CYCLIN INHIBITORS

Information

  • Patent Application
  • 20240408169
  • Publication Number
    20240408169
  • Date Filed
    May 17, 2024
    7 months ago
  • Date Published
    December 12, 2024
    15 days ago
Abstract
Disclosed herein are compounds of Formula (I), intermediates thereof, and methods for making the same:
Description
BACKGROUND

Cyclins are a family of proteins that play a central role in the regulation of the cell cycle. Specific cyclins, including Cyclins D, E, A and B, are expressed at the different stages of the cell cycle, during which they bind and activate their cognate cyclin dependent kinases (CDKs), including CDKs 1, 2, 4 and 6, to form cyclin-CDK complexes that orchestrate progression and transitions through the different stages of the cell cycle. Disruptions of the normal regulatory functions of cyclin-CDK complexes are common drivers of oncogenesis and the rapid proliferation of cancer cells. The central role of cyclins and CDKs in the cell cycle makes these proteins and their complexes attractive targets for treating proliferative disorders and cancer. To date, most inhibitors of cyclin-CDK complexes target the kinase activity of CDKs (“CDK inhibitors”) and include therapeutics both in development and approved for clinical use. Alternative approaches could include disrupting the association of cyclins with CDKs or the interaction of a particular cyclin-CDK complex with its substrates or regulators.


Although CDK inhibitors have been developed and proven successful in certain cancers, they are currently limited by their relative lack of selectivity, small therapeutic window, and ultimately the development of resistance. As such, there is a need to develop agents that offer alternative approaches to inhibiting the function of cyclin-CDK complexes as a means to modulate the cell cycle. Such agents could provide new tools in the treatment of proliferative diseases. The present disclosure addresses this need by providing compounds that inhibit the binding of substrates to various cyclins, thereby disrupting the function of cyclin-CDK complexes.


BRIEF SUMMARY

In one embodiment, provided herein is a compound of Formula (I):




embedded image


wherein

    • R3 is
      • (a) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C1-6 haloalkyl, each substituted with 0 to 5 R3a,
      • (b) C3-12 cycloalkyl substituted with 0 to 5 R3b, or
      • (c) heterocycloalkyl having 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 5 R3c;
      • (g) heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heteroaryl is substituted with 0, 1, 2, 3, 4, or 5 R3g;
    • each R3a is —OH, C1-6 alkoxy, C1-6 haloalkoxy, or —O—C(O)C1-6 alkyl;
    • each R3b is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, —OH, C1-6 alkoxy, halo, C1-6 haloalkyl, C1-6 haloalkoxy, or cyano;
    • each R3c is independently C1-6 alkyl, halo, C1-6 haloalkyl, cyano, C3-6 cycloalkyl, or oxo;
    • each R3g is independently C1-6 alkyl, halo, C1-6 haloalkyl, or C3-6 cycloalkyl;
    • R4a is H or C1-6 alkyl;
    • R4b and R4c are each independently H, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;
    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 4 R4a1;
    • each R4a1 is independently C1-6 alkyl, —OH, C1-6 alkyl-OH, C1-6 alkoxy, or halo;
    • R5a is H or C1-6 alkyl;
    • R5b and R5c are each independently H, C1-6 alkyl, —C1-6 alkyl-OH, C2-6 alkoxyalkyl, C1-6 haloalkyl, —C1-6 alkyl-C(O)NR5b1R5b2, —C1-6 alkyl-C(O)OR5b1, C3-6 cycloalkyl, C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or
    • —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R5b5;
    • each R5b1 and R5b2 are independently H, C1-4 alkyl, or C1-4 haloalkyl;
    • alternatively, R5b1 and R5b2 together with the nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members and 0 to 1 additional heteroatoms, wherein the heterocycloalkyl is substituted with 0 to 2 R5b3;
    • each R5b3 and R5b5 is independently C1-6 alkyl, halo, C1-6 haloalkyl, or cyano;
    • X6 is C6-7 alkylene or C6-7 alkenylene;
    • R6a is H, C1-6 alkyl, C1-6 deuteroalkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein the heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and the heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;
    • R6b and R6d are each independently H or C1-6 alkyl;
    • R7a is H or C1-6 alkyl;
    • R7b and R7c are each independently H, C1-6 alkyl, C2-6 alkenyl, C1-6 haloalkyl, C1-6 alkyl-OH, C3-6 cycloalkyl, or —C1-6 alkyl-C3-6 cycloalkyl;
    • R8a is H, C1-6 alkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl or —C1-6 alkyl-C3-6 cycloalkyl;
    • R8b, R8d, and R8e are each independently H or C1-6 alkyl;
    • ring B is C6-12 aryl or heteroaryl having 5 to 12 ring members and 1 to 6 heteroatoms, each independently N, O or S;
    • the subscript m8 is an integer from 0 to 5;
    • each R8f is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 deuteroalkoxy, halo, C1-6 haloalkyl, cyano, —X8f-cyano, C3-12 cycloalkyl,
    • —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl,
    • —X8f-heterocycloalkyl, C6-12 aryl, —X8f—C6-12 aryl, heteroaryl, or
    • —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein each alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl is substituted with 0 to 3 R8f3;
    • alternatively, two R8f groups on adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the cycloalkyl or heterocycloalkyl is substituted with 0 to 3 R8f3;
    • each X8f is independently C1-6 alkylene, C2-6 alkenylene, —O—C1-6 alkylene, C(O), O, or S;
    • each R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3;
    • each R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalkyl,
    • —Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,
    • —C1-6 alkyl-Y8—C1-6 alkyl, —(C1-2 alkyl-O)1-4—C1-2 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, cyano, —C1-6 alkyl-cyano, —C1-6 alkyl-NR8gR8h, oxo, C3-6 cycloalkyl, —X8f3—C3-6 cycloalkyl, heterocycloalkyl, —X8f3-heterocycloalkyl, phenyl, —X8f3-phenyl, heteroaryl, or —X8f3-heteroaryl, wherein each heterocycloalkyl has 3 to 12 members and 1 to 4 heteroatoms, each independently N, O, S, or S(O)2, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and wherein each heterocycloalkyl and heteroaryl are substituted with 0 to 2 C1-4 alkyl or halo;
    • each X8f3 is independently C1-6 alkylene, O, C(O), or S(O)2;
    • each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2;
    • each R8g and R8h is independently H, C1-3 alkyl, or C1-6 haloalkyl;
    • alternatively, two R8f3 groups on the same or adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein nitrogen atom ring members in the heterocycloalkyl are substituted with 0 to 1 C1-4 alkyl; and
    • each R8f4 is independently H or C1-6 alkyl;


      or a pharmaceutically acceptable salt thereof.


In another embodiment, the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure, and a pharmaceutically acceptable excipient.


In another embodiment, the present disclosure provides a method of treating a disease or disorder mediated at least in part by cyclin activity, the method comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure, thereby treating the disorder or condition.


In another embodiment, the present disclosure provides a method of treating a cancer mediated at least in part by cyclin A, the method comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure, thereby treating the cancer.


In another embodiment, the present invention provides intermediates useful in the preparation of compounds of Formula (I).


Other objects, features, and advantages of the present disclosure will be apparent to one of skill in the art from the following detailed description and FIGURES.





BRIEF DESCRIPTION OF THE DRAWINGS

NOT APPLICABLE





DETAILED DESCRIPTION
I. General

Provided herein are compounds and compositions that disrupt the typical cellular function of cyclins. Also provided herein are, for example, methods of treating or preventing a disease, disorder or condition, or a symptom thereof, mediated by cyclin activity.


Complexes between cyclins and cyclin dependent kinases (CDKs) are responsible for phosphorylating a wide range of substrates, thereby modulating the activity of the substrates. Many of these substrates are important in the cell cycle and the cyclin and CDKs that regulate these substrates therefore play key roles in regulating the cell cycle, including Cyclins D, A, E and B, and CDKs 1, 2, 4 and 6. Without being bound to any particular theory, certain substrates, including p21, p27, Rb, E2F and CDC6, first bind to the cyclin-CDK complex via a conserved R×L motif within the substrate (Adams et al. Mol Cell Biol. 1996. 16(12):6223-33.) and bind to a region with the cyclin that is referred to as an R×L binding domain or a “hydrophobic patch” (Brown et al. Nat Cell Biol. 1999.1(7):438-43) and contains a highly conserved MRAIL motif. Compounds that disrupt the binding of substrates to cyclins have been posited to be of potential therapeutic utility, including in the disruption of cancer cell proliferation (Chen et al. Proc Natl Acad Sci USA. 1999. 96(8):4325-9).


Without being bound to any particular theory, it is believed that compounds of the present disclosure inhibit the binding of substrates to the hydrophobic patch region of cyclins including, but not limited to, Cyclins A, E and B. Compounds of the present disclosure include compounds that bind more potently to one or more cyclins.


II. Definitions

As used herein, the term “about” means a range of values including the specified value, which a person of ordinary skill in the art would consider reasonably similar to the specified value. In some embodiments, the term “about” means within a standard deviation using measurements generally acceptable in the art. In some embodiments, about means a range extending to +/−10% of the specified value. In some embodiments, about means the specified value.


“Alkyl” refers to a straight or branched, saturated, aliphatic radical having the number of carbon atoms indicated. Alkyl can include any number of carbons, such as C1-2, C1-3, C1-4, C1-5, C1-6, C1-7, C1-8, C1-9, C1-10, C2-3, C2-4, C2-5, C2-6, C3-4, C3-5, C3-6, C4-5, C4-6 and C5-6. For example, C1-6 alkyl includes, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, hexyl, etc. Alkyl can also refer to alkyl groups having up to 20 carbons atoms, such as, but not limited to heptyl, octyl, nonyl, decyl, etc. Alkyl groups can be substituted or unsubstituted.


“Alkylene” refers to a straight or branched, saturated, aliphatic radical having the number of carbon atoms indicated, and linking at least two other groups, i.e., a divalent hydrocarbon radical. The two moieties linked to the alkylene can be linked to the same atom or different atoms of the alkylene group. For instance, a straight chain alkylene can be the bivalent radical of —(CH2)n—, where n is 1, 2, 3, 4, 5 or 6. Representative alkylene groups include, but are not limited to, methylene, ethylene, propylene, isopropylene, butylene, isobutylene, sec-butylene, pentylene and hexylene. Alkylene groups can be substituted or unsubstituted.


“Alkenyl” refers to a straight chain or branched hydrocarbon having at least 2 carbon atoms and at least one double bond. Alkenyl can include any number of carbons, such as C2, C2-3, C2-4, C2-5, C2-6, C2-7, C2-8, C2-9, C2-10, C3, C3-4, C3-5, C3-6, C4, C4-5, C4-6, C5, C5-6, and C6. Alkenyl groups can have any suitable number of double bonds, including, but not limited to, 1, 2, 3, 4, 5 or more. Examples of alkenyl groups include, but are not limited to, vinyl(ethenyl), propenyl, isopropenyl, 1-butenyl, 2-butenyl, isobutenyl, butadienyl, 1-pentenyl, 2-pentenyl, isopentenyl, 1,3-pentadienyl, 1,4-pentadienyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 1,3-hexadienyl, 1,4-hexadienyl, 1,5-hexadienyl, 2,4-hexadienyl, or 1,3,5-hexatrienyl. Alkenyl groups can be substituted or unsubstituted.


“Alkenylene” refers to a straight or branched hydrocarbon having at least 2 carbon atoms, one double bond, and linking at least two other groups, i.e., a divalent hydrocarbon radical. Alkenylene can include any number of carbons, such as C2, C2-3, C2-4, C2-5, C2-6, C2-7, C2-8, C2-9, C2-10, C3, C3-4, C3-5, C3-6, C3-6, C3-7, C4, C4-5, C4-6, C4-7, C6-7, C5, C6 and C7. The two moieties linked to the alkenylene can be linked to the same atom or different atoms of the alkenylene group. Representative alkenylene groups include, but are not limited to, (E)-hex-2-enylene, (Z)-hex-2-enylene, (E)-hept-2-enylene, (Z)-hept-2-enylene, (E)-hept-3-enylene, and (Z)-hept-3-enylene. Alkenylene moieties can be in the E or Z isomer. Alkenylene groups can be substituted or unsubstituted.


“Alkynyl” refers to either a straight chain or branched hydrocarbon having at least 2 carbon atoms and at least one triple bond. Alkynyl can include any number of carbons, such as C2, C2-3, C2-4, C2-5, C2-6, C2-7, C2-8, C2-9, C2-10, C3, C3-4, C3-5, C3-6, C4, C4-5, C4-6, C5, C5-6, and C6. Examples of alkynyl groups include, but are not limited to, acetylenyl, propynyl, 1-butynyl, 2-butynyl, butadiynyl, 1-pentynyl, 2-pentynyl, isopentynyl, 1,3-pentadiynyl, 1,4-pentadiynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 1,3-hexadiynyl, 1,4-hexadiynyl, 1,5-hexadiynyl, 2,4-hexadiynyl, or 1,3,5-hexatriynyl. Alkynyl groups can be substituted or unsubstituted.


“Alkoxy” refers to an alkyl group having an oxygen atom that connects the alkyl group to the point of attachment: alkyl-O—. As for alkyl group, alkoxy groups can have any suitable number of carbon atoms, such as C1-6. Alkoxy groups include, for example, methoxy, ethoxy, propoxy, iso-propoxy, butoxy, 2-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, pentoxy, hexoxy, etc. The alkoxy groups can be substituted or unsubstituted.


“Alkoxyalkyl” refers to alkyl group connected to an oxygen atom that is further connected to an second alkyl group, the second alkyl group being the point of attachment to the remainder of the molecule: alkyl-O-alkyl. The alkyl portion can have any suitable number of carbon atoms, such as C2-6. Alkoxyalkyl groups include, for example, methoxymethyl, ethoxymethyl, methoxyethyl, ethoxyethyl, etc. The alkoxy groups can be substituted or unsubstituted.


“Halo” or “halogen” refers to fluorine, chlorine, bromine and iodine.


“Haloalkyl” refers to alkyl, as defined above, where some or all of the hydrogen atoms are replaced with halogen atoms. As for alkyl group, haloalkyl groups can have any suitable number of carbon atoms, such as C1-6. For example, haloalkyl includes trifluoromethyl, flouromethyl, etc. In some instances, the term “perfluoro” can be used to define a compound or radical where all the hydrogens are replaced with fluorine. For example, perfluoromethyl refers to 1,1,1-trifluoromethyl.


“Haloalkoxy” refers to an alkoxy group where some or all of the hydrogen atoms are substituted with halogen atoms. As for an alkyl group, haloalkoxy groups can have any suitable number of carbon atoms, such as C1-6. The alkoxy groups can be substituted with 1, 2, 3, or more halogens. When all the hydrogens are replaced with a halogen, for example by fluorine, the compounds are per-substituted, for example, perfluorinated. Haloalkoxy includes, but is not limited to, trifluoromethoxy, 2,2,2,-trifluoroethoxy, perfluoroethoxy, etc.


“Cycloalkyl” refers to a saturated or partially unsaturated, monocyclic, spirocyclic, fused or bridged polycyclic ring assembly containing from 3 to 12 ring atoms, or the number of atoms indicated. Cycloalkyl can include any number of carbons, such as C3-6, C4-6, C5-6, C3-8, C4-8, C5-8, C6-8, C3-9, C3-10, C3-11, and C3-12. Saturated monocyclic cycloalkyl rings include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclooctyl. Saturated bicyclic and polycyclic cycloalkyl rings include, for example, norbornane, [2.2.2]bicyclooctane, decahydronaphthalene and adamantane. Cycloalkyl groups can also be partially unsaturated, having one or more double or triple bonds in the ring, but cycloalkyl groups are not aromatic. Representative cycloalkyl groups that are partially unsaturated include, but are not limited to, cyclobuteneyl, cyclopenteneyl, cyclohexeneyl, cyclohexadieneyl (1,3- and 1,4-isomers), cyclohepteneyl, cycloheptadieneyl, cycloocteneyl, cyclooctadieneyl (1,3-, 1,4- and 1,5-isomers), norborneneyl, and norbornadieneyl. When cycloalkyl is a C3-6 monocyclic cycloalkyl, exemplary groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexeneyl, cyclohexadieneyl (1,3- and 1,4-isomers). When cycloalkyl is a C5-10 fused bicyclic cycloalkyl, exemplary groups include, but are not limited to bicyclo[3.1.0]hexanyl, bicyclo[4.1.0]heptanyl, bicyclo[4.2.0]octanyl, and octahydro-1H-indenyl. When cycloalkyl is a C5-10 bridged polycyclic cycloalkyl, exemplary groups include, but are not limited to bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, and bicyclo[2.1.1]hexane. When cycloalkyl is a C5-10 spirocycloalkyl, exemplary groups include, but are not limited to spiro[3.3]heptane, spiro[3.4]octane, spiro[3.5]nonanyl, spiro[2.5]octane, and spiro[2.4]heptane. Cycloalkyl groups can be substituted or unsubstituted.


“Heterocycloalkyl” refers to a saturated or partially unsaturated, monocyclic, spirocyclic, fused or bridged polycyclic ring assembly having from 3 to 12 ring members and from 1 to 4 heteroatoms of N, O and S. The heteroatoms can also be oxidized, such as, but not limited to, —S(O)— and —S(O)2—. Heterocycloalkyl groups can include any number of ring atoms, such as, 3 to 6, 4 to 6, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, or 3 to 12 ring members. Any suitable number of heteroatoms can be included in the heterocycloalkyl groups, such as 1, 2, 3, or 4, or 1 to 2, 1 to 3, 1 to 4, 2 to 3, 2 to 4, or 3 to 4. The heterocycloalkyl group can include groups such as aziridine, azetidine, pyrrolidine, piperidine, azepane, azocane, quinuclidine, pyrazolidine, imidazolidine, piperazine (1,2-, 1,3- and 1,4-isomers), oxirane, oxetane, tetrahydrofuran, oxane (tetrahydropyran), tetrahydropyridine, oxepane, thiirane, thietane, thiolane (tetrahydrothiophene), thiane (tetrahydrothiopyran), oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, morpholine, thiomorpholine, dioxane, or dithiane. Heterocycloalkyl groups can be unsubstituted or substituted.


The heterocycloalkyl groups can be linked via any position on the ring. For example, aziridine can be 1- or 2-aziridine, azetidine can be 1- or 2-azetidine, pyrrolidine can be 1-, 2- or 3-pyrrolidine, piperidine can be 1-, 2-, 3- or 4-piperidine, pyrazolidine can be 1-, 2-, 3-, or 4-pyrazolidine, imidazolidine can be 1-, 2-, 3- or 4-imidazolidine, piperazine can be 1-, 2-, 3- or 4-piperazine, tetrahydrofuran can be 1- or 2-tetrahydrofuran, oxazolidine can be 2-, 3-, 4- or 5-oxazolidine, isoxazolidine can be 2-, 3-, 4- or 5-isoxazolidine, thiazolidine can be 2-, 3-, 4- or 5-thiazolidine, isothiazolidine can be 2-, 3-, 4- or 5-isothiazolidine, and morpholine can be 2-, 3- or 4-morpholine.


When heterocycloalkyl is a monocyclic heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, representative members include, but are not limited to, pyrrolidine, piperidine, tetrahydrofuran, oxane, tetrahydrothiophene, thiane, pyrazolidine, imidazolidine, piperazine, oxazolidine, isoxzoalidine, thiazolidine, isothiazolidine, morpholine, thiomorpholine, dioxane and dithiane. Heterocycloalkyl can also be monocyclic heterocycloalkyl having 5 to 6 ring members and 1 to 2 heteroatoms, with representative members including, but not limited to, pyrrolidine, piperidine, tetrahydrofuran, tetrahydrothiophene, pyrazolidine, imidazolidine, piperazine, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, and morpholine.


“Aryl” refers to an aromatic ring system having any suitable number of ring atoms and any suitable number of rings. Aryl groups can include any suitable number of ring atoms, such as, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16 ring atoms, as well as from 6 to 10, 6 to 12, or 6 to 14 ring members. Aryl groups can be monocyclic, fused to form bicyclic or tricyclic groups, or linked by a bond to form a biaryl group. Representative aryl groups include phenyl, naphthyl and biphenyl. Other aryl groups include benzyl, having a methylene linking group. Some aryl groups have from 6 to 12 ring members, such as phenyl, naphthyl or biphenyl. Other aryl groups have from 6 to 10 ring members, such as phenyl or naphthyl. Some other aryl groups have 6 ring members, such as phenyl. Aryl groups can be substituted or unsubstituted.


“Heteroaryl” refers to a monocyclic or fused bicyclic or tricyclic aromatic ring assembly containing 5 to 12 ring atoms, where from 1 to 6 of the ring atoms are a heteroatom such as N, O or S. The heteroatoms can also be oxidized, such as, but not limited to, —S(O)— and —S(O)2—. Heteroaryl groups can include any number of ring atoms, such as, 5 to 6, 5 to 8, 5 to 9, 5 to 10, 5 to 12, or 9 to 12 ring members. Any suitable number of heteroatoms can be included in the heteroaryl groups, such as 1, 2, 3, 4, 5, or 6, or 1 to 2, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, 2 to 5, 2 to 6, 3 to 4, 3 to 5, or 3 to 6. Heteroaryl groups can have from 5 to 8 ring members and from 1 to 4 heteroatoms, or from 5 to 8 ring members and from 1 to 3 heteroatoms, or from 5 to 6 ring members and from 1 to 4 heteroatoms, or from 5 to 6 ring members and from 1 to 3 heteroatoms. The heteroaryl group can include groups such as pyrrole, pyridine, imidazole, pyrazole, triazole, tetrazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3,5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole. The heteroaryl groups can also be fused to aromatic ring systems, such as a phenyl ring, to form members including, but not limited to, benzopyrroles such as indole and isoindole, benzopyridines such as quinoline and isoquinoline, benzopyrazine (quinoxaline), benzopyrimidine (quinazoline), benzopyridazines such as phthalazine and cinnoline, benzothiophene, and benzofuran. Other heteroaryl groups include heteroaryl rings linked by a bond, such as bipyridine. Heteroaryl groups can be substituted or unsubstituted.


The heteroaryl groups can be linked via any position on the ring. For example, pyrrole includes 1-, 2- and 3-pyrrole, pyridine includes 2-, 3- and 4-pyridine, imidazole includes 1-, 2-, 4- and 5-imidazole, pyrazole includes 1-, 3-, 4- and 5-pyrazole, triazole includes 1-, 4- and 5-triazole, tetrazole includes 1- and 5-tetrazole, pyrimidine includes 2-, 4-, 5- and 6-pyrimidine, pyridazine includes 3- and 4-pyridazine, 1,2,3-triazine includes 4- and 5-triazine, 1,2,4-triazine includes 3-, 5- and 6-triazine, 1,3,5-triazine includes 2-triazine, thiophene includes 2- and 3-thiophene, furan includes 2- and 3-furan, thiazole includes 2-, 4- and 5-thiazole, isothiazole includes 3-, 4- and 5-isothiazole, oxazole includes 2-, 4- and 5-oxazole, isoxazole includes 3-, 4- and 5-isoxazole, indole includes 1-, 2- and 3-indole, isoindole includes 1- and 2-isoindole, quinoline includes 2-, 3- and 4-quinoline, isoquinoline includes 1-, 3- and 4-isoquinoline, quinazoline includes 2- and 4-quinoazoline, cinnoline includes 3- and 4-cinnoline, benzothiophene includes 2- and 3-benzothiophene, and benzofuran includes 2- and 3-benzofuran.


Some heteroaryl groups include those having from 5 to 10 ring members and from 1 to 3 ring atoms including N, O or S, such as pyrrole, pyridine, imidazole, pyrazole, triazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3,5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, isoxazole, indole, isoindole, quinoline, isoquinoline, quinoxaline, quinazoline, phthalazine, cinnoline, benzothiophene, and benzofuran. Other heteroaryl groups include those having from 5 to 8 ring members and from 1 to 3 heteroatoms, such as pyrrole, pyridine, imidazole, pyrazole, triazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3,5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole. Some other heteroaryl groups include those having from 9 to 12 ring members and from 1 to 3 heteroatoms, such as indole, isoindole, quinoline, isoquinoline, quinoxaline, quinazoline, phthalazine, cinnoline, benzothiophene, benzofuran and bipyridine. Still other heteroaryl groups include those having from 5 to 6 ring members and from 1 to 2 ring atoms including N, O or S, such as pyrrole, pyridine, imidazole, pyrazole, pyrazine, pyrimidine, pyridazine, thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole.


“Thiophenyl” refers to a thiophene radical.


As used herein, the term “oxo” refers to an oxygen atom connected to the point of attachment by a double bond (═O).


“Pharmaceutically acceptable excipient” refers to a substance that aids the formulation and/or administration of an active agent to a subject. Pharmaceutical excipients useful in the present disclosure include, but are not limited to, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors and colors. One of skill in the art will recognize that other pharmaceutical excipients are useful in the present disclosure.


“Subject” refers to animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice and the like. In some embodiments, the subject is a human.


“Administering” refers to oral administration, administration as a suppository, topical contact, parenteral, intravenous, intraperitoneal, intramuscular, intralesional, intranasal or subcutaneous administration, intrathecal administration, or the implantation of a slow-release device e.g., a mini-osmotic pump, to the subject.


“Therapeutically effective amount” refers to a dose that produces therapeutic effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins)


“Treat”, “treating” and “treatment” refers to any indicia of success in the treatment or amelioration of an injury, pathology, condition, or symptom (e.g., pain), including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the symptom, injury, pathology or condition more tolerable to the patient; decreasing the frequency or duration of the symptom or condition. The treatment or amelioration of symptoms can be based on any objective or subjective parameter; including, e.g., the result of a physical examination.


III. Compounds

In some embodiments, the present disclosure provides a compound of Formula (I)




embedded image




    • R3 is
      • (a) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C1-6 haloalkyl, each substituted with 0 to 5 R3a,
      • (b) C3-12 cycloalkyl substituted with 0 to 5 R3b, or
      • (c) heterocycloalkyl having 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 5 R3c;
      • (g) heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heteroaryl is substituted with 0, 1, 2, 3, 4, or 5 R3g;

    • each R3a is —OH, C1-6 alkoxy, C1-6 haloalkoxy, or —O—C(O)C1-6 alkyl;

    • each R3b is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, —OH, C1-6 alkoxy, halo, C1-6 haloalkyl, C1-6 haloalkoxy, or cyano;

    • each R3c is independently C1-6 alkyl, halo, C1-6 haloalkyl, cyano, C3-6 cycloalkyl, or oxo;

    • each R3g is independently C1-6 alkyl, halo, C1-6 haloalkyl, or C3-6 cycloalkyl;

    • R4a is H or C1-6 alkyl;

    • R4b and R4c are each independently H, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;

    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 4 R4a1;

    • each R4a1 is independently C1-6 alkyl, —OH, C1-6 alkyl-OH, C1-6 alkoxy, or halo;

    • R5a is H or C1-6 alkyl;

    • R5b and R5c are each independently H, C1-6 alkyl, —C1-6 alkyl-OH, C2-6 alkoxyalkyl, C1-6 haloalkyl, —C1-6 alkyl-C(O)NR5b1R5b2, —C1-6 alkyl-C(O)OR5b1, C3-6 cycloalkyl, C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or

    • —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R5b5;

    • each R5b1 and R5b2 are independently H, C1-4 alkyl, or C1-4 haloalkyl; alternatively, R5b1 and R5b2 together with the nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members and 0 to 1 additional heteroatoms, wherein the heterocycloalkyl is substituted with 0 to 2 R5b3;

    • each R5b3 and R5b5 is independently C1-6 alkyl, halo, C1-6 haloalkyl, or cyano;

    • X6 is C6-7 alkylene or C6-7 alkenylene;

    • R6a is H, C1-6 alkyl, C1-6 deuteroalkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein the heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and the heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;

    • R6b and R6d are each independently H or C1-6 alkyl;

    • R7a is H or C1-6 alkyl;

    • R7b and R7c are each independently H, C1-6 alkyl, C2-6 alkenyl, C1-6 haloalkyl, C1-6 alkyl-OH, C3-6 cycloalkyl, or —C1-6 alkyl-C3-6 cycloalkyl;

    • R8a is H, C1-6 alkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl or —C1-6 alkyl-C3-6 cycloalkyl;

    • R8b, R8d, and R8e are each independently H or C1-6 alkyl;

    • ring B is C6-12 aryl or heteroaryl having 5 to 12 ring members and 1 to 6 heteroatoms, each independently N, O or S;

    • the subscript m8 is an integer from 0 to 5;

    • each R8f is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 deuteroalkoxy, halo, C1-6 haloalkyl, cyano, —X8f-cyano, C3-12 cycloalkyl,

    • —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl,

    • —X8f-heterocycloalkyl, C6-12 aryl, —X8f—C6-12 aryl, heteroaryl, or

    • —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein each alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl is substituted with 0 to 3 R8f3;

    • alternatively, two R8f groups on adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the cycloalkyl or heterocycloalkyl is substituted with 0 to 3 R8f3;

    • each X8f is independently C1-6 alkylene, C2-6 alkenylene, —O—C1-6 alkylene, C(O), O, or S;

    • each R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3;

    • each R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalkyl,

    • —Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,

    • —C1-6 alkyl-Y8—C1-6 alkyl, —(C1-2 alkyl-O)1-4—C1-2 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, cyano, —C1-6 alkyl-cyano, —C1-6 alkyl-NR8gR8h, oxo, C3-6 cycloalkyl, —X8f3—C3-6 cycloalkyl, heterocycloalkyl, —X8f3-heterocycloalkyl, phenyl, —X8f3-phenyl, heteroaryl, or —X8f3-heteroaryl, wherein each heterocycloalkyl has 3 to 12 members and 1 to 4 heteroatoms, each independently N, O, S, or S(O)2, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and wherein each heterocycloalkyl and heteroaryl are substituted with 0 to 2 C1-4 alkyl or halo;

    • each X8f3 is independently C1-6 alkylene, O, C(O), or S(O)2;

    • each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2;

    • each R8g and R8h is independently H, C1-3 alkyl, or C1-6 haloalkyl;

    • alternatively, two R8f3 groups on the same or adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein nitrogen atom ring members in the heterocycloalkyl are substituted with 0 to 1 C1-4 alkyl; and

    • each R8f4 is independently H or C1-6 alkyl;

    • or a pharmaceutically acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of Formula (I)




embedded image


wherein

    • R3 is
      • (a) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C1-6 haloalkyl, each substituted with 0 to 5 R3a,
      • (b) C3-12 cycloalkyl substituted with 0 to 5 R3b, or
      • (c) heterocycloalkyl having 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 5 R3c;
    • each R3a is —OH, C1-6 alkoxy, C1-6 haloalkoxy, or —O—C(O)C1-6 alkyl;
    • each R3b is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, —OH, C1-6 alkoxy, halo, C1-6 haloalkyl, C1-6 haloalkoxy, or cyano;
    • each R3c is independently C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or oxo;
    • R4a is H or C1-6 alkyl;
    • R4b and R4c are each independently H, C1-6 alkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;
    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members, wherein the heterocycloalkyl is substituted with 0 to 4 R4a1;
    • each R4a1 is independently C1-6 alkyl, —OH, C1-6 alkyl-OH, C1-6 alkoxy, or halo;
    • R5a is H or C1-6 alkyl;
    • R5b and R5c are each independently H, C1-6 alkyl, —C1-6 alkyl-OH, C2-6 alkoxyalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or
    • —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R5b5;
    • each R5b5 is independently C1-6 alkyl, halo, or C1-6 haloalkyl;
    • X6 is C6-7 alkylene or C6-7 alkenylene;
    • R6a is H, C1-6 alkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein the heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and the heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;
    • R6b and R6d are each independently H or C1-6 alkyl;
    • R7a is H or C1-6 alkyl;
    • R7b and R7c are each independently H, C1-6 alkyl, C2-6 alkenyl, C1-6 haloalkyl, C1-6 alkyl-OH, C3-6 cycloalkyl, or —C1-6 alkyl-C3-6 cycloalkyl;
    • R8a is H, C1-6 alkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl or —C1-6 alkyl-C3-6 cycloalkyl;
    • R8b, R8d, and R8e are each independently H or C1-6 alkyl;
    • ring B is C6-12 aryl or heteroaryl having 5 to 12 ring members and 1 to 6 heteroatoms, each independently N, O or S;
    • the subscript m8 is an integer from 0 to 5;
    • each R8f is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 deuteroalkoxy, halo, C1-6 haloalkyl, cyano, —X8f-cyano, C3-6 cycloalkyl,
    • —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl,
    • —X8f-heterocycloalkyl, C6-12 aryl, —X8f—C6-12 aryl, heteroaryl, or
    • —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein each alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl is substituted with 0 to 3 R8f3;
    • alternatively, two R8f groups on adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the cycloalkyl or heterocycloalkyl is substituted with 0 to 3 R8f3;
    • each X8f is independently C1-6 alkylene, C2-6 alkenylene, —O—C1-6 alkylene, O, or S;
    • each R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3;
    • each R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalkyl,
    • —Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,
    • —C1-6 alkyl-Y8—C1-6 alkyl, —(C1-2 alkyl-O)1-4—C1-2 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, cyano, —C1-6 alkyl-cyano, oxo, C3-6 cycloalkyl, —X8f3—C3-6 cycloalkyl, heterocycloalkyl, —X8f3-heterocycloalkyl, phenyl, —X8f3-phenyl, heteroaryl, or —X8f3-heteroaryl, wherein each heterocycloalkyl has 3 to 12 members and 1 to 4 heteroatoms, each independently N, O, S, or S(O)2, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and wherein nitrogen atom ring members in each heterocycloalkyl and heteroaryl are substituted with 0 to 1 C1-4 alkyl;
    • each X8f3 is independently C1-6 alkylene, O, C(O), or S(O)2;
    • each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2;
    • alternatively, two R8f3 groups on the same or adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein nitrogen atom ring members in the heterocycloalkyl are substituted with 0 to 1 C1-4 alkyl; and
    • each R8f4 is independently H or C1-6 alkyl;


      or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a compound of Formula (I)




embedded image


wherein

    • R3 is
      • (a) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C1-6 haloalkyl, each substituted with 0 to 5 R3a,
      • (b) C3-12 cycloalkyl substituted with 0 to 5 R3b, or
      • (c) heterocycloalkyl having 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 5 R3c;
    • each R3a is —OH, C1-6 alkoxy, C1-6 haloalkoxy, or —O—C(O)C1-6 alkyl;
    • each R3b is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, —OH, C1-6 alkoxy, halo, C1-6 haloalkyl, C1-6 haloalkoxy, or cyano;
    • each R3c is independently C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or oxo;
    • R4a is H or C1-6 alkyl;
    • R4b and R4c are each independently H, C1-6 alkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;
    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members, wherein the heterocycloalkyl is substituted with 0 to 4 R4a1;
    • each R4a1 is independently C1-6 alkyl, —OH, C1-6 alkyl-OH, C1-6 alkoxy, or halo;
    • R5a is H or C1-6 alkyl;
    • R5b and R5c are each independently H, C1-6 alkyl, —C1-6 alkyl-OH, C2-6 alkoxyalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or
    • —C1-6 alkyl-heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R5b5;
    • each R5b5 is independently C1-6 alkyl, halo, or C1-6 haloalkyl;
    • X6 is C6-7 alkylene or C6-7 alkenylene;
    • R6a is H, C1-6 alkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl, —C1-6 alkyl-C3-6 cycloalkyl, heterocycloalkyl, —C1-6 alkyl-heterocycloalkyl, phenyl, —C1-6 alkyl-phenyl, heteroaryl, or —C1-6 alkyl-heteroaryl, wherein the heterocycloalkyl has 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and the heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S;
    • R6b and R6d are each independently H or C1-6 alkyl;
    • R7a is H or C1-6 alkyl;
    • R7b and R7c are each independently H, C1-6 alkyl, C2-6 alkenyl, C1-6 haloalkyl, C1-6 alkyl-OH, C3-6 cycloalkyl, or —C1-6 alkyl-C3-6 cycloalkyl;
    • R8a is H, C1-6 alkyl, C2-6 alkoxyalkyl, C3-6 cycloalkyl or —C1-6 alkyl-C3-6 cycloalkyl;
    • R8b, R8d, and R8e are each independently H or C1-6 alkyl;
    • ring B is aryl or heteroaryl having 5 to 12 ring members and 1 to 6 heteroatoms, each independently N, O or S;
    • the subscript m8 is an integer from 0 to 5;
    • each R8f is independently C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 alkoxy, C1-6 deuteroalkoxy, halo, C1-6 haloalkyl, cyano, —X8f-cyano, C3-12 cycloalkyl,
    • —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl,
    • —X8f-heterocycloalkyl, aryl, —X8f-aryl, heteroaryl, or —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 12 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, wherein each alkynyl, cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3;
    • alternatively, two R8f groups on adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S;
    • each X8f is independently C1-6 alkylene, C2-6 alkenylene, O, or S;
    • each R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3;
    • each R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalkyl,
    • —Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,
    • —C1-6 alkyl-Y8—C1-6 alkyl, C1-6 alkoxy, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, cyano, —C1-6 alkyl-cyano, oxo, C3-6 cycloalkyl, —X8f3—C3-6 cycloalkyl, heterocycloalkyl, —X8f3-heterocycloalkyl, phenyl, —X8f3-phenyl, heteroaryl, or —X8f3-heteroaryl, wherein each heterocycloalkyl has 3 to 12 members and 1 to 4 heteroatoms, each independently N, O, S, or S(O)2, and each heteroaryl has 5 to 10 ring members and 1 to 4 heteroatoms, each independently N, O, or S, and wherein nitrogen atom ring members in each heterocycloalkyl and heteroaryl are substituted with 0 to 1 C1-4 alkyl;
    • each X8f3 is independently C1-6 alkylene, O, C(O), or S(O)2;
    • each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2;
    • alternatively, two R8f3 groups on adjacent ring vertices combine to form a C3-6 cycloalkyl or a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S; and
    • each R8f4 is independently H or C1-6 alkyl;
    • or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure of Formula (Ib)




embedded image




    • R3, R4a, R4c, R5a, R5c, R6a, X6, R6d, R8a, m8, R8f, and ring B can each independently be as defined for any embodiment of Formula (Ib) as described herein.





In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure of Formula (Ib1)




embedded image


R3, R4a, R4c, R5a, R5c, R6a, X6, R6d, R8a, m8, R8f, and ring B can each independently be as defined for any embodiment of Formula (Ib1) as described herein.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure of Formula (Ic):




embedded image




    • R3, R4a1, R5a, R5c, R6a, X6, R6d, R8a, m8, R8f, and ring B can each independently be as defined for any embodiment of Formula (Ic) as described herein.





In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure of Formula (Ic1)




embedded image




    • R3, R4a1, R5a, R5c, R6a, X6, R6d, R8a, m8, R8f, and ring B can each independently be as defined for any embodiment of Formula (Ic1) as described herein.





In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure of Formula (Id)




embedded image


wherein the dashed bond is absent or a single bond.

    • R3, R4a1, R5a, R5c, R6a, R6d, R8a, m8, R8f, and ring B can each independently be as defined for any embodiment of Formula (Id) as described herein. When the dashed line is a single bond in Formula (Id), the wavy bond attached to the double bond indicates E, Z, or a mixture of both isomers.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure of Formula (Id1)




embedded image


wherein the dashed bond is absent or a single bond.

    • R3, R4a1, R5a, R5c, R6a, R6d, R8a, m8, R8f, and ring B can each independently be as defined for any embodiment of Formula (Id1) as described herein. When the dashed line is a bond in Formula (Id1), the wavy line attached to the double bond indicates E, Z, or a mixture or both linkages.


Residue 3

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (a) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C1-6 haloalkyl substituted with 0 to 5 R3a. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (a) C1-6 alkyl or C1-6 haloalkyl substituted with 0 to 2 R3a. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (a) C1-6 alkyl substituted with 0 to 2 R3a. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (a) C1-6 haloalkyl substituted with 0 to 2 R3a. These embodiments of R3 can be combined with any of the embodiments described herein for R3a.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R3a is —OH or C1-6 alkoxy. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R3a is —OH. These embodiments of R3a can be combined with any of the relevant embodiments described herein for R3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (b) C3-12 cycloalkyl substituted with 0 to 3 R3b. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (b) C3-7 cycloalkyl substituted with 0 to 3 R3b. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (b) C5-6 cycloalkyl substituted with 0 to 2 R3b. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (b) C3-7 cycloalkyl substituted with 0 to 2 R3b. These embodiments of R3 can be combined with any of the embodiments described herein for R3b.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R3b is halo, C1-4 haloalkyl, or cyano. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R3b is halo or C1-4 haloalkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R3b is fluoro or trifluoromethyl. These embodiments of R3b can be combined with any of the relevant embodiments described herein for R3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (c) heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 5 R3c. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (c) heterocycloalkyl having 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 2 R3c. These embodiments of R3 can be combined with any of the relevant embodiments described herein for R3c.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3c is halo or C1-4 haloalkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R3c is fluoro or trifluoromethyl. These embodiments of R3c can be combined with any of the embodiments described herein for R3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is (g) heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heteroaryl is substituted with 0, 1, 2, 3, 4, or 5 R3g.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3g is independently C1-6 alkyl, halo, C1-6 haloalkyl, or C3-6 cycloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R3 is




embedded image


Any of the embodiments described herein for residue 3 can be combined with any of the embodiments described herein for residues 4, 5, 6, 7, and 8. For example, any of the embodiments of R3 as described herein, can be combined with any of the embodiments described herein for R4a, R4b, R4c, R5a, R5b, R5e, X6, R6a, R6b, R6d, R7a, R7b, R7e, R8a, R8b, R8d, R8e, ring B, m8, and R8f.


Residue 4

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (Ib1), wherein


R4a, R4b, and R4c are each independently H or C1-6 alkyl;

  • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 4 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 2 R4a1; and


    each R4a1 is independently halo.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (Ib1), wherein


R4a, R4b, and R4c are each independently H or C1-6 alkyl;

  • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 4 to 6 ring members, wherein the heterocycloalkyl is substituted with 0 to 2 R4a1; and


    each R4a1 is independently halo.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (Ib1), wherein


R4a, R4b, and R4c are each independently H or C1-6 alkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (Ib1), wherein


R4b is H or C1-6 alkyl; and

  • R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 4 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 3 R4a1.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (Ib1), wherein


R4b is H or C1-6 alkyl; and

  • R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 4 to 6 ring members, wherein the heterocycloalkyl is substituted with 0 to 3 R4a1.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (Ib1), wherein


R4a and R4b are each H; and


R4c is ethyl;

  • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a pyrrolidinyl, substituted with 0 to 2 fluoro.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), or (ml), wherein


R4b is H; and



  • R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a pyrrolidinyl, substituted with 0 to 2 R4a1.



In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), (Id1), wherein R4a1 is C1-6 alkyl or halo. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), (Id1), wherein R4a1 is halo. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), (Id1), wherein R4a1 is fluoro. These embodiments of R4a1 can be combined with any of the relevant embodiments described herein for R4.


The embodiments described herein for R4a, R4b and R4c can be present in any combination. In addition, the embodiments described herein for residue 4 can be present in combination with any of the embodiments described herein for residues 3, 5, 6, 7, and 8. For example, any of the embodiments of R4a, R4b and R4c as described herein, can be combined with any of the embodiments described herein for R3, R5a, R5, R5e, X6, R6a, R6b, R6d, R7a, R7b, R7c, R8a, R8b, R8d, R8e, ring B, m8, and R8f.


Residue 5

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a is H or C1-6 alkyl;

  • R5b and R5c are each independently H, C1-6 alkyl, —C1-6 alkyl-OH, C2-6 alkoxyalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or C1-4 alkyl-C3-6 cycloalkyl, wherein each cycloalkyl is substituted with 0 to 3 R5b5; and


    each R5b5 is independently C1-4 alkyl, halo, or C1-4 haloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a is H or C1-6 alkyl;

  • R5b and R5c are each independently H, —C1-6 alkyl-C(O)NR5b1R5b2, or —C1-6 alkyl-C(O)OR5b1;


    each R5b1 and R5b2 are independently H, C1-4 alkyl, or C1-4 haloalkyl;
  • alternatively, R5b1 and R5b2 together with the nitrogen to which each is attached combine to form a heterocycloalkyl having 3 to 6 ring members and 0 to 1 additional heteroatoms, wherein the heterocycloalkyl is substituted with 0 to 2 R5b3; and
  • each R5b3 is independently C1-6 alkyl, halo, C1-6 haloalkyl, or cyano each R5b5 is independently C1-4 alkyl, halo, or C1-4 haloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a is H or C1-6 alkyl; and


R5b and R5c are each independently H, C1-6 alkyl, C3-6 cycloalkyl, C1-4 alkyl-C3-6 cycloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a is H or C1-6 alkyl;


R5b is H; and

R5c is C3-6 cycloalkyl or C1-4 alkyl-C3-6 cycloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R5a and R5b are each H; and


R5c is H, methyl, ethyl,




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a and R5b are each H; and


R5c is H, methyl, ethyl,




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a is H or C1-6 alkyl;


R5b is H; and
R5c is



embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R5a is H or C1-6 alkyl;


R5b is H; and
R5c is



embedded image


The embodiments described herein for R5a, R5b and R5c can be present in any combination. In addition, the embodiments described herein for residue 5 can be present in combination with any of the embodiments described herein for residues 3, 4, 6, 7, and 8. For example, any of the embodiments of R5a, R5b and R5c as described herein, can be combined with any of the embodiments described herein for R3, R4a, R4b, R4c, X6, R6a, R6b, R6d, R7a, R7b, R7c, R8a, R8b, R8d, R8c, ring B, m8, and R8f.


Residue 6

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), or (Ic1), wherein X6 is C6-7 alkylene. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), or (Ic1), wherein X6 is C6-7 alkenylene. These embodiments of X6 can be combined with any of the embodiments described herein for R6a, R6b and R6d.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), or (Ic1), wherein X6 is




embedded image


wherein the wavy bond attached to the double bond indicates E, Z, or a mixture of both isomers. These embodiments of X6 can be combined with any of the embodiments described herein for R6a, R6b and R6d.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), or (Ic1), wherein X6 is




embedded image


wherein the wavy bond attached to the double bond indicates E, Z, or a mixture of both isomers. This embodiment of X6 can be combined with any of the embodiments described herein for R6a, R6b and R6d.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), or (Ic1), wherein X6 is




embedded image


This embodiment of X6 can be combined with any of the embodiments described herein for R6a, R6b and R6d.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R6a is H, C1-4 alkyl, C1-4 deuteroalkyl, or —C1-4 alkyl-C3-6 cycloalkyl; and R6b and R6d are each independently H or C1-6 alkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1),


wherein


R6a is H, C1-4 alkyl, or —C1-4 alkyl-C3-6 cycloalkyl; and R6b and R6d are each independently H or C1-6 alkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein


R6a is C1-4 alkyl; R6b is H; and R6d is C1-4 alkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R6a is C1-4 alkyl; and R6b and R6d are each H. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R6a, R6b, and R6d are each H. These embodiments of R6a, R6b and R6d can be combined with any of the embodiments described herein for X6.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R6a is H, methyl, ethyl,




embedded image


R6b is H; and R6d is H, methyl, or ethyl. This embodiment of R6a, R6b and R6d can be combined with any of the embodiments described herein for X6.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R6a is H, methyl, ethyl, or




embedded image


R6b is H; and R6d is H, methyl, or ethyl. This embodiment of R6a, R6b and R6d can be combined with any of the embodiments described herein for X6.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R6a is H, methyl, ethyl, or




embedded image


R6b is H; and R6d is H or methyl. This embodiment of R6a, R6b and R6d can be combined with any of the embodiments described herein for X6.


The embodiments described herein for X6, R6a, R6b and R6d can be present in any combination. In addition, the embodiments described herein for residue 6 can be present in combination with any of the embodiments described herein for residues 3, 4, 5, 7, and 8. For example, any of the embodiments of X6, R6a, R6b and R6d as described herein, can be combined with any of the embodiments described herein for R3, R4a, R4b, R4, R5a, R5b, R5, R7a, R7b, R7e, R8a, R8b, R8d, R8e, ring B, m8, and R8f.


Residue 7

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), wherein R7a and R7b are each independently H or C1-6 alkyl; and R7c is C1-6 alkyl, C2-6 alkenyl, C1-6 haloalkyl, C1-6 alkyl-OH, or


—C1-6 alkyl-C3-6 cycloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), wherein R7a and R7b are each independently H or C1-6 alkyl; and R7c is C1-6 alkyl or C1-6 haloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), wherein R7a and R7b are each H; and R7c is C1-6 alkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), wherein R7a and R7b are each H; and R7c is isobutyl,




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), wherein R7a and R7b are each H; and R7c is isobutyl.


The embodiments described herein for R7a, R7b and R7c can be present in any combination. In addition, the embodiments described herein for residue 7 can be present in combination with any of the embodiments described herein for residues 3, 4, 5, 6, and 8. For example, any of the embodiments of R7a, R7b and R7c as described herein, can be combined with any of the embodiments described herein for R3, R4a, R4b, R4e, R5a, R5b, R50, X6, R6a, R6b, R6d, R8a, R8b, R8d, R8e, ring B, m8, and R8f.


Residue 8

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R8a, R8b, R8d, and R8e are each independently H or C1-6 alkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein R8a is H or methyl; and R8b, R8d and R8e are each H. These embodiments of R8a, R8b, R8d, and R8e can be combined with any of the embodiments described herein for m8, ring B, R8f, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is phenyl or heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, wherein each heteroatom is N, O, or S. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is phenyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is naphthyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is biphenyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is a heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, wherein each heteroatom is N, O, or S. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is thiophenyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is a heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, wherein each heteroatom is N. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is pyridyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is benzofuranyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is indolyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is indazolyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is quinolinyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is pyrid-3-yl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is phenyl or pyridyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein ring B is phenyl or pyrid-3-yl. These embodiments of ring B can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, R8f, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 1, 2, or 3. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 0. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 1. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 2. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 3. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 4. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the subscript m8 is 5. These embodiments of m8 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, R8f, R80, and ring B.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the moiety




embedded image


These embodiments can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, R8f, R8f3, m8 and ring B.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the moiety




embedded image


These embodiments can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, R8f, R8f3, m8 and ring B.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein the moiety




embedded image


These embodiments can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8, R8f, R8f3, m8 and ring B.


In some embodiments, at least one R8f is halo. In some embodiments, at least one R8f is fluoro or chloro. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently C1-6 alkyl, C2-6 alkenyl, C1-6 alkoxy, C1-6 deuteroalkoxy, halo, C1-6 haloalkyl, cyano, or —X8f-cyano. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, C3-6 cycloalkyl, —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl, —X8f-heterocycloalkyl, phenyl, —X8f-phenyl, heteroaryl, or —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3; each X8f is independently C1-6 alkylene, C2-6 alkenylene, O, or S; and each R8f1 and R8f are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, C3-6 cycloalkyl, —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl, —X8f-heterocycloalkyl, heteroaryl, or —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, and heteroaryl is substituted with 0 to 3 R8f3; each X8f is independently C1-6 alkylene, C2-6 alkenylene, O, or S; and each R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently C1-6 alkylene. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently C2-6 alkenylene. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently —O—C1-6 alkylene. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently C(O). In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently O. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently S. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f is independently C1-6 alkylene or O. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl, phenyl, or heteroaryl, wherein each heterocycloalkyl has 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3; each R8f1 and R8f are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently C1-4 alkyl, C2-4 alkenyl, C1-4 alkoxy, C1-4 deuteroalkoxy, halo, C1-4 haloalkyl, cyano, or —C1-2 alkyl-cyano. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, C3-6 cycloalkyl, —O—C3-6 cycloalkyl, heterocycloalkyl, —C2-4 alkenyl-heterocycloalkyl, —O-heterocycloalkyl, —CH═CR8f1R8f2, phenyl, —O-phenyl, heteroaryl, or —O-heteroaryl, wherein each heterocycloalkyl has 3 to 9 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 9 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3; and R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, C3-6 cycloalkyl, —O—C3-6 cycloalkyl, heterocycloalkyl, —C2-4 alkenyl-heterocycloalkyl, —O-heterocycloalkyl, phenyl, —O-phenyl, heteroaryl, or —O— heteroaryl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 2 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, C3-6 cycloalkyl, —O—C3-6 cycloalkyl, heteroaryl, or —O-heteroaryl, wherein each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, and heteroaryl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, heterocycloalkyl, —O-heterocycloalkyl, phenyl, or —O-phenyl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 2 heteroatoms, each independently N, O, or S, wherein each heterocycloalkyl and phenyl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f is independently halo, heterocycloalkyl, or —O-heterocycloalkyl, wherein each heterocycloalkyl has 3 to 6 ring members and 1 to 2 heteroatoms, each independently N, O, or S, wherein each heterocycloalkyl is substituted with 0 to 3 R8f3. These embodiments of R8f can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each


R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalky,


—Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,


—C1-6 alkyl-Y8—C1-6 alkyl, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, or oxo; each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2; and each R8f4 is independently H or C1-6 alkyl. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each


R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalky,


—Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,


—C1-6 alkyl-Y8—C1-6 alkyl, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, —C1-6 alkyl-NR8gR8h, or oxo; each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2; and each R8f4 is independently H or C1-6 alkyl. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each each R8g and R8h is independently H, C1-3 alkyl, or C1-3 haloalkyl.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently —Y8—C1-6 alkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently —Y8-methyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently C(O). In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently C(O)O. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently NHC(O). In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently O. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently S. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently S(O)2. These embodiments of Y8 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, R8f, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f3 is independently C1-6 alkyl, C1-6 deuteroalky, —OH, —C1-6 alkyl-OH, halo, C1-6 haloalkyl, or oxo. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f3 is independently C1-6 alkyl or —Y8—C1-6 alkyl. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each Y8 is independently a C(O) or C(O)O. These embodiments of R8f3 and Y8 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f3 is independently C3-6 cycloalkyl, —X8f3—C3-6 cycloalkyl, heterocycloalkyl, or —X8f3-heterocycloalkyl, wherein each heterocycloalkyl has 3 to 6 members and 1 to 2 heteroatoms, each independently N, O, S, or S(O)2; and each X8f3 is independently C1-6 alkylene, C(O), or S(O)2. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f3 is independently C1-6 alkylene. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f3 is independently C(O). In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each X8f3 is independently S(O)2. These embodiments of X8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, R8f, and R8f3.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f3 is independently C3-6 cycloalkyl or heterocycloalkyl, wherein each heterocycloalkyl has 3 to 6 members and 1 to 2 heteroatoms, each independently N, O, S, or S(O)2. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein two R8f3 groups on adjacent ring vertices combine to form a heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O or S. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f3 is independently C1-4 alkyl, C1-4 alkoxy, C2-6 alkoxyalkyl, —S(O)2—C1-4 alkyl, —C1-4 alkyl-S(O)2—C1-4 alkyl, halo, C1-4 haloalkyl, oxo, —C(O)—C1-4 alkyl, or —C(O)O—C1-4 alkyl. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein each R8f3 is independently C1-4 alkyl, C1-4 alkoxy, C2-6 alkoxyalkyl, halo, C1-4 haloalkyl, oxo, —S(O)2—C1-4 alkyl, —C1-4 alkyl-S(O)2—C1-4 alkyl, —C(O)—C1-4 alkyl, —C(O)O—C1-4 alkyl, C3-6 cycloalkyl, —C(O)—C3-6 cycloalkyl, —S(O)2—C3-6 cycloalkyl, heterocycloalkyl, —C1-4 alkyl-heterocycloalkyl, or —S(O)2-heterocycloalkyl, wherein each heterocycloalkyl has 4 to 6 members and 1 to 2 heteroatoms, each independently N, O, S, or S(O)2; alternatively, two R8f3 groups on adjacent ring vertices combine to form a non-aromatic cyclic moiety having 3 to 6 ring members and 0 to 2 additional heteroatoms, each independently N, O or S. These embodiments of R8f3 can be combined with any of the embodiments described herein for R8a, R8b, R8d, R8e, m8, ring B, and R8f.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein

    • m8 is 1, 2, or 3; and
    • each R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro, cyano,




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




    • alternatively, ring B and three R8f groups form the moiety







embedded image


wherein the wavy line identifies attachment to the remainder of the molecule.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id11), wherein

    • m8 is 1, 2, or 3; and
    • each R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro, cyano,




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




    • alternatively, ring B and three R8f groups form the moiety







embedded image




    • wherein the wavy line identifies attachment to the remainder of the molecule.





In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein

    • m8 is 1, 2, or 3; and
    • each R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro,




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




    • alternatively, ring B and three R8f groups form the moiety







embedded image


wherein the wavy line identifies attachment to the remainder of the molecule.


The embodiments described herein for R8a, R8b, R8d, R8e, m8, R8f, and ring B can be present in any combination. In addition, the embodiments described herein for residue 8 can be present in combination with any of the embodiments described herein for residues 3, 4, 5, 6, and 7. For example, any of the embodiments of R8a, R8b, R8d, R8e, m8, R8f, and ring B as described herein, can be combined with any of the embodiments described herein for R3, R4a, R4b, R5a, R5b, R5c, X6, R6a, R6b, R6d, R7a, R7b, and R7c.


Residues 3 to 8

In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein

    • R3 is




embedded image




    • R4a and R4b are each H;

    • R4c is ethyl;

    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a pyrrolidinyl, substituted with 0 to 2 fluoro;

    • R5a and R5b are each H;

    • R5c is H, methyl, ethyl,







embedded image




    • X6 is







embedded image




    • R6a is H, methyl, ethyl,







embedded image




    • R6b is H;

    • R6d is H, methyl, or ethyl;

    • R7a and R7b are each H;

    • R7c is isobutyl,







embedded image




    • R8a is H or methyl;

    • R8b, R8d and R8e are each H;

    • ring B is phenyl, naphthyl, biphenyl, thiophenyl, pyridyl, benzofuranyl, indolyl, indazolyl, or quinolinyl;

    • m8 is 1, 2, or 3; and

    • each R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro, cyano,







embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




    • alternatively, ring B and three R8f groups form the moiety







embedded image


wherein the wavy line identifies attachment to the remainder of the molecule.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein

    • R3 is




embedded image




    • R4a and R4b are each H;

    • R4c is ethyl;

    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a pyrrolidinyl, substituted with 0 to 2 fluoro;

    • R5a and R5b are each H;

    • R5c is H, methyl, ethyl,







embedded image




    • X6 is







embedded image




    • R6a is H, methyl, ethyl, or







embedded image




    • R6b is H;

    • R6d is H, methyl, or ethyl;

    • R7a and R7b are each H;

    • R7c is isobutyl,







embedded image




    • R8a is H or methyl;

    • R8b, R8d and R8e are each H;

    • ring B is phenyl, biphenyl, or pyrid-3-yl;

    • m8 is 1, 2, or 3; and

    • each R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro, cyano,







embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image




    • alternatively, ring B and three R8f groups form the moiety







embedded image




    • wherein the wavy line identifies attachment to the remainder of the molecule.





In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1), (Id), or (Id1), wherein

    • R3 is




embedded image




    • R4a and R4b are each H;

    • R4c is ethyl;

    • alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a pyrrolidinyl, substituted with 0 to 2 fluoro;

    • R5a and R5b are each H;

    • R5c is H, methyl, ethyl







embedded image




    • X6 is







embedded image




    • R6a is H, methyl, ethyl, or







embedded image




    • R6b is H;

    • R6d is H or methyl;

    • R7a and R7b are each H;

    • R7c is isobutyl,







embedded image




    • R8a is H or methyl;

    • R8b, R8d and R8e are each H;

    • ring B is phenyl or pyrid-3-yl;

    • m8 is 1, 2, or 3; and

    • each R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro,







embedded image


embedded image


embedded image


embedded image


embedded image




    • alternatively, ring B and three R8f groups form the moiety







embedded image


wherein the wavy line identifies attachment to the remainder of the molecule.


For the above embodiment, R3, R4a, R4b, R4c, R5a, R5b, R5e, X6, R6a, R6b, R6d, R7a, R7b, R7c, R8a, R8b, R8d, R8e, ring B, m8, and R8f can each independently be as defined for any embodiment of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) as described herein.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 1-426. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 1-389. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 1-334. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 1-50. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 51-100. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 101-150. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 151-200. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 201-250. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 251-300. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 301-334. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 335-389. In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) having the structure of any one of Examples 390-426.


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


In some embodiments, the compound, or the pharmaceutically acceptable salt thereof, is the compound of Formula (I) having the structure




embedded image


The present disclosure includes all tautomers and stereoisomers of the compounds described herein, either in admixture or in pure or substantially pure form. The compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can have asymmetric centers at one or more carbon atoms, and therefore compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can exist in diastereomeric or enantiomeric forms or mixtures thereof. All conformational isomers (e.g., cis and trans isomers) and all optical isomers (e.g., enantiomers and diastereomers), racemic, diastereomeric and other mixtures of such isomers, as well as solvates, hydrates, and tautomers are within the scope of the present disclosure. Compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can be prepared using diastereomers, enantiomers or racemic mixtures as starting materials. Furthermore, diastereomer and enantiomer products can be separated by chromatography, fractional crystallization or other methods known to those of skill in the art. Unless otherwise indicated, when a stereochemical depiction is shown, it is meant that the isomer with the depicted stereochemistry is present and substantially free of the other isomer(s). “Substantially free of” another isomer indicates at least an 80/20 ratio of the two isomers, more preferably 90/10, or 95/5 or more. When a structure includes a wavy bond attached to a double bond, this indicates E, Z, or a mixture of both isomers.


The compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can also be in the salt forms, such as acid or base salts of the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1). Illustrative examples of pharmaceutically acceptable salts are mineral acid (hydrochloric acid, hydrobromic acid, phosphoric acid, and the like) salts, organic acid (acetic acid, propionic acid, glutamic acid, citric acid and the like) salts, quaternary ammonium (methyl iodide, ethyl iodide, and the like) salts. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, which is incorporated herein by reference.


Pharmaceutically acceptable salts of the acidic compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) are salts formed with bases, namely cationic salts such as alkali and alkaline earth metal salts, such as sodium, lithium, potassium, calcium, magnesium, as well as ammonium salts, such as ammonium, trimethyl-ammonium, diethylammonium, and tris-(hydroxymethyl)-methyl-ammonium salts.


Similarly acid addition salts, such as of mineral acids, organic carboxylic and organic sulfonic acids, e.g., hydrochloric acid, methanesulfonic acid, maleic acid, are also possible provided a basic group, such as pyridyl, constitutes part of the structure.


The neutral forms of the compounds can be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present disclosure.


The present disclosure also includes isotopically-labeled compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1), wherein one or more atoms are replaced by one or more atoms having specific atomic mass or mass numbers. Examples of isotopes that can be incorporated into compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) include, but are not limited to, isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, sulfur, and chlorine (such as 2H, 3H, 13C, 14C, 15N, 18O, 17O, 18F, 35S and 36Cl). Isotopically-labeled compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can be useful in assays of the tissue distribution of the compounds and their prodrugs and metabolites; preferred isotopes for such assays include 3H and 14C. In addition, in certain circumstances substitution with heavier isotopes, such as deuterium (2H), can provide increased metabolic stability, which offers therapeutic advantages such as increased in vivo half-life or reduced dosage requirements. Isotopically-labeled compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can generally be prepared according to methods known in the art.


IV. Compositions

The compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein are useful in the manufacture of a pharmaceutical composition or a medicament for modulating one or more cyclins (e.g. cyclin A, cyclin B, cycline E). In some embodiments, the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure, and a pharmaceutically acceptable excipient. In some embodiments, a pharmaceutical composition or medicament comprising one or more compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can be administered to a subject for the treatment of a cancer.


Pharmaceutical compositions or medicaments for use in the present disclosure can be formulated by standard techniques or methods well-known in the art of pharmacy using one or more physiologically acceptable carriers or excipients. Suitable pharmaceutical carriers are described herein and in, e.g., “Remington's Pharmaceutical Sciences” by E. W. Martin. Compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) and their physiologically acceptable salts and solvates can be formulated for administration by any suitable route, including, but not limited to, orally, topically, nasally, rectally, pulmonary, parenterally (e.g., intravenously, subcutaneously, intramuscularly, etc.), and combinations thereof. In some embodiments, the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) is dissolved in a liquid, for example, water. The most suitable route of administration for a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) in any given case will depend, in part, on the nature, severity, and optionally, and the stage of the cancer.


The pharmaceutical compositions or medicaments of the present disclosure can include a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) with as an active ingredient and a pharmaceutically acceptable carrier and/or excipient or diluent. Any carrier and/or excipient suitable for the form of preparation desired for administration is contemplated for use with the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) disclosed herein.


In some embodiments, the pharmaceutical compositions or medicaments described herein are suitable for systemic administration. Systemic administration includes enteral administration (e.g., absorption of the compound through the gastrointestinal tract) or parenteral administration (e.g., injection, infusion, or implantation). In some embodiments, the pharmaceutical compositions or medicaments can be administered via a syringe or intravenously. In preferred embodiments, the pharmaceutical compositions or medicaments are injected subcutaneously.


For oral administration, a pharmaceutical composition or a medicament can take the form of, e.g., a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient. Preferred are tablets and gelatin capsules comprising the active ingredient(s), together with (a) diluents or fillers, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose (e.g., ethyl cellulose, microcrystalline cellulose), glycine, pectin, polyacrylates and/or calcium hydrogen phosphate, calcium sulfate, (b) lubricants, e.g., silica, anhydrous colloidal silica, talcum, stearic acid, its magnesium or calcium salt (e.g., magnesium stearate or calcium stearate), metallic stearates, colloidal silicon dioxide, hydrogenated vegetable oil, corn starch, sodium benzoate, sodium acetate and/or polyethyleneglycol; for tablets also (c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone and/or hydroxypropyl methylcellulose; if desired (d) disintegrants, e.g., starches (e.g., potato starch or sodium starch), glycolate, agar, alginic acid or its sodium salt, or effervescent mixtures; (e) wetting agents, e.g., sodium lauryl sulfate, and/or (f) absorbents, colorants, flavors and sweeteners. In some embodiments, the tablet contains a mixture of hydroxypropyl methylcellulose, polyethyleneglycol 6000 and titatium dioxide. Tablets can be either film coated or enteric coated according to methods known in the art.


Liquid preparations for oral administration can take the form of, for example, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives, for example, suspending agents, for example, sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, for example, lecithin or acacia; non-aqueous vehicles, for example, almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, for example, methyl or propyl-p-hydroxybenzoates or sorbic acid. The preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate. If desired, preparations for oral administration can be suitably formulated to give controlled release of the active compound.


Typical formulations for topical administration include creams, ointments, sprays, lotions, and patches. The pharmaceutical composition can, however, be formulated for any type of administration, e.g., intradermal, subdermal, intravenous, intramuscular, intranasal, intracerebral, intratracheal, intraarterial, intraperitoneal, intravesical, intrapleural, intracoronary or intratumoral injection, with a syringe or other devices. Formulation for administration by inhalation (e.g., aerosol), or for oral, rectal, or vaginal administration is also contemplated.


Pharmaceutical compositions for pulmonary administration include, but are not limited to, dry powder compositions consisting of the powder of a compound described herein, or a salt thereof, and the powder of a suitable carrier and/or lubricant. The compositions for pulmonary administration can be inhaled from any suitable dry powder inhaler device known to a person skilled in the art. In certain instances, the compositions can be conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer, with the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas. In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, for example, gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound(s) and a suitable powder base, for example, lactose or starch.


The compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) can also be formulated in rectal compositions, for example, suppositories or retention enemas, for example, containing conventional suppository bases, for example, cocoa butter or other glycerides.


The compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) set forth herein can be formulated for parenteral administration by injection, for example by bolus injection. Formulations for injection can be presented in unit dosage form, for example, in ampoules or in multi-dose containers, with an added preservative. Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are preferably prepared from fatty emulsions or suspensions. The compositions can be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. Alternatively, the compound(s) can be in powder form for reconstitution with a suitable vehicle, for example, sterile pyrogen-free water, before use. In addition, they may also contain other therapeutically valuable substances. The compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the compound(s).


In some embodiments, the compositions described herein are prepared with a polysaccharide such as chitosan or derivatives thereof (e.g., chitosan succinate, chitosan phthalate, etc.), pectin and derivatives thereof (e.g., amidated pectin, calcium pectinate, etc.), chondroitin and derivatives thereof (e.g., chondroitin sulfate), and alginates.


In some embodiments, the compositions described herein further include a pharmaceutical surfactant. In other embodiments, the compositions further include a cryoprotectant. Non-limiting examples of cryoprotectants include glucose, sucrose, trehalose, lactose, sodium glutamate, PVP, cyclodextrin, 2-hydroxypropyl-β-cyclodextrin (HPI3CD) glycerol, maltose, mannitol, saccharose, and mixtures thereof.


V. Methods

The present disclosure contemplates the use of the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein in the treatment or prevention of diseases or disorders modulated, at least in part, by one or more cyclins. In some embodiments, the cyclin mediated disease is a proliferative condition or disorder, including cancer. In some embodiments, the present disclosure provides a method of treating a cancer mediated at least in part by cyclin activity, the method comprising administering to a subject in need there of, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure, thereby treating the cancer.


In some embodiments, provided herein are compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for use in therapy.


The present disclosure contemplates the use of the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein in the treatment or prevention of diseases or disorders modulated, at least in part, by cyclin A. In some embodiments, the cyclin A mediated disease is a proliferative condition or disorder, including cancer. In some embodiments, the present disclosure provides a method of treating a cancer mediated at least in part by cyclin A, the method comprising administering to a subject in need there of, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure, thereby treating the cancer.


In some embodiments, provided herein are methods of treating a proliferative condition or disorder mediated at least in part by cyclin A comprising administering a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein.


In some embodiments, provided herein are compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for use in a method for treating a proliferative condition or disorder mediated at least in part by cyclin A.


In some embodiments, provided herein are uses of compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for the manufacture of a medicament for the treatment of a proliferative condition or disorder mediated at least in part by cyclin A.


The present disclosure contemplates the use of the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein in the treatment or prevention of diseases or disorders modulated, at least in part, by cyclin B. In some embodiments, the cyclin B mediated disease is a proliferative condition or disorder, including cancer. In some embodiments, the present disclosure provides a method of treating a cancer mediated at least in part by cyclin B, the method comprising administering to a subject in need there of, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure, thereby treating the cancer.


In some embodiments, provided herein are methods of treating a proliferative condition or disorder mediated at least in part by cyclin B comprising administering a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein.


In some embodiments, provided herein are compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for use in a method for treating a proliferative condition or disorder mediated at least in part by cyclin B.


In some embodiments, provided herein are uses of compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for the manufacture of a medicament for the treatment of a proliferative condition or disorder mediated at least in part by cyclin B.


The present disclosure contemplates the use of the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein in the treatment or prevention of diseases or disorders modulated, at least in part, by cyclin E. In some embodiments, the cyclin E mediated disease is a proliferative condition or disorder, including cancer. In some embodiments, the present disclosure provides a method of treating a cancer mediated at least in part by cyclin E, the method comprising administering to a subject in need there of, a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present disclosure, thereby treating the cancer.


In some embodiments, provided herein are methods of treating a proliferative condition or disorder mediated at least in part by cyclin E comprising administering a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein.


In some embodiments, provided herein are compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for use in a method for treating a proliferative condition or disorder mediated at least in part by cyclin E.


In some embodiments, provided herein are uses of compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) for the manufacture of a medicament for the treatment of a proliferative condition or disorder mediated at least in part by cyclin E.


In some embodiments, the compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein can be used to treat or prevent a proliferative condition or disorder, including a cancer, for example, cancer of the uterus, cervix, breast, prostate, testes, gastrointestinal tract (e.g., esophagus, oropharynx, stomach, small or large intestines, colon, or rectum), kidney, renal cell, bladder, bone, bone marrow, skin, head or neck, liver, gall bladder, bile ducts, heart, lung (e.g., non-small-cell lung carcinoma, small cell lung cancer), pancreas, salivary gland, adrenal gland, thyroid, brain, ganglia, central nervous system (CNS) and peripheral nervous system (PNS), and cancers of the hematopoietic system and the immune system (e.g., spleen or thymus).


The present disclosure also provides methods of treating or preventing other cancer-related diseases, disorders or conditions, including, for example, virus-induced cancers (e.g., epithelial cell cancers, endothelial cell cancers, squamous cell carcinomas and papillomavirus), adenocarcinomas, lymphomas, carcinomas, melanomas, leukemias, myelomas, sarcomas, teratocarcinomas, chemically-induced cancers, metastasis, and angiogenesis.


In some embodiments, the tumor or cancer is colon cancer, ovarian cancer, breast cancer, melanoma, lung cancer, glioblastoma, or leukemia.


In some embodiments, the tumor or cancer is small cell lung cancer (SCLC).


The use of the term(s) cancer-related diseases, disorders and conditions is meant to refer broadly to conditions that are associated, directly or indirectly, with cancer, and includes, e.g., angiogenesis and precancerous conditions such as dysplasia.


In some embodiments, the cancer is a blood cancer (e.g., leukemia, lymphoma, multiple myeloma).


In some embodiments, the leukemia is acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, or hairy cell leukemia.


In some embodiments, the lymphoma is non-Hodgkin's lymphoma, Hodgkin's lymphoma, B-cell lymphoma, or Burkitt's lymphoma.


In some embodiments, the cancer is an Rb mutated cancer. In some embodiments, the cancer has a mutation in the Rb/E2F pathway.


VI. Administration

The present disclosure contemplates the administration of compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) and compositions thereof, in any appropriate manner. Suitable routes of administration include oral, parenteral (e.g., intramuscular, intravenous, subcutaneous (e.g., injection or implant), intraperitoneal, intracisternal, intraarticular, intraperitoneal, intracerebral (intraparenchymal) and intracerebroventricular), nasal, vaginal, sublingual, intraocular, rectal, topical (e.g., transdermal), buccal and inhalation.


Pharmaceutical compositions comprising compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) are preferably in unit dosage form. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.


Compounds of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) or pharmaceutical compositions or medicaments thereof can be administered to a subject diagnosed or suspected of having a disease or disorder mediated at least in part by cyclin A in an amount sufficient to elicit an effective therapeutic response in the subject.


The dosage of compounds administered is dependent on a variety of factors including the subject's body weight, age, individual condition, and/or on the form of administration. The size of the dose will also be determined by the existence, nature, and extent of any adverse effects that accompany the administration of a particular compound in a particular subject. Typically, a dosage of the active compounds is a dosage that is sufficient to achieve the desired effect. Optimal dosing schedules can be calculated from measurements of compound accumulation in the body of a subject. In general, dosage can be given once or more daily, weekly, or monthly. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies, and repetition rates.


In some embodiments, a unit dosage for oral administration of a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein to a subject (e.g., a human) of about 50 to about 70 kg may contain between about 1 and about 5,000 mg, about 1 and about 3,000 mg, about 1 and about 2,000 mg, or about 1 to about 1,000 mg of the compound(s).


In some embodiments, a unit dosage for subcutaneous administration of a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein to a subject (e.g., human) of about 50 to about 70 kg may contain between about 0.1 and about 500 mg, about 0.5 and about 300 mg, about 0.5 and about 200 mg, about 0.5 and about 100 mg, or about 0.5 and about 50 mg.


The dose can be administered once per day or divided into sub-doses and administered in multiple doses, e.g., twice, three times, or four times per day. However, as will be appreciated by a skilled artisan, depending on the route of administration different amounts can be administered at different times.


In some embodiments, the compounds are administered for about 1 to 31 days, or for about 1 to 12 months. In some embodiments, the compounds are administered for one or more weeks, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or more weeks. In some embodiments, the compounds are administered for one or more months, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more months.


Optimum dosages, toxicity, and therapeutic efficacy of such compounds may vary depending on the relative potency of individual compounds and can be determined by standard pharmaceutical procedures in experimental animals, for example, by determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio, LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side-effects can be used, care should be taken to design a delivery system that targets such compounds to the affected site to minimize potential damage to normal cells and, thereby, reduce side-effects.


The dosage of a pharmaceutical composition or medicament of the present disclosure can be monitored and adjusted throughout treatment, depending on severity of symptoms, frequency of recurrence, and/or the physiological response to the therapeutic regimen. Those of skill in the art commonly engage in such adjustments in therapeutic regimens.


Single or multiple administrations of the pharmaceutical compositions or medicaments can be administered depending on the dosage and frequency as required and tolerated by the patient. In any event, the composition or medicament should provide a sufficient quantity of the compounds of the disclosure to effectively treat the patient. Generally, when treating cancer, the dose is sufficient to stop tumor growth or cause tumor regression without producing unacceptable toxicity or side-effects to the patient.


VII. Intermediates

In some embodiments, the present disclosure provides intermediates useful in the preparation of compounds of Formula (I). Certain intermediates useful in the preparation of a compound of Formula (I) can be found, for example, in the Examples section of the current disclosure.


In some embodiments, the intermediate is an External Building Block described herein. In some embodiments the intermediate is a compound produced in one of Methods A-D or Methods 1-14 for any one of the compounds exemplified herein. In some embodiments, the intermediate is one of Int. 1-438. In some embodiments, the intermediate is one of Int. 1-405. In some embodiments, the intermediate is one of Int. 1-154.


In some embodiments, the intermediate is Int. 7




embedded image


In some embodiments, the intermediate is Int. 30




embedded image


In some embodiments, the intermediate is Int. 142




embedded image


In some embodiments, the intermediate is a combination of one or more convalently linked External Building Blocks.


VIII. Kits

The present disclosure contemplates kits comprising a compound of Formula (I), (Ib), (Ib1), (Ic), (Ic1) (Id), or (Id1) described herein described herein, and pharmaceutical compositions thereof. The kits are generally in the form of a physical structure housing various components, as described below, and can be utilized, for example, in practicing the methods described above.


A kit can include one or more of the compounds disclosed herein (provided in, e.g., a sterile container), which may be in the form of a pharmaceutical composition suitable for administration to a subject. The compounds described herein can be provided in a form that is ready for use (e.g., a tablet, capsule, syringe) or in a form requiring, for example, reconstitution or dilution (e.g., a powder) prior to administration. When the compounds described herein are in a form that needs to be reconstituted or diluted by a user, the kit may also include diluents (e.g., sterile water), buffers, pharmaceutically acceptable excipients, and the like, packaged with or separately from the compounds described herein. Each component of the kit can be enclosed within an individual container, and all of the various containers can be within a single package. A kit of the present disclosure can be designed for conditions necessary to properly maintain the components housed therein (e.g., refrigeration or freezing).


A kit may contain a label or packaging insert including identifying information for the components therein and instructions for their use (e.g., dosing parameters, clinical pharmacology of the active ingredient(s), including mechanism of action, pharmacokinetics and pharmacodynamics, adverse effects, contraindications, etc.). Labels or inserts can include manufacturer information such as lot numbers and expiration dates. The label or packaging insert may be, e.g., integrated into the physical structure housing the components, contained separately within the physical structure, or affixed to a component of the kit (e.g., an ampule, tube or vial).


Labels or inserts can additionally include, or be incorporated into, a computer readable medium, such as a disk (e.g., hard disk, card, memory disk), optical disk such as CD- or DVD-ROM/RAM, DVD, MP3, magnetic tape, or an electrical storage media such as RAM and ROM or hybrids of these such as magnetic/optical storage media, FLASH media or memory-type cards. In some embodiments, the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e.g., via the internet, are provided.


IX. EXAMPLES

The following examples illustrate how various intermediates and exemplary compounds of Formula (I) are prepared. The following examples are offered to illustrate, but not to limit the current disclosure.


A. External Building Blocks

The compounds of Formula (I) described herein are prepared by covalently linking the external building blocks described in this section. The external building blocks of the present disclosure are identified in Table 1, below, by intermediate number (INT #), IUPAC name, and CAS number, if known. For those without a CAS number, an experimental write-up is provided herein. The order and details related to covalently linking these external building blocks are described in another section.









TABLE 1







External Building Blocks of the Present Disclosure











CAS Number,


INT #
IUPAC
if known





Int. 1
hex-5-en-1-amine
34825-70-2


Int. 2
4-Penten-1-amine
22537-07-1


Int. 3
N-Methylhex-5-en-1-amine
55863-02-0


Int. 4
N-Methyl-4-penten-1-amine
5831-72-1


Int. 5
3-Buten-1-amine
2524-49-4


Int. 6
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-



(2,5-dichlorophenyl)propanoic acid


Int. 7
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-cyclopropoxypyridin-3-yl)propanoic acid


Int. 8
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-(5-methyl-1,3,4-oxadiazol-2-



yl)phenyl)propanoic acid


Int. 9
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(2-



cyclopropoxy-5-fluoropyridin-3-yl)propanoic acid


Int. 10
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-isopropoxypyridin-3-yl)propanoic acid


Int. 11
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-(pyrimidin-2-yloxy)phenyl)propanoic



acid


Int. 12
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-cyclobutoxypyridin-3-yl)propanoic acid


Int. 13
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-methoxypyridin-3-yl)propanoic acid


Int. 14
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-ethoxypyridin-3-yl)propanoic acid


Int. 15
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-isobutoxypyridin-3-yl)propanoic acid


Int. 16
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-



yl)phenyl)propanoic acid


Int. 17
(S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-



chlorophenyl)-2-((tert-



butoxycarbonyl)(methyl)amino)propanoic acid


Int. 18
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridin-3-



yl)propanoic acid


Int. 19
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridin-3-



yl)propanoic acid


Int. 20
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-(1-



methylcyclopropoxy)phenyl)propanoic acid


Int. 21
(S)-2-((((9H-fluoren-9-
146549-21-5



yl)methoxy)carbonyl)amino)pent-4-enoic acid


Int. 22
(S)-3-(1′-acetyl-5-chloro-1′,2′,3′,6′-tetrahydro-



[2,4′-bipyridin]-3-yl)-2-((tert-



butoxycarbonyl)(methyl)amino)propanoic acid


Int. 23
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-cyclopropoxyphenyl)propanoic acid


Int. 24
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-morpholinophenyl)propanoic acid


Int. 25
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-morpholinopyridin-3-yl)propanoic acid


Int. 26
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(7-



chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-



yl)propanoic acid


Int. 27
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-methylpyridin-3-yl)propanoic acid


Int. 28
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-



chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridin-



3-yl)propanoic acid


Int. 29
(S)-3-(2-((1-acetylazetidin-3-yl)oxy)-5-



chloropyridin-3-yl)-2-((tert-



butoxycarbonyl)(methyl)amino)propanoic acid


Int. 30
(S)-3-(2-bromo-5-chlorophenyl)-2-((tert-



butoxycarbonyl)(methyl)amino)propanoic acid


Int. 31
(S)-3-(2-bromo-5-fluorophenyl)-2-((tert-



butoxycarbonyl)(methyl)amino)propanoic acid


Int. 32
(tert-butoxycarbonyl)-L-alanine
15761-38-3


Int. 33
((2S,4R)-4-fluoro-1-(1-



(trifluoromethyl)cyclopropane-1-



carbonyl)pyrrolidine-2-carbonyl)glycine


Int. 34
((S)-2-(1-(trifluoromethyl)cyclopropane-1-



carboxamido)butanoyl)-L-alanine


Int. 35
(S)-2-((2S,4R)-4-fluoro-1-(1-



(trifluoromethyl)cyclopropane-1-



carbonyl)pyrrolidine-2-carboxamido)butanoic acid


Int. 36
ethyl carbonochloridate
541-41-3


Int. 37
isobutyryl chloride
79-30-1


Int. 38
pivaloyl chloride
3282-30-2


Int. 39
(2S,4R)-4-fluoro-1-(2-(trifluoromethyl)tetrahydro-



2H-pyran-2-carbonyl)pyrrolidine-2-carboxylic



acid


Int. 40
1-(4-((4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-



yl)methylene)piperidin-1-yl)ethan-1-one


Int. 41
(S)-2-((tert-butoxycarbonyl)(ethyl)amino)pent-4-



enoic acid


Int. 42
(S)-2-((tert-butoxycarbonyl)(methyl)amino)hex-5-



enoic acid


Int. 43
(S)-2-((tert-butoxycarbonyl)(methyl)amino)hept-6-



enoic acid


Int. 44
1-(methylsulfonyl)-3-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole


Int. 45
1-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-



yl)-8-azabicyclo[3.2.1]oct-2-en-8-yl)ethan-1-one


Int. 46
2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-
287944-16-5



tetramethyl-1,3,2-dioxaborolane


Int. 47
1-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1227068-67-8



yl)-3,6-dihydropyridin-1(2H)-yl)ethan-1-one


Int. 48
(3-chlorophenyl)boronic acid
63503-60-6


Int. 49
2-(4,4-difluorocyclohex-1-en-1-yl)-4,4,5,5-
1227068-84-9



tetramethyl-1,3,2-dioxaborolane


Int. 50
1-(methylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-
1382137-67-8



dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine


Int. 51
4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1310384-24-7



yl)cyclohex-3-en-1-ol


Int. 52
1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-
761446-44-0



dioxaborolan-2-yl)-1H-pyrazole


Int. 53
1-(oxetan-3-yl)-4-(4,4,5,5-tetramethyl-1,3,2-
1339890-99-1



dioxaborolan-2-yl)-1H-pyrazole


Int. 54
2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1040377-08-9



yl)-1H-pyrazol-1-yl)ethan-1-ol


Int. 55
pyrimidin-5-ylboronic acid
109299-78-7


Int. 56
1-(2-methoxyethyl)-4-(4,4,5,5-tetramethyl-1,3,2-
847818-71-7



dioxaborolan-2-yl)-1H-pyrazole


Int. 57
2-(cyclopent-1-en-1-yl)-4,4,5,5-tetramethyl-1,3,2-
287944-10-9



dioxaborolane


Int. 58
1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-
454482-11-2



dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine


Int. 59
1-cyclopropyl-4-(4,4,5,5-tetramethyl-1,3,2-
1151802-22-0



dioxaborolan-2-yl)-1H-pyrazole


Int. 60
1-(methoxymethyl)-4-(4,4,5,5-tetramethyl-1,3,2-
1301198-65-1



dioxaborolan-2-yl)-1H-pyrazole


Int. 61
1-(2,2-difluoroethyl)-4-(4,4,5,5-tetramethyl-1,3,2-
1048730-40-6



dioxaborolan-2-yl)-1H-pyrazole


Int. 62
1-(tetrahydro-2H-pyran-4-yl)-4-(4,4,5,5-
1040377-03-4



tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole


Int. 63
1-(tetrahydrofuran-3-yl)-4-(4,4,5,5-tetramethyl-
1029715-63-6



1,3,2-dioxaborolan-2-yl)-1H-pyrazole


Int. 64
1-(methylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-
944994-03-0



dioxaborolan-2-yl)-1H-pyrazole


Int. 65
1-cyclobutyl-4-(4,4,5,5-tetramethyl-1,3,2-
1002309-48-9



dioxaborolan-2-yl)-1H-pyrazole


Int. 66
cyclopropyl(4-(4,4,5,5-tetramethyl-1,3,2-
1616388-38-5



dioxaborolan-2-yl)-3,6-dihydropyridin-1(2H)-



yl)methanone


Int. 67
4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1121057-75-7



1,2,3,6-tetrahydropyridine hydrochloride


Int. 68
4,4,5,5-tetramethyl-2-(prop-1-en-2-yl)-1,3,2-
126726-62-3



dioxaborolane


Int. 69
4,4,5,5-tetramethyl-2-(2-methylprop-1-en-1-yl)-
126689-00-7



1,3,2-dioxaborolane


Int. 70
2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-
1218791-01-5



dioxaborolan-2-yl)thiazole


Int. 71
1-isopropyl-4-(4,4,5,5-tetramethyl-1,3,2-
879487-10-2



dioxaborolan-2-yl)-1H-pyrazole


Int. 72
cyclopropylboronic acid
411235-57-9


Int. 73
3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1314138-13-0



5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole


Int. 74
1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-
1083180-01-1



dioxaborolan-2-yl)-1H-imidazole


Int. 75
4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
269410-08-4



1H-pyrazole


Int. 76
benzo[c][1,2,5]oxadiazol-5-ylboronic acid
426268-09-9


Int. 77
6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1218790-54-5



[1,2,5]oxadiazolo[3,4-b]pyridine


Int. 78
6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1416437-27-8



yl)pyrazolo[1,5-a]pyrimidine


Int. 79
3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-
1257554-02-1



dioxaborolan-2-yl)-3H-imidazo[4,5-b]pyridine


Int. 80
6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1314137-24-0



[1,2,4]triazolo[4,3-a]pyridine


Int. 81
7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1210048-18-2



[1,2,4]triazolo[1,5-a]pyridine


Int. 82
(6-(ethylthio)pyridin-3-yl)boronic acid
1217501-38-6


Int. 83
(6-ethoxypyridin-3-yl)boronic acid
612845-44-0


Int. 84
1-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-
1256359-15-5



dioxaborolan-2-yl)-1H-pyrazole


Int. 85
3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1383675-83-9



6,7-dihydro-5H-pyrazolo[5,1-b][1,3]oxazine


Int. 86
pyridin-4-ylboronic acid hydrate
1692-15-5


Int. 87
(2-methoxypyridin-4-yl)boronic acid
762262-09-9


Int. 88
(5-fluoropyridin-3-yl)boronic acid
872041-86-6


Int. 89
5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1207623-97-9



1,3-dihydro-2H-pyrrolo[2,3-b]pyridin-2-one


Int. 90
1-(difluoromethyl)-4-(4,4,5,5-tetramethyl-1,3,2-
1206640-82-5



dioxaborolan-2-yl)-1H-pyrazole


Int. 91
(6-(methylsulfonyl)pyridin-3-yl)boronic acid
1088496-41-6


Int. 92
(5-(methylthio)pyridin-3-yl)boronic acid
477251-98-2


Int. 93
1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-
1020174-04-2



dioxaborolan-2-yl)-1H-pyrazole


Int. 94
2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-
1192056-62-4



dioxaborolan-2-yl)oxazole


Int. 95
2,4-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-
859833-13-9



dioxaborolan-2-yl)thiazole


Int. 96
(2-oxo-1,2-dihydropyridin-3-yl)boronic acid
951655-49-5


Int. 97
2-(2,5-dihydrofuran-3-yl)-4,4,5,5-tetramethyl-
212127-80-5



1,3,2-dioxaborolane


Int. 98
(3,5-dimethylisoxazol-4-yl)boronic acid
16114-47-9


Int. 99
3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-
936250-20-3



dioxaborolan-2-yl)-1H-pyrazole


Int. 100
5-methyl-4-(4,4,5,5-tetramethyl-1,3,2-
1346808-41-0



dioxaborolan-2-yl)isoxazole


Int. 101
2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1093307-35-7



yl)-1H-pyrazol-1-yl)acetonitrile


Int. 102
4-fluoro-1-isopropyl-2-methyl-6-(4,4,5,5-
1231930-37-2



tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-



benzo[d]imidazole


Int. 103
6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1160790-18-0



[1,2,4]triazolo[1,5-a]pyridine


Int. 104
1,5-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-
1036991-40-8



dioxaborolan-2-yl)-1H-pyrazole


Int. 105
1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-
847818-79-5



dioxaborolan-2-yl)-1H-pyrazole


Int. 106
7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
908268-52-0



yl)imidazo[1,2-a]pyridine


Int. 107
1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-
1227068-65-6



dioxaborolan-2-yl)-5,6-dihydropyridin-2(1H)-one


Int. 108
1-(methyl-d3)-4-(4,4,5,5-tetramethyl-1,3,2-
1022151-50-3



dioxaborolan-2-yl)-1H-pyrazole


Int. 109
(2-methyl-2H-pyrazolo[3,4-b]pyridin-5-yl)boronic
2304634-03-3



acid


Int. 110
1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-
1312312-78-9



dioxaborolan-2-yl)-1H-pyrazolo[3,4-b]pyridine


Int. 111
1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-
1107627-02-0



dioxaborolan-2-yl)-1H-benzo[d]imidazole


Int. 112
1-(cyclopropylsulfonyl)-4-(4,4,5,5-tetramethyl-
1612172-62-9



1,3,2-dioxaborolan-2-yl)-1H-pyrazole


Int. 113
3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1233526-31-2



yl)-1H-pyrazol-1-yl)tetrahydrothiophene 1,1-



dioxide


Int. 114
3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1776065-66-7



yl)-1H-pyrazol-1-yl)thietane 1,1-dioxide


Int. 115
4-(2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
864754-18-7



yl)-1H-pyrazol-1-yl)ethyl)morpholine


Int. 116
1-(2-(methylsulfonyl)ethyl)-4-(4,4,5,5-
1339892-52-2



tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole


Int. 117
4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
568577-88-8



yl)phenyl)morpholine


Int. 118
(4-(methylsulfonyl)phenyl)boronic acid
149104-88-1


Int. 119
(6-acetamidopyridin-3-yl)boronic acid
947533-21-3


Int. 120
(4-(pyrrolidin-1-ylsulfonyl)phenyl)boronic acid
486422-57-5


Int. 121
pyridin-3-ylboronic acid
1692-25-7


Int. 122
1-(3,3-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)-3,6-dihydropyridin-1(2H)-



yl)ethan-1-one


Int. 123
4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
863422-41-7



yl)pyridazine


Int. 124
3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-
1350543-95-1



dioxaborolan-2-yl)pyridazine


Int. 125
3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1169690-88-3



yl)pyrazolo[1,5-a]pyrimidine


Int. 126
pyrazolo[1,5-a]pyrazin-3-ylboronic acid
2304634-86-2


Int. 127
(S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-



(trifluoromethyl)cyclobutane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxamido)acetic acid


Int. 128
tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-
212127-83-8



dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole-1-



carboxylate


Int. 129
(S)-2-cyclopropyl-2-((2S,4R)-4-fluoro-1-(1-



(trifluoromethyl)cyclopropane-1-



carbonyl)pyrrolidine-2-carboxamido)acetic acid


Int. 130
Methylboronic acid
13061-96-6


Int. 131
5-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-
1073354-91-2



yl)benzo[d]thiazole


Int. 132
5-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-
1054483-78-1



yl)pyridin-2(1H)-one


Int. 133
3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
329214-79-1



yl)pyridine


Int. 134
tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-
900503-08-4



dioxaborolan-2-yl)-8-azabicyclo[3.2.1]oct-2-ene-



8-carboxylate


Int. 135
tert-butyl 7-(4,4,5,5-tetramethyl-1,3,2-
1313034-29-5



dioxaborolan-2-yl)-3-oxa-9-azabicyclo[3.3.1]non-



6-ene-9-carboxylate


Int. 137
tert-butyl (4-(4,4,5,5-tetramethyl-1,3,2-
1251732-64-5



dioxaborolan-2-yl)cyclohex-3-en-1-yl)carbamate


Int. 138
tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-
286961-14-6



dioxaborolan-2-yl)-3,6-dihydropyridine-1(2H)-



carboxylate


Int. 140
(2S,4R)-4-fluoro-1-(1-



(trifluoromethyl)cyclopropane-1-



carbonyl)pyrrolidine-2-carboxylic acid


Int. 141
(2S,4R)-1-(3,3-difluoro-1-



(trifluoromethyl)cyclobutane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxylic acid


Int. 142
(2S,4R)-4-fluoro-1-((R)-3,3,3-trifluoro-2-hydroxy-



2-methylpropanoyl)pyrrolidine-2-carboxylic acid


Int. 143
(2S,4R)-4-fluoro-1-(2-



(trifluoromethyl)spiro[3.3]heptane-2-



carbonyl)pyrrolidine-2-carboxylic acid


Int. 144
(2S,4R)-4-fluoro-1-(1-



(trifluoromethyl)cyclobutane-1-



carbonyl)pyrrolidine-2-carboxylic acid


Int. 145
(1-(trifluoromethyl)cyclobutane-1-carbonyl)-L-



proline


Int. 146
(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-



carbonyl)-L-proline


Int. 147
(2S,4R)-1-(1-cyanocyclobutane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxylic acid


Int. 148
(2S,4R)-4-fluoro-1-((R)-2-



(trifluoromethyl)tetrahydro-2H-pyran-2-



carbonyl)pyrrolidine-2-carboxylic acid


Int. 149
(2S,4R)-4-fluoro-1-((S)-2-



(trifluoromethyl)tetrahydro-2H-pyran-2-



carbonyl)pyrrolidine-2-carboxylic acid


Int. 150
(2S,4R)-4-fluoro-1-(3,3,3-trifluoro-2,2-



dimethylpropanoyl)pyrrolidine-2-carboxylic acid


Int. 151
(tert-butoxycarbonyl)-L-leucine
13139-15-6


Int. 152
(S)-2-((tert-butoxycarbonyl)(methyl)amino)pent-4-
136092-76-7



enoic acid


Int. 153
(S)-2-((tert-butoxycarbonyl)amino)-2-
155976-13-9



cyclopropylacetic acid


Int. 154
tert-butyl (S)-2-((((9H-fluoren-9-



yl)methoxy)carbonyl)(methyl)amino)pent-4-



enoate


Int. 155
4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
364794-79-6



yl)benzyl)morpholine


Int. 156
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
938043-30-2



dioxaborolan-2-yl)benzyl)piperazine


Int. 157
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
747413-21-4



dioxaborolan-2-yl)phenyl)piperazine


Int. 158
3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
929203-04-3



yl)phenyl)pyridine


Int. 159
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
747413-21-4



dioxaborolan-2-yl)phenyl)piperazine


Int. 160
4-((4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
1401222-64-7



yl)phenyl)sulfonyl)morpholine


Int. 161
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
1607838-14-1



dioxaborolan-2-yl)phenyl)-1H-pyrazole


Int. 162
(S)-1-(3,3-difluoro-1-



(trifluoromethyl)cyclobutane-1-carbonyl)-4,4-



difluoropyrrolidine-2-carboxylic acid


Int. 163
(3-methyl-4-morpholinophenyl)boronic acid
1426245-63-7


Int. 164
4-(2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
690636-28-3



yl)phenoxy)ethyl)morpholine


Int. 165
4-methyl-7-(4,4,5,5-tetramethyl-1,3,2-
519054-54-7



dioxaborolan-2-yl)-3,4-dihydro-2H-



benzo[b][1,4]oxazine


Int. 166
(3-chloro-4-morpholinophenyl)boronic acid
1426246-59-4


Int. 167
(4-((tetrahydro-2H-pyran-4-yl)oxy)phenyl)boronic
279261-92-6



acid


Int. 168
1-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
912369-50-7



yl)phenyl)piperazine


Int. 169
1-isopropyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
1073354-18-3



dioxaborolan-2-yl)phenyl)piperazine


Int. 170
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
1247000-92-5



dioxaborolan-2-yl)phenyl)piperidine


Int. 171
(3-fluoro-4-morpholinophenyl)boronic acid
279262-09-8


Int. 172
1-methyl-5′-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)-3′H-spiro[azetidine-3,1′-



isobenzofuran]


Int. 173
4-(3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-
957198-29-7



dioxaborolan-2-yl)pyridin-2-yl)morpholine


Int. 174
(2S,4R)-1-(1-(difluoromethyl)-3,3-



difluorocyclobutane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxylic acid


Int. 175
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
124700-48-9



dioxaborolan-2-yl)phenoxy)piperidine


Int. 176
2-((1S,6R)-3-oxabicyclo[4.1.0]heptan-6-yl)-5-



(4,4,5,5-tetraethyl-1,3,2-dioxaborolan-2-



yl)pyridine


Int. 177
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
1447696-00-5



dioxaborolan-2-yl)phenyl)-1,2,3,6-



tetrahydropyridine


Int. 178
8-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)-3,8-



diazabicyclo[3.2.1]octane


Int. 179
(1S,4S)-2-methyl-5-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)-2,5-



diazabicyclo[2.2.1]heptane


Int. 180
(2S,4R)-1-(3,3-difluoro-1-methylcyclobutane-1-



carbonyl)-4-fluoropyrrolidine-2-carboxylic acid


Int. 181
2,2-dimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)morpholine


Int. 182
4-(2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)morpholine


Int. 183
1-(pyridin-2-ylmethyl)-4-(4-(4,4,5,5-tetramethyl-



1,3,2-dioxaborolan-2-yl)phenyl)piperazine


Int. 184
(S)-3-(methoxymethyl)-4-(4-(4,4,5,5-tetramethyl-



1,3,2-dioxaborolan-2-yl)phenyl)morpholine


Int. 185
1-methyl-4-(2-methyl-4-(4,4,5,5-tetramethyl-
1310704-51-8



1,3,2-dioxaborolan-2-yl)phenyl)piperazine


Int. 186
4-fluoro-1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperidine


Int. 187
2-morpholino-5-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)benzonitrile


Int. 188
1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-
1046793-62-7



dioxaborolan-2-yl)cyclohex-3-en-1-yl)piperazine


Int. 189
(2S,4R)-4-fluoro-1-(2,3,3,3-tetrafluoro-2-



methylpropanoyl)pyrrolidine-2-carboxylic acid


Int. 190
3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-



yl)phenyl)-6,7-dihydro-4H-pyrazolo[5,1-



c][1,4]oxazine . . .


Int. 191
1-methyl-4-(4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)-1H-pyrazol-1-



yl)piperidine . . .


Int. 192
(2R,6R)-2,6-dimethyl-4-(4-(4,4,5,5-tetramethyl-



1,3,2-dioxaborolan-2-yl)phenyl)morpholine


Int. 193
tert-butyl 3-(4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperazin-1-



yl)azetidine-1-carboxylate


Int. 194
(4-(1-(4-Methylpiperazin-1-
1643923-77-6



yl)cyclopropyl)phenyl)bornoic acid pinacol ester


Int. 195
N-ethylhex-5-en-1-amine


Int. 196
(2R,6S)-1,2,6-trimethyl-4-(4-(4,4,5,5-tetramethyl-



1,3,2-dioxaborolan-2-yl)phenyl)piperazine . . .


Int. 197
3-[4-(4,4,5,5-Tetramethyl-[1,3,2]dioxaborolan-2-
1362703-30-7



yl)-phenoxy]-pyridine


Int. 198
2,3-Difluoro-4-morpholinophenylboronic acid
2377609-97-5


Int. 199
1-(oxetan-3-yl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperazine


Int. 400
(3aR,6aS)-5-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)hexahydro-1H-furo[3,4-



c]pyrrole


Int. 401
(R)-8-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-



2-yl)phenyl)octahydropyrazino[2,1-c][1,4]oxazine


Int. 402
4-(3-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-
1366131-51-2



dioxaborolan-2-yl)phenyl)morpholine


Int. 403
1-(2-(2-methoxyethoxy)ethyl)-4-(4-(4,4,5,5-



tetramethyl-1,3,2-dioxaborolan-2-



yl)phenyl)piperazine


Int. 404
1-(2,2-difluoroethyl)-4-(4-(4,4,5,5-tetramethyl-



1,3,2-dioxaborolan-2-yl)phenyl)piperazine


Int. 405
1-((1-methyl-1H-pyrazol-4-yl)methyl)-4-(4-



(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-



yl)phenyl)piperazine


Int. 406
(S)-2-((tert-butoxycarbonyl)(methyl)amino)-4-



methoxy-4-oxobutanoic acid


Int. 407
(S)-2-((tert-butoxycarbonyl)amino)-5-methoxy-5-
45214-91-3



oxopentanoic acid


Int. 408
(S)-2-((tert-butoxycarbonyl)(methyl-



d3)amino)pent-4-enoic acid


Int. 409
[4-(4-Methylpiperazine-1-carbonyl)phenyl]boronic
832114-06-4



acid pinacol ester


Int. 410
1-methyl-4-(5-(4,4,5,5-tetramethyl-1,3,2-
918524-63-7



dioxaborolan-2-yl)pyridin-2-yl)piperazine


Int. 411
7-bromo-5-chloro-1-methyl-1H-indole


Int. 412
4-bromo-6-chloro-1-methyl-1H-indole


Int. 413
7-bromo-5-chlorobenzofuran
286836-07-5


Int. 414
2-((1-methylpiperidin-4-yl)oxy)-5-(4,4,5,5-



tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine


Int. 415
(2S,4R)-1-(1-chlorocyclopropane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxylic acid


Int. 416
(2S,4R)-1-((R)-2,2-difluoro-1-



methylcyclopropane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxylic acid


Int. 417
(2S,4R)-1-((S)-2,2-difluoro-1-



methylcyclopropane-1-carbonyl)-4-



fluoropyrrolidine-2-carboxylic acid . . .


Int. 418
1-(2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-
1408088-34-5



dioxaborolan-2-yl)phenyl)-4-methylpiperazine


Int. 419
2-(3-fluoro-3-methylazetidin-1-yl)-5-(4,4,5,5-



tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine


Int. 420
4-(1-(4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-
1206594-12-8



2-yl)phenyl)ethyl)morpholine


Int. 421
2-methyl-7-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)-1,2,3,4-tetrahydroisoquinoline


Int. 422
2-Methyl-1,2,3,4-tetrahydroisoquinoline-6-
922718-57-8



boronicacid pinacol ester


Int. 423
4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-
928631-04-3



yl)phenyl]morpholin-3- one


Int. 424
2-methyl-6-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)-2,6-



diazaspiro[3.4]octane . . .


Int. 425
(S)-1,2-dimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperazine . . .


Int. 426
(S)-N,N-dimethyl-1-(4-(4-(4,4,5,5-tetramethyl-



1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-



yl)methanamine


Int. 427
(R)-1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperidine . . .


Int. 428
(S)-1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperidine


Int. 429
1-(2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)-4-isopropylpiperazine . . .


Int. 430
(R)-2-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)morpholine


Int. 431
(S)-1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-



dioxaborolan-2-yl)phenyl)piperidine


Int. 432
8-Bromo-6-fluoroquinoline
22960-18-5


Int. 433
4-bromo-6-chloro-1-methyl-1H-indazole


Int. 434
1-Bromo-3-chloronaphthalene
51671-06-8


Int. 435
2-bromo-4-chlorothiophene
32431-92-8


Int. 436
5-Bromo-2-cyanopyridine
97483-77-7


Int. 437
1-Methylpiperazine
109-01-3


Int. 438
(S)-3-(2-bromo-5-chlorophenyl)-2-((tert-



butoxycarbonyl)amino)propanoic acid









Int. 6: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(2,5-dichlorophenyl)propanoic acid



embedded image


Step 1: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(2,5-dichlorophenyl)propanoate



embedded image


To a stirred mixture of Zn (29.95 g, 458.06 mmol, 2.5 eq) and TMSCl (1.33 g, 12.28 mmol, 0.067 eq) in DMA was added methyl (2R)-2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (1.03 g, 3.11 mmol, 1.7 eq) dropwise below 45° C. under nitrogen atmosphere. The resulting mixture was stirred for 16 h at rt under nitrogen atmosphere. The above mixture was added into a solution of 1,4-dichloro-2-iodo-benzene (50 g, 183.22 mmol, 1 eq), CuI (6.96 g, 36.64 mmol, 0.2 eq) and Pd(dppf)Cl2·CH2Cl2 (14.93 g, 18.32 mmol, 0.10 eq) at rt under nitrogen. The resulting mixture was stirred for additional 16 h at 85° C. Desired product could be detected by LCMS. The reaction was quenched by the addition of water (1.5 L) at rt. The aqueous layer was extracted with EtOAc (3×500 mL). The combined organics were washed with (5×300 mL) of water and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (25:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(2,5-dichlorophenyl)propanoate (40 g, 63%) as a white solid. LCMS (ESI+): m/z 292.15 (M−56+). 1H NMR (300 MHz, DMSO-d6): δ 1.11-1.47 (m, 12H), 2.89 (dd, 1H), 3.22 (dd, 1H), 3.64 (d, 4H), 3.91-4.37 (m, 1H), 6.87-7.89 (m, 4H).


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(2,5-dichlorophenyl)propanoate



embedded image


To a stirred solution of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(2,5-dichlorophenyl)propanoate (200 mg, 0.574 mmol, 1 eq) and methyl iodide (214.00 g, 1507.67 mmol, 15 eq) in DMF (350 mL) was added (argentiooxy)silver (93.17 g, 402.04 mmol, 4 eq) in portions at 25° C. under nitrogen atmosphere. The reaction mixture was stirred for 16 h at 25° C. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×100 mL). The filtrate was diluted water (500 mL) and extracted with EtOAc (3×300 mL). The combined organic layers were washed with water (4×200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.05% TFA), 10% to 50% gradient in 10 min; detector, UV 220 nm. This resulted in methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(2,5-dichlorophenyl)propanoate as a white solid. LCMS (ESI+): m/z 262.20 (M+Na+).


Step 3: Int. 6



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(2,5-dichlorophenyl)propanoate (35 g, 96.62 mmol, 1 eq) and LiOH (4.63 g, 193.24 mmol, 2 eq) in THF (500 mL)/H2O (300 mg) was stirred for 16 h at 25° C. Desired product could be detected by LCMS. The mixture was acidified to pH=4 with diluted HCl (aq.). The aqueous layer was extracted with EtOAc (3×500 mL). The combined organics was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 220 nm. This resulted in Int. 6 (25.26 g, 75.07%) as a light-yellow solid. LCMS (ESI+): m/z 346.00 (M−H). 1H NMR (400 MHz, DMSO-d6): δ 1.23 (d, 9H), 2.66 (d, 3H), 3.06-3.39 (m, 2H), 7.24-7.56 (m, 3H), 13.00 (s, 1H).


Int. 7: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoic acid



embedded image


Step 1: 3-Bromo-5-chloro-2-cyclopropoxypyridine



embedded image


Into a 1000 mL round-bottom flask were added 3-bromo-5-chloro-2-fluoropyridine (50 g, 237.61 mmol, 1 eq) in DMF (400 mL), Cs2CO3 (232.97 g, 712.83 mmol, 3 eq) was added at rt under nitrogen atmosphere. The mixture was stirred at rt for 50 min. cyclopropanol (9.94 g, 171.08 mmol, 1.2 eq) was added dropwise over 2 min at rt. The resulting mixture was stirred at 80° C. for 16 h. The reaction was quenched with ice-water (700 mL) and extracted with EA (3×100 mL). The organic layer combined and washed with brine (300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (0˜20%). This resulted in 3-bromo-5-chloro-2-cyclopropoxypyridine (40 g, 68%) as a colorless oil. LCMS (ESI+): m/z 249.95. 1H NMR (400 MHz, DMSO-d6) δ 0.76 (ddd, J=5.7, 4.5, 3.1 Hz, 2H), 0.96-0.78 (m, 2H), 4.32 (tt, J=6.4, 3.1 Hz, 1H), 8.17 (d, J=2.5 Hz, 1H), 8.26 (d, J=2.4 Hz, 1H).


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoate



embedded image


To a mixture of Zn (26.84 g, 410.46 mmol, 1.7 eq) in DMA (135 mL) was added 1,2-dibromoethane (6.80 g, 36.22 mmol, 0.15 eq) in one portion under N2. Then chlorotrimethylsilane (2.62 g, 24.15 mmol, 0.1 eq) was added slowly and the mixture was stirred for 30 min at 25° C. A solution of methyl 2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (95.36 g, 289.74 mmol, 1.2 eq) in DMA (135 mL) was added dropwise slowly (60 min) to maintain temperature below 50° C., the resulting mixture was stirred at rt for 2 h and then added 1000 mL 3-necked round-bottom flask a cannula to a solution of 3-bromo-5-chloro-2-cyclopropoxypyridine (60 g, 241.45 mmol, 1 eq), Pd(dppf)Cl2·CH2Cl2 (19.67 g, 24.14 mmol, 0.1 eq) and CuI (9.20 g, 48.29 mmol, 0.2 eq) in DMA (135 ml) under N2, the color of the mixture turned brown, then the mixture was heated and stirred at 80° C. for 2 h under N2. The mixture was quenched with ice-water (700 ml) and extracted with EA (3×500 ml). The organic layer was combined and washed with brine (300 ml), dried with anhydrous Na2SO4, filtered, and concentrated in vacuum to give the crude product. The crude product was purified by silica gel chromatography eluted with PE/EA (0-50%) to afford methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoate (30 g, 34%) as a white solid. LCMS (ESI+): m/z 371.10.


Step 3: Methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoate



embedded image


Into a 1000 mL round-bottom flask were added methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoate (25 g, 67.42 mmol, 1 eq) in DMF (400 mL), Ag2O (78.11 g, 337.08 mmol, 5 eq) was added at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 30 min, CH3I (95.69 g, 674.16 mmol, 10 eq) was added dropwise over 2 min at 0° C. The resulting mixture was stirred at rt for 16 h. The reaction was quenched with ice-water (500 mL) and extracted with EtOAc (3×500 mL). The organic layer was combined and washed with brine (300 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (7:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoate (22 g, 85%) as a colorless oil. LCMS (ESI+): m/z 385.10. 1H NMR (400 MHz, DMSO-d6) δ 0.83-0.62 (m, 2H), 1.19 (t, J=7.1 Hz, 2H), 1.28 (s, 9H), 2.65 (d, J=3.3 Hz, 3H), 2.95 (dd, J=13.6, 10.4 Hz, 1H), 3.07 (ddd, J=14.2, 4.9, 1.9 Hz, 2H), 3.73-3.61 (m, 3H), 4.69 (dd, J=10.5, 4.6 Hz, 1H), 8.12 (dd, J=17.7, 2.6 Hz, 2H).


Step 4: Int. 7



embedded image


Into a 1000 mL round-bottom flask were added methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-cyclopropoxypyridin-3-yl)propanoate (20 g, 51.97 mmol, 1 eq) in THF (250 ml) and NaOH (10.39 g, 259.84 mmol, 5 eq) in water (50 ml) was added dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred at rt for 2 h. The solvent was removed by reduce pressure and the residue was purified by reverse flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.5% FA), 0% to 100% gradient in 40 min; detector, UV 254 nm. This resulted in Int. 7 (13.95 g, 72%) as a yellow oil. LCMS (ESI+): m/z 371.00. 1H NMR (400 MHz, DMSO-d6) δ 0.79-0.61 (m, 4H), 1.21 (s, 9H), 2.95-2.84 (m, 2H), 3.00 (s, 3H), 3.09-3.01 (m, 1H), 4.86 (dd, J=11.3, 4.6 Hz, 1H), 7.67 (d, J=2.6 Hz, 1H), 8.11 (dd, J=18.4, 2.6 Hz, 1H).


Int. 8: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl)propanoic acid



embedded image


Step 1: 4-chloro-2-iodobenzohydrazide



embedded image


Into a stirred mixture of 4-chloro-2-iodobenzoic acid (10 g, 35.40 mmol, 1 eq) and 2-methylpropyl carbonochloridate (4.84 g, 35.40 mmol, 1 eq) in THF (100 mL) were added TEA (10.75 g, 106.21 mmol, 3 eq) and hydrazine (5.67 g, 177.02 mmol, 5 eq) dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred for an additional 4 h at r. Desired product could be detected by LCMS. The resulting mixture was diluted with water (100 mL). The resulting mixture was extracted with EtOAc (3×100 mL). The combined organic layers were washed with 0.5N HCl solution (3×50 mL). The combined aqueous layers were basified to pH 8 with NaOH (0.5N). The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 5% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in 4-chloro-2-iodobenzohydrazide (7.5 g, 72%) as a white solid. LCMS (ESI+): m/z 296.90 (M+H+).


Step 2: 2-(4-chloro-2-iodophenyl)-5-methyl-1,3,4-oxadiazole



embedded image


Into a 200 mL vial were added 4-chloro-2-iodobenzohydrazide (8 g, 26.98 mmol, 1 eq) and triethyl orthoacetate (80 mL) at rt. The resulting mixture was stirred overnight at 140° C. Desired product could be detected by LCMS. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% TFA), 5% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in 2-(4-chloro-2-iodophenyl)-5-methyl-1,3,4-oxadiazole (10.2 g, 118%) as a yellow solid. LCMS (ESI+): m/z 320.85 (M+H+).


Step 3: methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate



embedded image


Into a stirred solution of in Zn (3.84 g, 58.73 mmol, 2 eq) DMA (30 mL) were added 1,2-dibromoethane (0.55 g, 2.937 mmol, 0.1 eq) and TMSCl (0.21 g, 1.967 mmol, 0.067 eq) dropwise at rt under nitrogen atmosphere. To the above mixture was added methyl (2R)-2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (9.66 g, 29.37 mmol, 1 eq) in DMA (30 mL) dropwise. The resulting mixture was stirred for an additional 3 h at rt. To a stirred mixture of 2-(4-chloro-2-iodophenyl)-1,3,4-oxadiazole (9 g, 29.37 mmol, 1 eq) in DMA (30 mL) were added Pd(dppf)Cl2CH2Cl2 (2.39 g, 2.94 mmol, 0.10 eq) and CuI (1.12 g, 5.87 mmol, 0.2 eq) in portions at rt under nitrogen atmosphere. Then, two mixtures were added together. The resulting mixture was stirred for an additional 16 h at rt. The resulting mixture was stirred for additional 3 h at 80° C. Desired product could be detected by LCMS. The reaction was quenched with water 100 mL) at rt. The resulting mixture was filtered, and the filter cake was washed with EtOAc (3×150 mL). The filtrate was concentrated under reduced pressure. The resulting mixture was extracted with EtOAc (3×100 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 5% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (4.5 g, 39%) as a yellow solid. LCMS (ESI+): m/z 396.1 (M+H+).


Step 4: (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoic acid



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (5 g, 12.63 mmol, 1 eq) in THF (40 mL) and water (10 mL) was treated with LiOH (1.51 g, 63.16 mmol, 5 eq) for 4 h at rt. Desired product could be detected by LCMS. The mixture was diluted with water and EA. The water phase was acidified by diluted HCl (0.5N) and extracted with EA. The organic layer was dried over anhydrous Na2SO4 and concentrated in vacuo. This resulted in (2S)-2-[(tert-butoxycarbonyl) amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoic acid (4.5 g) as a light-yellow solid. LCMS (ESI+): m/z 404.0 (M+Na+).


Step 5: methyl (2S)-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]-2-[(isopropoxycarbonyl)(methyl)amino]propanoate



embedded image


A solution of (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoic acid (4.5 g, 11.79 mmol, 1 eq) in DMF (50 mL) was treated with Ag2O (13.66 g, 58.93 mmol, 5 eq) for 20 min at rt under nitrogen atmosphere followed by the addition of CH3I (25.09 g, 176.79 mmol, 15 eq) dropwise at rt. The resulting mixture was stirred for 4 h at rt. The resulting mixture was diluted with water (50 mL). The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (3×50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% TFA), 5% to 100% gradient in 50 min; detector, UV 220 nm. This resulted in methyl (2S)-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]-2-[(isopropoxycarbonyl)(methyl)amino]propanoate (3.7 g, 79%) as a yellow solid. LCMS (ESI+): m/z 410.1 (M+H+).


Step 6: Int. 8



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (2.6 g, 6.34 mmol, 1 eq) in THF (40 mL) and H2O (10 mL) was treated with LiOH (1.08 g, 45.14 mmol, 5 eq) for 4 h at rt under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was diluted with water (20 mL) and acidified to pH 6 with HCl (0.5 mol/L). The mixture was concentrated under reduced pressure and extracted with EtOAc (3×50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% TFA), 5% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in Int. 8 (3.4289 g, 96%) as a yellow solid. LCMS (ESI+): m/z 390.10 (M+H+). 1H NMR (300 MHz, DMSO-d6): δ 1.26 (s, 4H), 1.39 (s, 5H) 2.67 (d, J=1.9 Hz, 3H), 2.84 (d, J=8.8 Hz, 3H), 3.33 (ddd, J=14.3, 10.9, 3.4 Hz, 1H), 3.91 (dt, J=14.6, 2.9 Hz, 1H), 5.06 (ddd, J=18.1, 10.8, 3.8 Hz, 1H), 7.34-7.26 (m, 1H), 7.40 (ddd, J=8.5, 6.1, 2.1 Hz, 1H), 7.90 (dd, J=8.4, 3.8 Hz, 1H), 10.00 (s, 1H).


Int. 9: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(2-cyclopropoxy-5-fluoropyridin-3-yl)propanoic acid



embedded image


Step 1: 3-bromo-2-cyclopropoxy-5-fluoropyridine



embedded image


To a solution of cyclopropanol (15.55 g, 268.10 mmol, 1.3 eq) in THF (500 mL) was added sodium hydride (16.6 g, 691.67 mmol, 2 eq) portion wise at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 1 h. 3-bromo-2,5-difluoropyridine (40 g, 206.21 mmol, 1 eq) was added dropwise over 30 min at 0° C. The resulting mixture was stirred at rt for 16 h. The reaction was quenched with ice-water (700 mL) and extracted with DCM (3×100 mL). The organic layer combined and washed with brine (300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 3-bromo-2-cyclopropoxy-5-fluoropyridine (34 g, crude) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 233.90 (M+H+).


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(2-cyclopropoxy-5-fluoropyridin-3-yl)propanoate



embedded image


To a mixture of Zn (15.94 g, 249.06 mmol, 1.7 eq) in DMA (80 mL) was added 1,2-dibromoethane (4.13 g, 21.98 mmol, 0.15 eq) in one portion under N2. Then chlorotrimethylsilane (1.58 g, 14.66 mmol, 0.1 eq) was added slowly and the mixture was stirred for 30 min at 25° C. A solution of methyl 2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (57.86 g, 175.86 mmol, 1.2 eq) in DMA (80 mL) was added dropwise slowly (60 min) to maintain temperature below 50° C., the resulting mixture was stirred at rt for 2 h and then added 250 mL 3-necked round-bottom flask a cannula to a solution of 3-bromo-2-cyclopropoxy-5-fluoropyridine (34 g, 146.55 mmol, 1 equiv), Pd(dppf)Cl2·CH2Cl2 (23.94 g, 29.31 mmol, 0.2 eq) and CuI (5.57 g, 29.31 mmol, 0.2 eq) in DMA (80 mL) under N2, the color of the mixture turned brown, then the mixture was heated and stirred at 80° C. for 2 h under N2. The mixture was quenched with ice-water (400 ml) and extracted with EA (3×500 ml). The organic layer was combined and washed with brine (300 ml), dried with anhydrous Na2SO4, filtered, and concentrated in vacuum to give the crude product. The crude product was purified by silica gel chromatography eluted with PE/EtOAc=0-50% to afford methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(2-cyclopropoxy-5-fluoropyridin-3-yl)propanoate (11.2 g, 23%) as a yellow oil. LCMS (ESI+): m/z 355.10 (M+H+).


Step 3: Int. 9



embedded image


Into a 1000 mL round-bottom flask were added methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(2-cyclopropoxy-5-fluoropyridin-3-yl)propanoate (11 g, 31.07 mmol, 1 eq) in DMF (150 mL), Ag2O (35.89 g, 155.37 mmol, 5 eq) was added at 0° C. under nitrogen atmosphere. The mixture was stirred at 0° C. for 30 min, methyl iodide (44.12 g, 310.73 mmol, 10 eq) was added dropwise over 2 min at 0° C. The resulting mixture was stirred at rt for 16 h. The reaction was quenched with ice-water (100 mL) and extracted with EtOAc (3×300 mL). The organic layer was combined and washed with brine (3×200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (7:1) to afford Int. 9 (10.8 g, 94%) as a yellow oil. LCMS (ESI+): m/z 369.15 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 8.06 (dd, J=18.7, 3.0 Hz, 1H), 7.55 (ddd, J=18.5, 8.7, 3.0 Hz, 1H), 4.68 (dd, J=10.6, 4.5 Hz, 1H), 4.24 (ddt, J=11.7, 8.6, 4.0 Hz, 1H), 3.68 (d, J=7.6 Hz, 3H), 3.13-2.84 (m, 2H), 2.63 (d, J=7.7 Hz, 3H), 1.24 (d, J=18.3 Hz, 9H), 0.80-0.63 (m, 4H).


Int. 10: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-isopropoxypyridin-3-yl)propanoic acid



embedded image


Step 1: 3-bromo-5-chloro-2-isopropoxypyridine



embedded image


To a stirred mixture of 3-bromo-5-chloro-2-fluoropyridine (50 g, 237.61 mmol, 1 eq) and K2CO3 (98.52 g, 712.83 mmol, 3 eq) in IPA (500 mL, 5261.52 mmol, 22.14 eq) at 20° C. The resulting mixture was stirred for 48 h at 80° C. The resulting mixture was concentrated under vacuum and the mixture was dissolved in DCM (300 mL). The resulting mixture was filtered, and the filter cake was washed with DCM (3×50 mL). The filtrate was concentrated under reduced pressure. This resulted in 3-bromo-5-chloro-2-isopropoxypyridine (50 g, 84.0%) as a yellow oil. LCMS (ESI+): m/z 252.05 (M+H+).


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl) propanoate



embedded image


To a stirred mixture of 3-bromo-5-chloro-2-isopropoxypyridine (50 g, 199.59 mmol, 1 eq) and Pd(dppf)Cl2 DCM adduct (16.26 g, 19.96 mmol, 0.1 eq), CuI (7.60 g, 39.92 mmol, 0.2 eq) in DMA (500 mL) was added methyl (2S)-2-[(tert-butoxycarbonyl)amino]-2-(iodozincio)acetate (162.6 mL, 199.59 mmol, 10 eq) dropwise at 20° C. under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 80° C. under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×500 mL). The filtrate was extracted with EtOAc (3×800 mL). The combined organic layers were washed with H2O (3×1000 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/PE (5:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl)propanoate (110 g) as a yellow oil. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 80% gradient in 35 min; detector, UV 280 nm. The resulting mixture was concentrated under vacuum. This resulted in methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl) propanoate (53 g, 77%) as a white solid. LCMS (ESI+): m/z 373.25 (M+H+).


Step 3: Methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl)propanoate



embedded image


To a stirred mixture of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl)propanoate (20 g, 53.64 mmol, 1 eq) and Ag2O (62.15 g, 268.21 mmol, 5 eq) in DMF (500 mL) was added MeI (77.66 g, 536.41 mmol, 10 eq) at 20° C. The resulting mixture was stirred for 16 h at 60° C. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×500 mL). The filtrate was dissolved in H2O (800 mL) and the mixture was extracted with EtOAc (3×1000 mL). The combined organic layers were washed with H2O (4×1200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl)propanoate (20.5 g, 99%) as a yellow oil. LCMS (ESI+): m/z 387.15 (M+H+).


Step 4: Int. 10



embedded image


To a stirred mixture of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-isopropoxypyridin-3-yl)propanoate (20 g, 51.70 mmol, 1 eq) in EtOH (30 mL) and THF (60 mL) were added NaOH (6.20 g, 155.09 mmol, 3 eq) in H2O (120 mL) dropwise at 0° C. The resulting mixture was stirred for 16 h at 20° C. The resulting mixture was extracted with EtOAc (2×100 mL). The combined organic layers were washed with H2O (1×150 mL). The combined water phase was acidified to pH=6 with diluted citric acid (1N). The resulting mixture was extracted with EtOAc (2×200 mL). The combined organic layers were washed with H2O (1×100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum to afford Int. 10 (14.2 g, 74%) as a yellow oil. LCMS (ESI+): m/z 373.15 (M+H+).


Int. 11: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-(pyrimidin-2-yloxy)phenyl)propanoic acid




embedded image


Step 1: 5-(2-bromo-4-chlorophenyl)pyrimidine



embedded image


To a stirred solution of pyrimidin-5-ylboronic acid (10 g, 80.70 mmol, 1 eq) and 2-bromo-4-chloro-1-iodobenzene (25.61 g, 80.70 mmol, 1 eq) in 1,4-dioxane (80 mL) and H2O (20 mL) were added Pd(dppf)Cl2CH2Cl2 (6.57 g, 8.06 mmol, 0.10 eq) and Na2CO3 (25.66 g, 242.11 mmol, 3 eq) in portions at rt under nitrogen atmosphere. The resulting mixture was stirred for additional 4 h at 80° C. Desired product could be detected by LCMS. The resulting mixture was filtered, and the filter cake was washed with EtOAc (3×10 mL). The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% TFA), 5% to 100% gradient in 30 min; detector, UV 220 nm. This resulted in 5-(2-bromo-4-chlorophenyl)pyrimidine (9.6 g, 44%) as a brown oil. LCMS (ESI+): m/z 270.90 (M+H+).


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(pyrimidin-5-yl)phenyl]propanoate



embedded image


Into a stirred solution of Zn (6.31 g, 96.46 mmol, 2 eq) in DMA (30 mL) were added 1,2-dibromoethane (0.91 g, 4.82 mmol, 0.1 eq) and TMSCl (0.35 g, 3.23 mmol, 0.067 eq) dropwise at rt under nitrogen atmosphere. To the above mixture was added methyl (2R)-2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (15.87 g, 48.23 mmol, 1 eq) in DMA (30 mL) dropwise. The resulting mixture was stirred for additional 3 h at rt. To a stirred solution of 5-(2-bromo-4-chlorophenyl)pyrimidine (13 g, 48.23 mmol, 1 eq) in DMA (40 mL) were added Pd(dppf)Cl2CH2Cl2 (3.93 g, 4.82 mmol, 0.1 eq) and CuI (1.84 g, 9.65 mmol, 0.2 eq) in portions at rt under nitrogen atmosphere. Then, the two mixtures were added together. The resulting mixture was stirred for additional 3 h at 80° C. Desired product could be detected by LCMS. The reaction was quenched by the addition of water (100 mL) at rt. The resulting mixture was filtered, and the filter cake was washed with EA (3×100 mL). The residue was diluted with water and EA. The organic phase was washed with brine (100 mL×3), dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 5% to 100% gradient in 30 min; detector, UV 220 nm. This resulted in methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(pyrimidin-5-yl)phenyl]propanoate (8 g, 42%) as a yellow solid. LCMS (ESI+): m/z 392.05 (M+H+).


Step 3: Methyl(S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-(pyrimidin-2-yloxy)phenyl)propanoate



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(pyrimidin-2-yloxy)phenyl]propanoate (1.3 g, 3.19 mmol, 1 eq) in DMF (15 mL) was treated with Ag2O (3.69 g, 15.94 mmol, 5 eq) at rt under nitrogen atmosphere followed by the addition of CH3I (6.79 g, 47.81 mmol, 15 eq) dropwise at rt. The resulting mixture was stirred for additional 3 h at rt. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with DMF (30 mL). The filtrate was used in the next step directly without further purification. LCMS (ESI+): m/z 378.0 (M+H+).


Step 4: Int. 11



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(pyrimidin-2-yloxy)phenyl]propanoate (3 g, 7.111 mmol, 1 eq) in DMF (45 mL) and H2O (10 mL) was treated with NaOH (0.85 g, 21.333 mmol, 3 eq) at rt. The resulting mixture was stirred for additional 16 h at rt. Desired product could be detected by LCMS. The resulting mixture was diluted with water (40 mL). The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 5% to 100% gradient in 10 min; detector, UV 220 nm. This resulted in Int. 11 (1.86 g, 62%) as a yellow solid. LCMS (ESI+): m/z 408.05 (M+H+).


Int. 12: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-cyclobutoxypyridin-3-yl)propanoic acid



embedded image


Step 1: 3-Bromo-5-chloro-2-cyclobutoxypyridine



embedded image


To a suspension of NaH (6.35 g, 158.67 mmol, 1.2 eq, 60% in oil) in THF (250 mL) was added cyclobutanol (10.49 g, 145.45 mmol, 1.1 eq) in THF (20 mL) at 0° C. dropwise. The resulting mixture was stirred for 1 h at 0° C. under nitrogen atmosphere. 3-bromo-2,5-dichloropyridine (30.0 g, 132.23 mmol, 1.0 eq) in THF (30 mL) was added and the mixture was stirred for 16 h at 60° C. under nitrogen atmosphere. The reaction was quenched by the addition of sat. NH4Cl (aq.) (100 mL) at 0° C. The resulting mixture was diluted with water (500 mL) and then extracted with EtOAc (3×150 mL). The combined organic layers were washed with brine (1×300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (40:1) to afford 3-bromo-5-chloro-2-cyclobutoxypyridine (31.0 g, 89%) as a colorless oil. LCMS (ESI+): m/z 262 (M+H+).


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-cyclobutoxypyridin-3-yl) propanoate



embedded image


To a stirred mixture of 3-bromo-5-chloro-2-cyclobutoxypyridine (15 g, 57.14 mmol, 1.0 eq), Pd(dppf)Cl2·CH2Cl2 (2.33 g, 2.86 mmol, 0.05 eq) and CuI (1.09 g, 5.71 mmol, 0.1 eq) in N, N-dimethylacetamide (300 mL) was added methyl (2R)-2-[(tert-butoxycarbonyl)amino]-3-(iodozincio) propanoate (171.41 mL, 171.41 mmol, 3.00 eq) in one portion at rt under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 80° C. under nitrogen atmosphere. The resulting mixture was diluted with water (600 mL) and EtOAc (200 mL), and then filtered. The filter cake was washed with EtOAc (1×20 mL). The filtrate was separated, and the aqueous layer was extracted with EtOAc (2×200 mL). The combined organic layers were washed with sat. NH4Cl (aq.) (2×300 mL), brine (1×300 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 80% gradient in 20 min; detector, UV 220 nm. This resulted in methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-cyclobutoxypyridin-3-yl) propanoate (16.4 g, 75%) as a colorless oil. LCMS (ESI+): m/z 385 (M+H+).


Step 3: Methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-cyclobutoxypyridin-3-yl) propanoate



embedded image


To a stirred mixture of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-cyclobutoxypyridin-3-yl) propanoate (16.4 g, 42.61 mmol, 1 eq) and Ag2O (39.50 g, 170.45 mmol, 4.0 eq) in DMF (200 mL) was added CH3I (96.8 g, 681.81 mmol, 16.0 eq) in one portion at rt under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 50° C. under nitrogen atmosphere. The starting material was transformed completely. The resulting mixture was diluted with water (500 mL) and EtOAc (200 mL) and was then filtered. The filter cake was washed with EtOAc (1×20 mL). The resulting mixture was separated, and the aqueous layer was extracted with EtOAc (2×150 mL). The combined organic layers were washed with sat. NH4Cl (aq.) (2×300 mL), brine (1×300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (20:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-cyclobutoxypyridin-3-yl) propanoate (17.0 g, 95%) as a colorless oil.


Step 4: Int. 12



embedded image


To a stirred solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-(5-chloro-2-cyclobutoxypyridin-3-yl) propanoate (17 g, 42.62 mmol, 1 eq) in THF (300 mL) and H2O (100 mL) was added LiOH (3.06 g, 127.86 mmol, 3.00 eq) in H2O (100 mL) dropwise at 0° C. The resulting mixture was stirred for 16 h at rt. The resulting mixture was diluted with water (300 mL) and then acidified to pH 2 with HCl (2 M aq.). The resulting mixture was extracted with EtOAc (3×150 mL). The combined organic layers were washed with brine (1×300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 60% gradient in 25 min; detector, UV 220 nm. This resulted in Int. 12 (15.7 g, 95%) as a white solid. LCMS (ESI+): m/z 385 (M+H+). 1H NMR (300 MHz, DMSO-d6) δ 1.23 (d, J=21.1 Hz, 9H), 1.54-1.88 (m, 2H), 1.93-2.20 (m, 2H), 2.28-2.47 (m, 2H), 2.68 (d, J=1.4 Hz, 3H), 2.86-3.03 (m, 1H), 3.04-3.19 (m, 1H), 4.66-5.03 (m, 1H), 5.05-5.21 (m, 1H), 7.49-7.81 (m, 1H), 7.88-8.23 (m, 1H), 12.95 (s, 1H).


Int. 13: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoic acid



embedded image


Step 1: 5-Chloro-2-methoxy-3-iodopyridine



embedded image


2,5-dichloro-3-iodopyridine (5.0 g, 1 eq, 18 mmol) solution in THF (26 mL) was brought to 0° C. and added NaH (1.5 g, 60% Wt, 2 eq, 37 mmol) as a solid. Then MeOH was added dropwise and stirred for 1 hour at 0° C. Then the mixture was stirred at 50° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 100 mL, then extracted with EtOAc (150 mL×2), the combined organic layers were washed with brine, dried over MgSO4, and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-60%) to yield 5-chloro-2-methoxy-3-iodopyridine (4.3 g, 16 mmol, 87% yield) as yellow solid.


Step 2: Methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoate



embedded image


Zinc (2.4 g, 3 eq, 37 mmol) and DMF (15 mL) were added to a dry flask and degas with Ar. Then Boc-beta-iodo-Ala-OMe (4.0 g, 1 eq, 12 mmol) and iodine (0.47 g, 0.15 eq, 1.8 mmol) were added as solids. In a separate vial, 5-chloro-3-iodo-2-methoxypyridine (4.3 g, 1.3 eq, 16 mmol) and 2-Dicyclohexylphosphino-2,6-Dimethoxy-1,1-Biphenyl (0.13 g, 0.025 eq, 0.31 mmol) were dissolved in DMF (15 mL) and degassed by Ar. Then Pd2dba3 (0.14 g, 0.013 eq, 0.15 mmol) was added. Transfer the first solution into the second solution and stir at rt until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 50 mL, then extracted with EtOAc (100 mL×2), the combined organic layers were washed with brine, dried over MgSO4, and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-60%) to yield methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoate (1.8 g, 8.1 mmol, 66% yield) as white solid. LCMS (ESI+): m/z 345.3 (M+H+)


Step 3: (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoic acid



embedded image


Methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoate (2.8 g, 1 eq, 8.1 mmol) was dissolved into THF (20 mL), H2O (14 mL) and MeOH (6.8 mL), then bring to 0° C. Lithium hydroxide monohydrate (1.0 g, 0.68 mL, 3 eq, 24 mmol) was added and the mixture was stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. Then the 10% citric acid was added to the mixture at 0° C. until pH was acidic. The reaction mixture was concentrated via rotavapor and extracted with EtOAc (80 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography to give (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoic acid (2.3 g, 7.0 mmol, 86% yield). LCMS (ESI+): m/z 331.4 (M+H+)


Step 4: Int. 13



embedded image


(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-methoxypyridin-3-yl)propanoic acid (2.3 g, 1 eq, 7.0 mmol) solid was dissolved in THF (35 mL) and brought to 0° C. under Ar. Then NaH (0.56 g, 60% Wt, 2 eq, 14 mmol) was added as solid, and MeI (5.9 g, 2.6 mL, 6 eq, 42 mmol) was added dropwise. The mixture was stirred for 1 hour at 0° C. Then the mixture was stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was quenched with ice water 100 mL and 10% citric acid was added to the mixture at 0° C. until pH was acidic. The reaction mixture was concentrated via rotavapor and extracted with EtOAc (80 mL×2), the combined organic layers were washed with brine, dried over MgSO4, and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography to give Int. 13 (2.3 g, 6.7 mmol, 96% yield) as white solid. LCMS (ESI+): m/z 345.3 (M+H+).


Int. 14: (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-ethoxypyridin-3-yl)propanoic acid



embedded image


This material was synthesized by following the procedure for Int. 13 with ethanol as the starting alcohol. LCMS (ESI+): m/z 359.3 (M+H+)


Int. 15: (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-isobutoxypyridin-3-yl)propanoic acid



embedded image


This material was synthesized by following the procedure for Int. 13 with 2-methylpropan-1-ol as the starting alcohol. LCMS (ESI+): m/z 387.3 (M+H+)


Int. 16: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl)propanoic acid



embedded image


Step 1: 4-Chloro-N′-cyclopropanecarbonyl-2-iodobenzohydrazide



embedded image


To a stirred solution of 4-chloro-2-iodobenzoic acid (1 g, 3.54 mmol, 1 eq) and NMI (0.87 g, 10.62 mmol, 3 eq) in ACN (40 mL) were added cyclopropanecarbohydrazide (354.45 mg, 3.540 mmol, 1 eq) and TCFH (1.19 g, 4.25 mmol, 1.2 eq) in portions at 0° C. The resulting mixture was stirred for 16 h at rt. The resulting mixture was diluted with water (20 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (1×50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, Acetonitrile in Water (0.1% FA), 0% to 60% gradient in 30 min; detector, UV 254 nm. This resulted in 4-chloro-N′-cyclopropanecarbonyl-2-iodobenzohydrazide (815 mg, 62%) as an off-white solid. LCMS (ESI+): m/z 364.80 (M+H+).


Step 2: 2-(4-chloro-2-iodophenyl)-5-cyclopropyl-1,3,4-oxadiazole



embedded image


A solution of 4-chloro-N′-cyclopropanecarbonyl-2-iodobenzohydrazide (5 g, 13.72 mmol, 1 eq) in POCl3 (100 mL) was stirred for 2 h at 85° C. The resulting mixture was concentrated under vacuum and extracted with CH2Cl2 (3×200 mL). The combined organic layers were washed with NaHCO3 (aq.) (1×200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum to afford 2-(4-chloro-2-iodophenyl)-5-cyclopropyl-1,3,4-oxadiazole (4.5 g, 82%) as a brown solid. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 347.00 (M+H+).


Step 3: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate



embedded image


To a stirred solution of Zn (4.98 g, 76.18 mmol, 2.4 eq), 1,2-Dibromoethane (609.07 mg, 3.17 mmol, 0.1 eq) in DMA (20 mL) was added TMSCl (231.04 mg, 2.13 mmol, 0.067 eq) dropwise at 25° C. under nitrogen atmosphere. The resulting mixture was stirred for 30 min and methyl (2R)-2-[(tert-butoxycarbonyl)amino]-3-iodopropanoate (20.89 g, 63.48 mmol, 2.00 eq) in DMA (20 mL) was added to the above solution. The temperature risen and kept it under 50° C. After 30 min, the mixture was added to a solution of 2-(4-chloro-2-iodophenyl)-5-cyclopropyl-1,3,4-oxadiazole (11 g, 31.74 mmol, 1 eq), CuI (1.21 g, 6.35 mmol, 0.2 eq) and Pd(dppf)Cl2 (2.32 g, 3.174 mmol, 0.1 equiv.) in DMA (200 mL) dropwise and stirred for 2 h at 80° C. The reaction was quenched with sat. NH4Cl (aq.) at 0° C. The resulting mixture was filtered, the filter cake was washed with CH2Cl2 (500 mL) and the filtrate was extracted with CH2Cl2 (1000 mL). The combined organic layers were washed with brine (3×200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, Acetonitrile in Water (0.1% FA), 0% to 100% gradient in 40 min; detector, UV 254 nm. This resulted in methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (7.2 g, 197%) as a brown solid. LCMS (ESI+): m/z 422.20 (M+H+).


Step 4: Methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate



embedded image


To a stirred solution of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (7 g, 16.59 mmol, 1 eq) and Ag2O (15.38 g, 66.37 mmol, 4 eq) in DMF (140 mL) was added CH3I (23.55 g, 165.9 mmol, 10 eq) dropwise at 0° C. The resulting mixture was stirred for 72 h at rt. The resulting mixture was filtered, the filter cake was washed with EtOAc (3×100 mL) and the filtrate was extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (3×200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The crude product (7 g) was purified by Prep-HPLC with the following conditions (Column: XBridge C18 OBD Prep Column, 100 Å 10 μm, 19 mm×250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 60% B to 60% B in 24 min; Wave Length: 220 nm; RT1 (min): 18; Number Of Runs: 0) to afford methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (3.6 g, 50%) as a brown oil. LCMS (ESI+): m/z 565.60 (M+H+).


Step 5: Int. 16



embedded image


To a stirred solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(5-cyclopropyl-1,3,4-oxadiazol-2-yl)phenyl]propanoate (2.6 g, 5.97 mmol, 1 eq) in THF (24 mL)/H2O (8 mL) was added NaOH (715.69 mg, 17.90 mmol, 3.00 eq) in water (10 mL) dropwise at 0° C. The mixture was acidified to pH=5 with HCl (1N). The resulting mixture was extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (1×50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, Acetonitrile in Water (0.1% FA), 5% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in Int. 16 (2.73 g, 108%) as an off-white solid. LCMS (ESI+): m/z 421.95 (M+H+).


Int. 17: Preparation of (S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)(methyl)amino)propanoic acid



embedded image


Step 1: 1-(4-(4-chloro-2-iodophenoxy)piperidin-1-yl)ethan-1-one



embedded image


To a solution of 4-chloro-2-iodophenol (20 g, 78.60 mmol, 1 eq), 1-(4-hydroxy-1-piperidyl)ethanone (12.38 g, 86.46 mmol, 1.1 eq) and PPh3 (24.74 g, 94.32 mmol, 1.2 eq) in THF (200 mL) was added DIAD (19.07 g, 94.32 mmol, 18.34 mL, 1.2 eq) dropwise at 0° C. under N2 atmosphere. Then the reaction mixture was allowed to warm to 20° C. slowly and stirred at 20° C. for 12 h. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was quenched by water (200 mL), extracted with EtOAc (150 mL×2). The combined organics were washed with brine (150 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 330 g SepaFlash® Silica Flash Column, Eluent of 0-55% EtOAc/PE gradient@200 mL/min) to give 1-(4-(4-chloro-2-iodophenoxy)piperidin-1-yl)ethan-1-one (21.49 g, 39.55 mmol, 50% yield, 70% purity) as a light-yellow solid.


Step 2: methyl(S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoate



embedded image


A mixture of Zn (6.95 g, 106.34 mmol, 3.5 eq) in DMF (50 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 120° C. for 10 min under N2 atmosphere. Then I2 (2.31 g, 9.11 mmol, 1.84 mL, 0.3 eq) in DMF (10 mL) was added dropwise at 20° C., methyl (R)-2-((tert-butoxycarbonyl)amino)-3-iodopropanoate (10 g, 30.38 mmol, 1 eq) in DMF (30 mL) was added dropwise, I2 (2.31 g, 9.11 mmol, 1.84 mL, 0.3 eq) in DMF (10 mL) was added dropwise, the reaction mixture was stirred at 20° C. for 1 hr. TLC (PE:EtOAc=5:1, Rf=0.5) showed starting material was consumed completely and a new major spot was detected. The (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (11.99 g, crude) as a gray liquid was obtained, which was used into the next step without further purification.


To a solution of 1-(4-(4-chloro-2-iodophenoxy)piperidin-1-yl)ethan-1-one (5.58 g, 14.70 mmol, 1 eq), sPhos (603.43 mg, 1.47 mmol, 0.1 eq) and Pd2(dba)3 (673.00 mg, 734.95 umol, 0.05 eq) in DMF (50 mL) was degassed and purged with N2 3 times. Then (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (11.99 g, 30.39 mmol, 2.07 eq) in DMF was added dropwise. Then, the reaction mixture was stirred at 65° C. for 12 h. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was cooled to rt and quenched by water (150 mL), extracted with EtOAc (100 mL×2). The combined organics were washed with brine (150 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 800 g Agela C18; mobile phase: [water-ACN]; B %: 20-50% 30 min; 50% 20 min) to give methyl(S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoate (3.62 g, 7.66 mmol, 52% yield, 96% purity) as a white solid.


Step 3: (S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoic acid



embedded image


To a solution of methyl(S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoate (3.62 g, 7.96 mmol, 1 eq) in THF (40 mL) was added a solution of LiOH·H2O (400.69 mg, 9.55 mmol, 1.2 eq) in H2O (20 mL). Then the reaction mixture was stirred at 25° C. for 1 hr. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was concentrated under reduced pressure to remove THF. Then the mixture was acidified by 1N HCl to pH=2-3 at 0° C. and extracted with EtOAc (15 mL×2). The combined organics were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give compound (S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoic acid (3.35 g, 7.60 mmol, 95% yield, 100% purity) as a white solid.


Step 4: Int. 17



embedded image


To a solution of (S)-3-(2-((1-acetylpiperidin-4-yl)oxy)-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoic acid (3.35 g, 7.60 mmol, 1 eq) in THF (40 mL) was added NaH (759.71 mg, 18.99 mmol, 60% purity, 2.5 eq) at 0° C. under N2 atmosphere. The mixture was stirred at 20° C. for 30 min. Then MeI (14.02 g, 98.77 mmol, 6.15 mL, 13 eq) was added at 0° C. and the reaction mixture was stirred at 20° C. for 12 hr. LCMS showed 38% of starting material remained and 26% of desired mass was detected. The reaction mixture was cooled to 0° C. and quenched by NH4Cl (40 mL), extracted with EtOAc (30 mL×2). The combined organics were washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: 800 g Agela C18; mobile phase: [water (FA)-ACN]; B %: 25-55% 30 min; 55% 20 min) to Int. 17 (3.02 g, 6.41 mmol, 97% purity) was obtained as a light-yellow solid. Spectrum: 1H NMR (400 MHz, DMSO-d6) δ=12.80 (br s, 1H), 7.28-6.99 (m, 3H), 4.90-4.60 (m, 2H), 3.73-3.54 (m, 2H), 3.39 (td, J=3.8, 13.3 Hz, 2H), 3.18 (br dd, J=3.8, 13.8 Hz, 1H), 2.95-2.82 (m, 1H), 2.66 (s, 3H), 2.01 (s, 3H), 1.97-1.80 (m, 2H), 1.74-1.50 (m, 2H), 1.30-1.15 (m, 9H)


Int. 18: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid



embedded image


Step 1: 5-Fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine



embedded image


To a solution of 2-bromo-5-fluoropyridin-3-amine (12 g, 62.83 mmol, 1 eq) in dioxane (96 mL) and H2O (18 mL) was added, 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole, Pd(dppf)Cl2 (2.30 g, 3.14 mmol, 0.05 eq) and K2CO3 (17.37 g, 125.65 mmol, 2 eq), the mixture was stirred at 90° C. for 12 h under N2. The reaction was monitored by LCMS, which showed 2-bromo-5-fluoropyridin-3-amine was consumed and one of peak with desired mass was detected. Five other reactions of the same size were set up and all six reactions were combined to work up. After cooling to rt, the mixture was filtered. The filtrate was diluted with water (500 mL), extracted with EtOAc (200 mL×3). The combined organics were washed with brine (300 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (200 mL), and 250 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 400 g Agela flash silica gel column, eluted with 0% to 50% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and concentrated to give 5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine (63 g, crude) as a yellow solid.


Step 2: 5-Fluoro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine



embedded image


To a solution of 5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine (15.75 g, 81.95 mmol, 1 eq) in MeCN (450 mL) was added CuI (18.73 g, 98.34 mmol, 1.2 eq), KI (68.02 g, 409.75 mmol, 5 eq) and tert-butyl nitrite (42.25 g, 409.75 mmol, 48.73 mL, 5 eq), the mixture was stirred at 60° C. for 12 h under N2. The reaction was monitored by LCMS, which showed 5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine as consumed and one of peak with desired mass was detected. Three other reactions of the same size were set up and all four reactions were combined and filtered, and the filter cake was rinsed with EtOAc (100 mL×3). The combined filtrate were concentrated under reduced pressure to give a residue. The filtrate was diluted with water (200 mL), extracted with dichloromethane (80 mL×3). The combined organics were washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (100 mL), and 200 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 300 g Agela flash silica gel column, eluted with 0% to 20% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and concentrated to give 5-fluoro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine (30 g, 98.99 mmol, 60% yield, 100% purity) as a yellow solid.


Step 3: Methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate



embedded image


To a solution of 5-fluoro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine (30 g, 98.99 mmol, 1 eq), SPhos (4.06 g, 9.90 mmol, 0.1 eq) and Pd2(dba)3 (4.83 g, 5.28 mmol, 0.05 eq) in DMF (330 mL) was degassed and purged with N2 3 times. Then (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (78.10 g, 197.97 mmol, 2 eq) in DMF (200 mL) was added dropwise. Then the reaction mixture was stirred at 20° C. for 12 h. The reaction was monitored by LCMS, which showed 5-fluoro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine was consumed and one of peak with desired mass was detected. The mixture was filtered. The filtrate was diluted with water (200 mL), extracted with EtOAc (100 mL×3). The combined organics were washed with brine (200 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Welch Xtimate C18 250×100 mm #10 um; mobile phase: [water (NH4HCO3)-ACN]; B %: 30%-60%, 36 min) the HPLC fractions were combined, lyophilized. The product fraction was combined and concentrated to give methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate (4.2 g, 11.10 mmol, 11% yield, 100% purity) as a yellow solid.


Step 4: (S)-2-((tert-butoxycarbonyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid



embedded image


To a solution of methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate (4.2 g, 11.10 mmol, 1 eq) in THF (60 mL) was added a solution of LiOH·H2O (558.93 mg, 13.32 mmol, 1.2 eq) in H2O (30 mL), the mixture was stirred at 20° C. for 1 h. The mixture was acidified by adding hydrochloric acid (0.1 M, 10 mL) dropwise at 20° C. to pH=5. The filtrate was diluted with water (30 mL), extracted with EtOAc (20 mL×3). The combined organics were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (S)-2-((tert-butoxycarbonyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid (3.7 g, 10.15 mmol, 91% yield, 100% purity) as a yellow solid.


Step 5: Int. 18



embedded image


To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid (1.85 g, 5.08 mmol, 1 eq) in THF (76 mL) was added NaH (1.02 g, 25.39 mmol, 60% purity, 5 eq) at 0° C., the mixture was stirred at 0° C. for 1 h under N2. MeI (7.21 g, 50.77 mmol, 3.16 mL, 10 eq) was added to the reaction mixture, the mixture was stirred at 20° C. for 12 h under N2. LCMS showed the (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-fluoro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid was consumed and desired mass was detected. One other reaction of the same size was set up and all two reactions were combined to work up. The reaction was quenched by addition of saturated ammonium chloride aqueous solution (20 mL), acidified by adding hydrochloric acid (0.1 M, 10 mL) dropwise at 0° C. to pH=5, extracted with EtOAc (20 mL×3). The combined organics were washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give Int. 18 (2.5 g, 6.53 mmol, 64.3% yield, 99% purity) as a yellow solid. 1H NMR (400 MHz, MeOD −d4) δ=8.34 (dd, J=2.4, 15.3 Hz, 1H), 8.02 (d, J=7.0 Hz, 1H), 7.84 (d, J=3.1 Hz, 1H), 7.52 (dt, J=2.6, 9.2 Hz, 1H), 4.74-4.62 (m, 1H), 4.02-3.92 (m, 3H), 3.64-3.54 (m, 1H), 3.28-3.12 (m, 1H), 2.66 (d, J=6.9 Hz, 3H), 1.42-1.18 (m, 9H).


Int. 19: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid



embedded image


Step 1: 5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine



embedded image


To a solution of 2-bromo-5-chloropyridin-3-amine (30 g, 144.61 mmol, 1 eq) and 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (36.11 g, 173.53 mmol, 1.2 eq) in dioxane (720 mL) and H2O (120 mL) was added K2CO3 (39.97 g, 289.22 mmol, 2 eq) and Pd(dppf)Cl2 (3.17 g, 4.34 mmol, 0.03 eq), the mixture was stirred at 90° C. for 12 h under N2. The reaction was monitored by LCMS, which showed 2-bromo-5-chloropyridin-3-amine was consumed and one of peak with desired mass was detected. The reaction mixture was filtered, and the filter cake was washed with EtOAc (50 mL×3) and diluted with water (10 mL), extracted with EtOAc (200 mL×3). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (80 mL), and 40 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 180 g Agela flash silica gel column, eluted with 0% to 40% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and concentrated to give 5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine (30 g, 142.35 mmol, 98% yield, 99% purity) as a brown solid.


Step 2: 5-chloro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine



embedded image


To a solution of 5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine (9 g, 43.14 mmol, 1 eq) in MeCN (300 mL) was added CuI (9.86 g, 51.76 mmol, 1.2 eq), KI (35.80 g, 215.68 mmol, 5 eq) and tert-butyl nitrite (22.24 g, 215.68 mmol, 25.65 mL, 5 eq), the mixture was stirred at 60° C. for 12 h under N2. The reaction was monitored by LCMS, which showed 5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-amine was consumed and one of peak with desired mass was detected. After cooling to rt, the solid was collected by filtration, washed with EtOAc (100 mL×3) and dried under reduced pressure to give a brown solid. The solid was triturated with EtOAc at 20° C. for 30 min. Then the crude product was triturated with ammonia water at 20° C. for 30 min and filtered, the filter cake was concentrated to give 5-chloro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine (20.5 g, 50.04 mmol, 58% yield, 78% purity) brown solid.


Step 3: Methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate



embedded image


To a solution of 5-chloro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine (20.5 g, 64.16 mmol, 1 eq) in DMF (200 mL) was added (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (50.62 g, 128.31 mmol, 2 eq), Pd2(dba)3 (2.94 g, 3.21 mmol, 0.05 eq) and sPhos (2.63 g, 6.42 mmol, 0.1 eq), the mixture was stirred at 20° C. for 12 h under N2. The reaction was monitored by LCMS, which showed 5-chloro-3-iodo-2-(1-methyl-1H-pyrazol-4-yl)pyridine was consumed and one of peak with desired mass was detected. The reaction was quenched by addition of saturated ammonium chloride aqueous solution (200 mL) and extracted with EtOAc (100 mL×3). The combined organics were washed with brine (100 mL×2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (50 mL), and 40 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 120 g Agela flash silica gel column, eluted with 0% to 55% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and concentrated to give methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate (8 g, 19.86 mmol, 31% yield, 98% purity) as a yellow solid.


Step 4: (S)-2-((tertbutoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid



embedded image


To a solution of methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate (8 g, 20.26 mmol, 1 eq) in THF (100 mL) was added a solution of LiOH·H2O (1.02 g, 24.31 mmol, 1.2 eq) in H2O (50 mL), the mixture was stirred at 20° C. for 1 h. The reaction was monitored by LCMS, which showed methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoate was consumed and one of peak with desired mass was detected. The mixture was extracted with EtOAc (100 mL). The aqueous phase was acidified by added hydrochloric acid (1 M, 50 mL) dropwise at 0° C. to pH=4 and extracted with EtOAc (200 mL×3). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated to give (S)-2-((tertbutoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid (7 g, 18.01 mmol, 89% yield, 98% purity) as a yellow solid.


Step 5: Int. 19



embedded image


To a solution of (S)-2-((tertbutoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid (3.7 g, 9.72 mmol, 1 eq) in THF (100 mL) was added NaH (1.94 g, 48.58 mmol, 60% purity, 5 eq) at 0° C., the mixture was stirred at 0° C. for 1 h under N2. MeI (13.79 g, 97.16 mmol, 6.05 mL, 10 eq) was added to the reaction mixture, the mixture was stirred at 20° C. for 12 h under N2. The reaction was monitored by LCMS, which showed (S)-2-((tertbutoxycarbonyl)amino)-3-(5-chloro-2-(1-methyl-1H-pyrazol-4-yl)pyridine-3-yl)propanoic acid was consumed and one of peak with desired mass was detected. The reaction mixture was cooled to 0° C. and quenched by NH4Cl (30 mL), extracted with EtOAc (50 mL×2). The combined organics were washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give Int. 19 (3 g, 7.51 mmol, 77% yield, 98% purity) as a yellow solid. 1H NMR (400 MHz, MeOD-d4) 1H NMR (400 MHz, METHANOL-d4) δ=8.48-8.32 (m, 1H), 8.08 (d, J=6.8 Hz, 1H), 7.92 (s, 1H), 7.76-7.64 (m, 1H), 4.80-4.66 (m, 1H), 3.98 (s, 3H), 3.64-3.56 (m, 1H), 3.28-3.16 (m, 1H), 2.66 (d, J=9.8 Hz, 3H), 1.24 (s, 9H)


Int. 20: Preparation of (S)-2-((tertbutoxycarbonyl)(methyl)amino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl)propanoic acid



embedded image


Step 1: 4-chloro-1-(1-methylcyclopropoxy)-2-nitrobenzene



embedded image


To a solution of 4-chloro-1-fluoro-2-nitrobenzene (21 g, 119.63 mmol, 1 eq) and 1-methylcyclopropanol (11.16 g, 131.59 mmol, 85% purity, 1.1 eq) in DMF (150 mL) was added Cs2CO3 (58.47 g, 179.44 mmol, 1.5 eq). Then, the reaction mixture was stirred at 75° C. for 3 h. HPLC showed starting material was consumed completely and a new major peak was detected. The reaction mixture was cooled to rt and quenched by water (200 mL), extracted with EtOAc (150 mL×2). The combined organics were washed with water (100 mL×3) and brine (150 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 4-chloro-1-(1-methylcyclopropoxy)-2-nitrobenzene (25 g, 104.54 mmol, 87% yield, 95.2% purity) as a brown oil.


Step 2: 5-chloro-2-(1-methylcyclopropoxy)aniline



embedded image


To a solution of 4-chloro-1-(1-methylcyclopropoxy)-2-nitrobenzene (25 g, 109.82 mmol, 1 eq) in EtOH (200 mL) and H2O (100 mL) was added NH4Cl (5.87 g, 109.82 mmol, 1 eq) and Fe (18.40 g, 329.46 mmol, 3 eq). Then, the reaction mixture was stirred at 80° C. for 2 h. LCMS showed starting material was consumed completely and desired mass was detected. The resultant mixture was filtered, and the filter cake was rinsed with EtOH (100 mL×3). Then the combined filtrates were concentrated under reduced pressure to remove EtOH. Then the aqueous phase was extracted with EtOAc (150 mL×2). The combined organics were washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 5-chloro-2-(1-methylcyclopropoxy)aniline (20.9 g, 104.54 mmol, 95% yield, 99% purity) as a black, brown oil.


Step 3: 4-chloro-2-iodo-1-(1-methylcyclopropoxy)benzene



embedded image


To a solution of 5-chloro-2-(1-methylcyclopropoxy)aniline (20.9 g, 105.74 mmol, 1 eq) in HCl (2.5 M, 209.00 mL, 4.94 eq) and ACN (100 mL) was added a solution of NaNO2 (8.02 g, 116.31 mmol, 1.1 eq) in H2O (100 mL) slowly at 0° C. Then, a solution of KI (43.88 g, 264.34 mmol, 2.5 eq) in H2O (100 mL) was added slowly. Then the reaction mixture was allowed to warm to 25° C. and stirred at 25° C. for 12 h. LCMS showed starting material was consumed completely and a new major spot was detected. The reaction mixture was adjusted pH=7 by adding saturated NaHCO3 aqueous solution slowly at 0° C., extracted with EtOAc (60 mL×2). The combined organics were washed with brine (80 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 220 g SepaFlash® Silica Flash Column, Eluent of 0˜0% EtOAc/PE gradient @ 150 mL/min) to give 4-chloro-2-iodo-1-(1-methylcyclopropoxy)benzene (23 g, 73.84 mmol, 70% yield, 99.1% purity) as a light-yellow oil.


Step 4: Methyl(S)-2-((tert-butoxycarbonylamino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl)propanoate



embedded image


A mixture of 4-chloro-2-iodo-1-(1-methylcyclopropoxy)benzene (15 g, 48.62 mmol, 1 eq), sPhos (2.00 g, 4.86 mmol, 0.1 eq) and Pd2(dba)3 (2.23 g, 2.43 mmol, 0.05 eq) in DMF (50 mL) was degassed and purged with N2 3 times, then [(2R)-2-(tert-butoxycarbonylamino)-3-methoxy-3-oxo-propyl]-iodo-zinc (41.95 g, 106.34 mmol, 2.19 eq) was added to the mixture and then the mixture was stirred at 65° C. for 12 h under N2 atmosphere. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was cooled to rt and quenched by water (300 mL), extracted with EtOAc (150 mL×2). The combined organics were washed with water (200 mL×2) and brine (200 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 250*70 mm #10 um; mobile phase: [water (NH4HCO3)-ACN]; B %: 50%-80%, 20 min) to give methyl(S)-2-((tert-butoxycarbonylamino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl)propanoate (15 g, 36.75 mmol, 76% yield, 94.0% purity) as a light-yellow solid.


Step 5: (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl) propanoic acid



embedded image


To a solution of methyl(S)-2-((tert-butoxycarbonylamino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl)propanoate (15 g, 39.08 mmol, 1 eq) in THF (200 mL) was added a solution of LiOH·H2O (2.46 g, 58.61 mmol, 1.5 eq) in H2O (100 mL) at 0-10° C. Then the reaction mixture was stirred at 20° C. for 0.5 hr. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was concentrated under reduced pressure to remove THF and extracted with EtOAc (80 mL×2). The aqueous phase was acidified by adding hydrochloric acid (1N) dropwise at 0° C. to pH=2-3. The aqueous phase was extracted with EtOAc (100 mL×2). The combined organics were washed with brine (150 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl) propanoic acid (12.7 g, 33.65 mmol, 86% yield, 98.1% purity) as a light-yellow oil.


Step 6: Int. 20



embedded image


To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-(1-methylcyclopropoxy)phenyl) propanoic acid (12.7 g, 34.34 mmol, 1 eq) in THF (400 mL) was added NaH (6.87 g, 171.70 mmol, 60% purity, 5 eq) at 0° C. under N2 atmosphere. The mixture was stirred at 20° C. for 0.5 hr. Then MeI (48.74 g, 343.39 mmol, 21.38 mL, 10 eq) was added and the reaction mixture was stirred at 20° C. for 12 h. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was cooled to 0° C. and quenched by NH4Cl (150 mL), acidified by adding hydrochloric acid (1 N) dropwise at 0° C. to pH=2-3 and extracted with EtOAc (200 mL×2). The combined organics were washed with brine (200 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give Int. 20 (9.98 g, 26.00 mmol, 76% yield, 100% purity) as a light-yellow solid. 1H NMR (400 MHz, DMSO-d6) δ=7.28-7.04 (m, 3H), 4.83-4.39 (m, 1H), 3.15-2.97 (m, 1H), 2.89-2.71 (m, 1H), 2.61 (br d, J=3.3 Hz, 3H), 1.49 (d, J=11.1 Hz, 3H), 1.30-1.12 (m, 9H), 0.95-0.81 (m, 2H), 0.81-0.69 (m, 2H)


Int. 22: Preparation of (S)-3-(1′-acetyl-5-chloro-1′,2′,3′,6′-tetrahydro-[2,4′-bipyridin]-3-yl)-2-((tert-butoxycarbonyl)(methyl)amino)propanoic acid



embedded image


Step 1: 2-bromo-3-(bromomethyl)-5-chloropyridine



embedded image


To a stirred mixture of 2-bromo-5-chloro-3-methylpyridine (25 g, 121.08 mmol, 1 eq.) and NBS (23706.05 mg, 133.19 mmol, 1.1 eq) in ACN (200 mL) was added AIBN (1988.33 mg, 12.11 mmol, 0.1 eq.) at rt under nitrogen atmosphere. The resulting mixture was stirred overnight at 80° C. The resulting mixture was concentrated in vacuo and diluted with water. The mixture was extracted with EtOAc (2×200 mL) and the combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. This resulted in 2-bromo-3-(bromomethyl)-5-chloropyridine (30 g, 87%) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 282.4.


Step 2: Tert-butyl (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(diphenylmethylidene)amino]propanoate



embedded image


To a stirred mixture of 2-bromo-3-(bromomethyl)-5-chloropyridine (15 g, 52.57 mmol, 1 eq.), (2S,4S,5R)-1-[(anthracen-9-yl)methyl]-5-ethenyl-2-[(R)-(prop-2-en-1-yloxy)(quinolin-4-yl)methyl]-1-azabicyclo[2.2.2]octan-1-ium bromide (1591.69 mg, 2.63 mmol, 0.05 eq.) and isopropyl 2-[(diphenylmethylidene)amino]acetate (29578.95 mg, 105.13 mmol, 2 eq.) in DCM (100 mL) was added KOH (29.49 g, 525.65 mmol, 10 eq.) in H2O (30 mL) in portions at −10° C. under air atmosphere. The resulting mixture was stirred for an additional 4 h at −10° C. The resulting mixture was extracted with EtOAc (2×200 mL), and the combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (10:1) to afford tert-butyl (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(diphenylmethylidene)amino]propanoate (25 g, 95%) as a yellow oil. LCMS (ESI+): m/z 501.1.


Step 3: (S)-2-amino-3-(2-bromo-5-chloropyridin-3-yl)propanoic acid



embedded image


A mixture of tert-butyl (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(diphenylmethylidene)amino]propanoate (5 g, 10.0 mmol, 1 eq.) and HCl (6M) (30 mL) in THF (10 mL) was stirred overnight at 55° C. The resulting mixture was concentrated under vacuum. This resulted in (2S)-2-amino-3-(2-bromo-5-chloropyridin-3-yl)propanoic acid (2.6 g, 93%) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 278.9.


Step 4: (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(tert-butoxycarbonyl)amino]propanoic acid



embedded image


To a stirred mixture of (2S)-2-amino-3-(2-bromo-5-chloropyridin-3-yl)propanoic acid (9 g, 32.20 mmol, 1 eq.) and Na2CO3 (10.24 g, 96.59 mmol, 3 eq.) in H2O (60 mL) was added Boc2O (14.05 g, 64.40 mmol, 2 eq.) in portions at rt under air atmosphere. The resulting mixture was stirred overnight at rt. The mixture was acidified to pH=6 with diluted HCl (aq. 1N). The resulting mixture was extracted with EtOAc (2×100 mL), and the combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 30 min; detector, UV 210 nm. This resulted in (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(tert-butoxycarbonyl)amino]propanoic acid (12 g, 98%) as a white solid. LCMS (ESI+): m/z 380.90.


Step 5: (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(tert-butoxycarbonyl)(methyl)amino]propanoic acid



embedded image


NaH (6.32 g, 263.41 mmol, 10 eq.) was added into a stirred mixture of (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(tert-butoxycarbonyl)amino]propanoic acid (10 g, 26.34 mmol, 1 eq.) in THF (100 mL) portion wise at 0° C. under nitrogen and the resulting mixture was stirred for 1 h at 0° C. MeI (56.08 g, 395.12 mmol, 15 eq.) was added dropwise at 0° C. under nitrogen atmosphere. The resulting mixture was stirred overnight at rt. The reaction was quenched with water at 0° C. The mixture was acidified to pH=6 with HCl (aq.). The resulting mixture was extracted with EtOAc (2×120 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 30 min; detector, UV 220 nm. This resulted in (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(tert-butoxycarbonyl)(methyl)amino]propanoic acid (7.3 g, 70%) as a yellow oil.


Step 6: Int. 22



embedded image


To a stirred mixture of (2S)-3-(2-bromo-5-chloropyridin-3-yl)-2-[(tert-butoxycarbonyl)(methyl)amino]propanoic acid (2.5 g, 6.35 mmol, 1 eq.) and 1-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2H-pyridin-1-yl]ethanone (2392.26 mg, 9.53 mmol, 1.5 eq.) in dioxane (20 mL) was added Na2CO3 (2019.28 mg, 19.05 mmol, 3 eq.) and Pd(dppf)Cl2·DCM Adduct (517.34 mg, 0.635 mmol, 0.1 eq) in portions at rt under nitrogen atmosphere. The resulting mixture was stirred for an additional 3 h at 80° C. The mixture was acidified to pH=6 with HCl (aq. 1N). The resulting mixture was extracted with EtOAc (2×60 mL), and the combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 30 min; detector, UV 254 nm. This resulted in Int. 22 (1.94 g, 66%) as a brown solid. LCMS (ESI+): m/z 438.2. 1H NMR (400 MHz, DMSO-d6) δ 12.99 (s, 1H), 8.45 (dd, J=15.5, 2.4 Hz, 1H), 7.81 (dd, J=53.7, 2.4 Hz, 1H), 5.86 (d, J=33.1 Hz, 1H), 4.85-4.50 (m, 1H), 4.09 (tt, J=30.1, 13.7 Hz, 2H), 3.90-3.47 (m, 2H), 3.42-3.24 (m, 2H), 3.11 (dt, J=26.2, 13.1 Hz, 1H), 2.58-2.37 (m, 4H), 2.41-2.15 (m, 1H), 2.05 (dd, J=12.3, 2.5 Hz, 3H), 1.23 (dd, J=31.0, 3.0 Hz, 9H).


Int. 23: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoic acid



embedded image


Step 1: 4-chloro-1-cyclopropoxy-2-nitrobenzene



embedded image


To a solution of 4-chloro-1-fluoro-2-nitrobenzene (40 g, 227.86 mmol, 1 eq) in DMF (400 mL) was added cyclopropanol (19.85 g, 341.79 mmol, 1.5 eq) and Cs2CO3 (111.36 g, 341.79 mmol, 1.5 eq), the mixture was stirred at 75° C. for 12 h under N2. The reaction was monitored by LCMS, which showed 4-chloro-1-fluoro-2-nitrobenzene was consumed and one of peak with desired mass was detected. After cooling to rt, the filtrate was diluted with water (400 mL), extracted with EtOAc (200 mL×3). The combined organics were washed with brine (400 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. 4-chloro-1-cyclopropoxy-2-nitrobenzene (35 g, crude) was obtained as a black, brown oil.


Step 2: 5-chloro-2-cyclopropoxyaniline



embedded image


To a solution of 4-chloro-1-cyclopropoxy-2-nitrobenzene (35 g, 163.84 mmol, 1 eq) in H2O (280 mL) and ethanol (280 mL) was added Fe (45.75 g, 819.22 mmol, 5 eq) and NH4Cl (4.38 g, 81.92 mmol, 0.5 eq), the mixture was stirred at 80° C. for 1 h. The reaction was monitored by LCMS, which showed 4-chloro-1-cyclopropoxy-2-nitrobenzene was consumed and one of peak with desired mass was detected. After cooling to rt, the reaction mixture was filtered, and the filter cake was rinsed with EtOAc (100 mL×3). Then the combined filtrates were concentrated under reduced pressure to give a residue, 5-chloro-2-cyclopropoxyaniline (26 g, 115.93 mmol, 71% yield, 82% purity) was obtained as a yellow oil.


Step 3: 4-chloro-1-cyclopropoxy-2-iodobenzene



embedded image


To a solution of 5-chloro-2-cyclopropoxyaniline (20 g, 108.91 mmol, 1 eq) in H2O (216 mL) was added hydrogen chloride (12 M, 27.23 mL, 3 eq) dropwise at 0° C. adjusted pH to 2, the mixture was stirred at 0° C. for 0.5 hr, a solution of NaNO2 (8.27 g, 119.80 mmol, 1.1 eq) in H2O (36 mL) was added to the mixture dropwise at 0° C. and stirred for 1 hr. Then the reaction mixture was added to a solution of KI (54.24 g, 326.74 mmol, 3 eq) in H2O (200 mL), the mixture was stirred at 20° C. for 12 hr. The reaction was monitored by LCMS, which showed 5-chloro-2-cyclopropoxyaniline was consumed and one of peak with desired mass was detected. The mixture was filtered, and filtrate was diluted with water (300 mL), extracted with EtOAc (150 mL×3). The combined organics were washed with brine (150 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give residue. The residue was dissolved in DCM (100 mL), and 60 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 120 g agela flash silica gel column, eluted with 0% to 12% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and evaporated. 4-chloro-1-cyclopropoxy-2-iodobenzene (20 g, 65.46 mmol, 60% yield, 96.4% purity) was obtained as a black, brown oil.


Step 4: Methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoate



embedded image


A mixture of Zn (34.77 g, 531.70 mmol, 3.5 eq) in DMF (210 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 120° C. for 10 min under N2 atmosphere. Then 12 (11.57 g, 45.57 mmol, 9.18 mL, 0.3 eq) in DMF (120 mL) was added dropwise at 20° C., methyl (R)-2-((tert-butoxycarbonyl)amino)-3-iodopropanoate (50 g, 151.92 mmol, 1 eq) in DMF (40 mL) was added dropwise, I2 (11.57 g, 45.57 mmol, 9.18 mL, 0.3 eq) in DMF (40 mL) was added dropwise, the reaction mixture was stirred at 20° C. for 1 hr. The reaction was monitored by TLC (PE:EtOAc=5:1, Rf=0.5) showed starting material was consumed completely and a new major spot was detected. The crude product was used into the next step without further purification. (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (59.93 g, crude) was obtained as a grey liquid. The zincate was used without further purification.


To a solution of 4-chloro-1-cyclopropoxy-2-iodobenzene (20 g, 67.91 mmol, 1 eq), sPhos (4.18 g, 10.19 mmol, 0.15 eq) and Pd2(dba)3 (6.22 g, 6.79 mmol, 0.1 eq) in DMF (400 mL) was degassed and purged with N2 for 3 times. Then (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (53.58 g, 135.82 mmol, 2 eq) in DMF was added dropwise. Then the reaction mixture was stirred at 20° C. for 12 hr. The reaction was monitored by LCMS, which showed 4-chloro-1-cyclopropoxy-2-iodobenzene was consumed and one of peaks with desired mass was detected. The reaction was quenched by addition of saturated ammonium chloride aqueous solution (200 mL) and filtered, the filtrate was diluted with water (100 mL), extracted with EtOAc (100 mL×3). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Waters Xbridge BEH C18 250×70 mm×10 um; mobile phase: [water (NH4HCO3)-ACN]; B %:50%-80%, 18 min) the HPLC fractions were combined, lyophilized. Methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoate (8 g, 19.53 mmol, 29% yield, 98% purity) was obtained as a yellow oil.


Step 5: (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoic acid



embedded image


To a solution of methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoate (8 g, 21.63 mmol, 1 eq) in THF (112 mL) was added LiOH·H2O (1.09 g, 25.96 mmol, 1.2 eq) in H2O (56 mL), the mixture was stirred at 20° C. for 1 hr. The reaction was monitored by LCMS, which showed methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoate was consumed and one of peak with desired mass was detected. The mixture was acidified pH=5 by adding hydrochloric acid (0.1 M, 15 mL) dropwise at 0° C. The residue was diluted with water (100 mL), extracted with EtOAc (40 mL×3). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoic acid (5 g, 13.77 mmol, 64% yield, 98% purity) was obtained as a yellow solid.


Step 6: Int. 23



embedded image


To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoic acid (5 g, 14.05 mmol, 1 eq) in THF (50 mL) was added NaH (2.81 g, 70.26 mmol, 60% purity, 5 eq) at 0° C., the mixture was stirred at 0° C. for 1 h under N2, MeI (19.95 g, 140.52 mmol, 8.75 mL, 10 eq) was added to the reaction mixture, the mixture was stirred at 20° C. for 12 h. The reaction was monitored by LCMS, which showed (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-cyclopropoxyphenyl)propanoic acid was consumed and one of peak with desired mass was detected. The reaction was quenched by addition of saturated ammonium chloride aqueous solution (20 mL), the mixture was acidified pH=5 by adding hydrochloric acid (0.1 M, 10 mL) dropwise at 0° C. The residue was extracted with EtOAc (20 mL×3). The combined organics were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue Int. 23 (3 g, 7.71 mmol, 54% yield, 95% purity) was obtained as a yellow oil. 1H NMR (400 MHz, DMSO-d6) 1H NMR (400 MHz, DMSO-d6) δ=7.31-7.08 (m, 3H), 4.85-4.52 (m, 1H), 3.91-3.80 (m, 1H), 3.12-2.99 (m, 1H), 2.94-2.80 (m, 1H), 2.65-2.55 (m, 3H), 1.24 (br d, J=19.6 Hz, 9H), 0.87-0.58 (m, 4H)


Int. 24: Preparation of (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)phenyl]propanoic acid



embedded image


Step 1: (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)phenyl]propanoate



embedded image


A solution of methyl (2S)-3-(2-bromo-5-chlorophenyl)-2-[(tert-butoxycarbonyl)(methyl)amino]propanoate (3 g, 7.376 mmol, 1 equiv.), Cs2CO3 (7232.46 mg, 22.128 mmol, 3 equiv.), morpholine (771.18 mg, 8.851 mmol, 1.2 equiv.), SPhos (605.66 mg, 1.475 mmol, 0.2 equiv.) and Pd2(dba)3 (675.48 mg, 0.738 mmol, 0.1 equiv.) in dioxane (50 mL) was stirred for 3 h at 80° C. under nitrogen atmosphere. The residual Cs2CO3 was removed by filtration. The residue was purified by silica gel column chromatography, eluted with PE/EA (5:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)phenyl]propanoate (1.15 g, 38%) as a brown oil. LCMS (ESI+): m/z 413 (M+H+).


Step 2: Int. 24



embedded image


To a stirred solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)phenyl]propanoate (1.2 g, 2.906 mmol, 1 equiv.) in tetrahydrofuran (15 mL) was added caustic soda (0.58 g, 14.530 mmol, 5 equiv.) in water (5 mL) dropwise at 0° C. The resulting mixture was stirred for 16 h at rt. The resulting mixture was concentrated under reduced pressure and then diluted with water (20 mL). The resulting mixture was acidified to pH=3 with diluted citric acid (1N) and then extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (1×100 mL) and dried over anhydrous Na2SO4. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 220 nm. After filtration, the filtrate was concentrated under reduced pressure. This resulted in Int. 24 (0.81 g, 70%) as a Brown yellow solid. LCMS (ESI+): m/z 399 (M+H+).


Int. 25: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-morpholinopyridin-3-yl)propanoic acid



embedded image


Step 1: 4-(3-bromo-5-chloropyridin-2-yl)morpholine



embedded image


A solution of 3-bromo-5-chloro-2-fluoropyridine (10 g, 47.52 mmol, 1 eq) in DMF (600 mL) was treated with Cs2CO3 (46.45 g, 142.57 mmol, 3 eq) at rt under nitrogen atmosphere followed by the addition of morpholine (6.21 g, 71.28 mmol, 1.5 eq.) in portions at rt. The resulting mixture was stirred for 16 h at rt under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was diluted with ice water (1 L) and extracted with EtOAc (2×1 L). The combined organic layers were washed with brine (500 mL×3) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 40 min; detector, UV 200.210 nm. This resulted in 4-(3-bromo-5-chloropyridin-2-yl)morpholine (3 g, 23%) as a light-yellow solid. LCMS (ESI+): m/z 279.00.


Step 2: Methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(morpholin-4-yl)pyridin-3-yl]propanoate



embedded image


A solution of 4-(3-bromo-5-chloropyridin-2-yl)morpholine (3.3 g, 11.89 mmol, 1 eq.) in DMA (30 mL) was treated with CuI (0.45 g, 2.38 mmol, 0.2 eq.) and Pd(dppf)Cl2·CH2Cl2 (0.87 g, 1.19 mmol. 0.1 eq.) at rt under nitrogen atmosphere followed by the addition of methyl (2R)-2-[(tert-butoxycarbonyl)amino]-3-(iodozincio)propanoate (33 mL) at rt. The resulting mixture was stirred for 16 h at 80° C. under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water (200 mL) and diluted with EA (200 mL). The precipitated solids were removed by filtration and washed with EtOAc (2×200 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 40 min; detector, UV 200.210 nm. The residue was purified by Prep-TLC (PE/EA 2:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(morpholin-4-yl)pyridin-3-yl]propanoate (1 g, 21%) as a light-yellow oil. LCMS (ESI+): m/z 400.15.


Step 3: Methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)pyridin-3-yl]propanoate



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)amino]-3-[5-chloro-2-(morpholin-4-yl)pyridin-3-yl]propanoate (700 mg, 1.75 mmol, 1 eq.) in DMF (10 mL, 129.22 mmol) was treated with Ag2O (2 g, 8.76 mmol, 5 eq.) at rt under nitrogen atmosphere followed by the addition of CH3I (2.4 g, 17.51 mmol, 10 eq.) at rt. The resulting mixture was stirred for 16 h at rt under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was extracted with EtOAc (2×100 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA 2:1) to afford methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)pyridin-3-yl]propanoate (500 mg, 69%) as a light-yellow oil. LCMS (ESI+): m/z 414.15. 1H NMR (400 MHz, DMSO-d6): δ1.15-1.34 (m, 8H), 2.61 (d, J=8.9 Hz, 2H), 2.94-3.13 (m, 4H), 3.23 (dd, J=14.7, 4.5 Hz, 1H), 3.66-3.75 (m, 5H), 7.69 (dd, J=34.4, 2.6 Hz, 1H), 8.22 (dd, J=13.9, 2.6 Hz, 1H).


Step 4: Int. 25



embedded image


A solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]-3-[5-chloro-2-(morpholin-4-yl)pyridin-3-yl]propanoate (660 mg, 1.60 mmol, 1 eq.) in THF (10 mL) was treated with NaOH (318.89 mg, 7.98 mmol, 5 eq.) in water (10 mL) at 0° C. The resulting mixture was stirred for 2 h at rt. Desired product could be detected by LCMS. The mixture neutralized to pH=2 with diluted HCl (1N). The resulting mixture was extracted with EtOAc (2×50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 40 min; detector, UV 200.210 nm. This resulted in Int. 25 (423 mg, 66%) as a white solid. LCMS (ESI+): m/z 400.15. 1H NMR (400 MHz, DMSO-d6): δ 1.26 (s, 9H), 2.61 (d, J=8.7 Hz, 3H), 3.00 (tq, J=13.5, 8.6, 7.2 Hz, 4H), 3.63-3.78 (m, 4H), 5.03 (ddd, J=148.8, 11.4, 4.2 Hz, 1H), 7.67 (dd, J=35.8, 2.6 Hz, 1H), 8.21 (dd, J=14.5, 2.6 Hz, 1H), 12.91 (s, 1H).


Int. 26: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoic acid



embedded image


Step 1: 5-chloro-2-hydroxy-3-nitrophenyl acetate



embedded image


Add TFAA (409.17 g, 1.95 mol, 270.97 mL, 10 eq.) to a three-necked flask, add H2O2 (110.44 g, 974.07 mmol, 93.59 mL, 30% purity, 5 eq.) dropwise at −10° C. After 10 min, add a solution of 1-(5-chloro-2-hydroxy-3-nitrophenyl)ethan-1-one (42 g, 194.81 mmol, 1 eq) in DCM (250 mL) dropwise. The reaction mixture was stirred at 20° C. for 12 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of peak with desired mass was detected. The reaction mixture was diluted with water (100 mL), extracted with DCM (300 mL×3). The combined organics were washed with sodium sulfite aqueous solution (100 mL, 1M) and brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 5-chloro-2-hydroxy-3-nitrophenyl acetate (30 g, 124.36 mmol, 64% yield, 96% purity) as a yellow solid. LCMS (ESI+): m/z 230.0 (M+H+).


Step 2: 5-chloro-3-nitrobenzene-1,2-diol



embedded image


To a solution of 5-chloro-2-hydroxy-3-nitrophenyl acetate (30 g, 129.54 mmol, 1 eq) in EtOH (300 mL) was added NaOH (12.95 g, 129.54 mmol, 40 mL, 40% purity in H2O, 1 eq), the mixture was stirred at 20° C. for 2 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of peak with desired mass was detected. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was acidified pH=4 by adding hydrochloric acid (1 M, 50 mL) dropwise at 0° C. The mixture was extracted with EtOAc (200 mL×3). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 5-chloro-3-nitrobenzene-1,2-diol (22 g, 113.74 mmol, 87.80% yield, 98% purity) as a yellow solid. LCMS (ESI+): m/z 188.0 (M+H+).


Step 3: 7-chloro-5-nitro-2,3,-dihydrobenzo[b][1,4]dioxine



embedded image


To a solution of 5-chloro-3-nitrobenzene-1,2-diol (10 g, 52.76 mmol, 1 eq) in DMF (100 mL) was added 1,2-dibromoethane (9.91 g, 52.76 mmol, 3.98 mL, 1 eq) and K2CO3 (14.58 g, 105.51 mmol, 2 eq) at 20° C. Then the mixture was stirred at 80° C. for 12 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of main peak was detected. The reaction mixture was cooled to rt and diluted by water (100 mL), extracted with EtOAc (60 mL×3). The combined organics were washed with water (100 mL×2). Then the combined organics were washed with brine (200 mL), dried over Na2SO4, filtered and the reaction mixture was concentrated under reduced pressure to give 7-chloro-5-nitro-2,3,-dihydrobenzo[b][1,4]dioxine (11 g, 51.0 mmol, 97% yield) as a black, brown oil.


Step 4: 7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-amine



embedded image


To a solution of 7-chloro-5-nitro-2,3,-dihydrobenzo[b][1,4]dioxine (16 g, 74.21 mmol, 1 eq) in EtOH (80 mL) and H2O (80 mL) were added Fe (20.72 g, 371.07 mmol, 5 eq) and NH4Cl (19.85 g, 371.07 mmol, 5 eq) at 20° C. The reaction was stirred at 80° C. for 2 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of main peak with desired mass was detected. The reaction mixture was filtered through a pad of celite and the celite was rinsed with EtOAc (100 mL). The filtrate was diluted with water (100 mL), extracted with EtOAc (120 mL×3). The combined organics were washed with brine (200 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (200 mL), and 50 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using an 80 g Agela flash silica gel column, eluted with 30% to 40% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and evaporated to give 7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-amine (8.6 g, 43.84 mmol, 30% yield, 94% purity) as a brown oil. LCMS (ESI+): m/z 186.3 (M+H+).


Step 5: 7-chloro-5-iodo-2,3-dihydrobenzo[b][1,4]dioxine



embedded image


To a solution of 7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-amine (4.6 g, 24.78 mmol, 1 eq) in MeCN (24 mL) was added hydrogen chloride (2.5 M, 49.57 mL, 5 eq) at 0° C. The mixture was stirred at 0° C. for 0.5 h. Then a solution of NaNO2 (1.88 g, 27.26 mmol, 1.1 eq) in H2O (20 mL) was added to the reaction mixture, the mixture was stirred at 0° C. for 0.5 h. Then a solution of KI (12.34 g, 74.35 mmol, 3 eq) in H2O (100 mL) was added to the reaction mixture, the reaction mixture was stirred at 0° C. for 2 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of main peak with desired mass was detected. The mixture was adjusted pH=7-8 by adding saturated NaHCO3 aqueous solution slowly at 0° C. Then the mixture was extracted with EtOAc (50 mL×3). The combined organics were washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (50 mL), and 20 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 40 g Agela flash silica gel column, eluted with 15% to 20% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and evaporated to give 7-chloro-5-iodo-2,3-dihydrobenzo[b][1,4]dioxine (6.5 g, 21.71 mmol, 88% yield, 99% purity) as a black, purple solid. LCMS (ESI+): m/z 298.1 (M+H+).


Step 6: methyl(S)-2-((tertbutoxycarbonyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoate



embedded image


A mixture of Zn (13.91 g, 212.68 mmol, 3.5 eq) in DMF (100 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 120° C. for 10 min under N2 atmosphere. Then I2 (4.63 g, 18.23 mmol, 3.7 mL, 0.3 eq) in DMF (40 mL) was added dropwise at 20° C., methyl (2R)-2-(tert-butoxycarbonylamino)-3-iodo-propanoate (20 g, 60.77 mmol, 1 eq) in DMF (30 mL) was added dropwise, I2 (4.63 g, 18.23 mmol, 3.67 mL, 0.3 eq) in DMF (30 mL) was added dropwise, the reaction mixture was stirred at 20° C. for 1 hr. The zincate was used into the next step without further purification. [(2R)-2-(tert-butoxycarbonylamino)-3-methoxy-3-oxo-propyl]-iodo-zinc (23.95 g, 60.71 mmol, 99.9% yield) was obtained as a gray liquid. To a solution of 7-chloro-5-iodo-2,3-dihydrobenzo[b][1,4]dioxine (9 g, 30.36 mmol, 1 eq) in DMF (90 mL) was added [(2R)-2-(tert-butoxycarbonylamino)-3-methoxy-3-oxo-propyl]-iodo-zinc (23.95 g, 60.71 mmol, 2 eq), Pd2(dba)3 (1.39 g, 1.52 mmol, 0.05 eq) and SPhos (1.25 g, 3.04 mmol, 0.1 eq) at 20° C. Then the mixture was stirred at 20° C. for 12 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of main peak with desired mass was detected. The reaction mixture was diluted by water (90 mL), extracted with EtOAc (100 mL×3). The combined organics were washed with water (150 mL×2). Then the combined organics were washed with brine (200 mL), dried over Na2SO4, filtered and the reaction mixture was concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (30 mL), and 18 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 40 g Agela flash silica gel column, eluted with 0% to 20% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and evaporated to give methyl(S)-2-((tertbutoxycarbonyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoate (6 g, 15.55 mmol, 51% yield, 96% purity) a yellow oil.


Step 7: (S)-2-((tert-butoxycarbonyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoic acid



embedded image


To a solution of methyl(S)-2-((tertbutoxycarbonyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoate (5.5 g, 14.79 mmol, 1 eq) in THF (36 mL) was added a solution of LiOH·H2O (744.89 mg, 17.75 mmol, 1.2 eq) in H2O (18 mL) at 0° C. The mixture was stirred at 20° C. for 2 h. The reaction was monitored by LCMS, which showed starting material was consumed and one of main peak with desired mass was detected. The mixture was acidified by adding hydrochloric acid (1 M) dropwise at 0° C. to pH=3. The mixture was extracted with EtOAc (50 mL×3). The combined organics were washed with brine (80 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was dissolved in DCM (30 mL), and 10 g of silica gel was added. The resulting mixture was evaporated under reduced pressure to give a dry flowing solid, and then it was loaded to Biotage® using a 40 g Agela flash silica gel column, eluted with 0% to 35% EtOAc in PE with the flower rate of 75 mL/min. The product fraction was combined and evaporated to give (S)-2-((tert-butoxycarbonyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoic acid (5.2 g, 14.53 mmol, 98% yield, 100% purity) as a yellow oil. LCMS (ESI+): m/z 380.2 (M+Na+).


Step 8: Int. 26



embedded image


To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(7-chloro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)propanoic acid (2.6 g, 7.27 mmol, 1 eq.) in THF (60 mL) was added NaH (1.45 g, 36.33 mmol, 60% purity, 5 eq) at 0° C., the mixture was stirred at 0° C. for 1 h under N2. MeI (10.31 g, 72.67 mmol, 4.52 mL, 10 eq.) was added to the reaction mixture, the mixture was stirred at 20° C. for 12 h under N2. (Batch 2) The reaction was monitored by LCMS, which showed starting material was consumed and one of main peaks with desired mass was detected. The reaction was quenched by saturated ammonium chloride (30 mL) at 0° C., extracted with EtOAc (60 mL×2). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered and the reaction mixture was concentrated under reduced pressure to give Int. 26 (3.9 g, 10.0 mmol, 69% yield, 96% purity) as a pale-yellow solid.). LCMS (ESI+): m/z 394.1 (M+Na+). 1H NMR (400 MHz, CHLOROFORM-d). δ ppm 1.40 (d, J=10.8 Hz, 9H) 2.67-2.82 (m, 3H) 3.00-3.13 (m, 1H) 3.15-3.35 (m, 1H) 4.18-4.34 (m, 4H) 4.61-4.91 (m, 1H) 6.68 (d, J=11.6 Hz, 1H) 6.79 (d, J=2.0 Hz, 1H).


Int. 27: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-methylpyridin-3-yl)propanoic acid



embedded image


Step 1: (5-chloro-2-methylpyridin-3-yl)methanol



embedded image


NaBH4 (6.11 g, 161.63 mmol, 3 eq.) was added into a mixture of methyl 5-chloro-2-methylpyridine-3-carboxylate (10 g, 53.88 mmol, 1 eq.) in MeOH (200 mL) at 0° C. under air atmosphere. The mixture was stirred overnight at rt. Then the reaction was quenched with water (100 mL) and concentrated in vacuo. The residual water phase was extracted with EtOAc (2×200 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (10:1) to afford (5-chloro-2-methylpyridin-3-yl)methanol (6.1 g, 72%) as a yellow oil. LCMS (ESI+): m/z 158.0.


Step 2: 3-(bromomethyl)-5-chloro-2-methylpyridine



embedded image


PBr3 (31431.18 mg, 116.12 mmol, 3 eq.) was added into a mixture of (5-chloro-2-methylpyridin-3-yl)methanol (6.1 g, 38.71 mmol, 1 eq.) in DCM (50 mL) dropwise at 0° C. under nitrogen atmosphere. The mixture was stirred for 3 h at 0° C. The resulting mixture was quenched with water (50 mL) at 0° C. and extracted with EtOAc (2×100 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. This resulted in 3-(bromomethyl)-5-chloro-2-methylpyridine (7 g, 82%) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 222.1.


Step 3: Tert-butyl (2S)-3-(5-chloro-2-methylpyridin-3-yl)-2-[(diphenylmethylidene)amino]propanoate



embedded image


To a stirred mixture of 3-(bromomethyl)-5-chloro-2-methylpyridine (6.1 g, 27.67 mmol, 1 eq.) and tert-butyl 2-[(diphenylmethylidene)amino]acetate (12257.90 mg, 41.50 mmol, 1.5 eq.) in DCM (60 mL) was added KOH (15521.82 mg, 276.66 mmol, 10 eq.) in H2O (15 mL) in portions at −10° C. under air atmosphere. The resulting mixture was stirred overnight at −10° C. The resulting mixture was extracted with DCM (2×100 mL) and the combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (10:1) to afford tert-butyl (2S)-3-(5-chloro-2-methylpyridin-3-yl)-2-[(diphenylmethylidene)amino]propanoate (6.4 g, 53%) as a yellow oil. LCMS (ESI+): m/z 435.0.


Step 4: (S)-2-amino-3-(5-chloro-2-methylpyridin-3-yl)propanoic acid



embedded image


A mixture of tert-butyl (2S)-3-(5-chloro-2-methylpyridin-3-yl)-2-[(diphenylmethylidene)amino]propanoate (300 mg, 0.69 mmol, 1 eq.) and HCl (6M) (100.00 mL, 3291.28 mmol, 119.30 eq.) in THF (10 mL) was stirred overnight at 55° C. under air atmosphere. The resulting mixture was concentrated under vacuum. This resulted in (2S)-2-amino-3-(5-chloro-2-methylpyridin-3-yl) propanoic acid (5 g, 84.43%) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 215.0.


Step 5: (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-methylpyridin-3-yl)propanoic acid



embedded image


To a stirred mixture of (2S)-2-amino-3-(5-chloro-2-methylpyridin-3-yl)propanoic acid (4 g, 18.64 mmol, 1 eq.) and Boc2O (12.20 g, 55.91 mmol, 3.00 eq.) in H2O (100 mL) was added Na2CO3 (5.93 g, 55.91 mmol, 3 eq.) in portions at rt under air atmosphere. The resulting mixture was stirred overnight at rt. The mixture was acidified to pH=6 with diluted HCl (aq. 1N). The resulting mixture was stirred overnight at 0° C. The resulting mixture was extracted with EtOAc (2×100 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% TFA), 10% to 100% gradient in 35 min; detector, UV 220 nm. This resulted in (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-methylpyridin-3-yl)propanoic acid (5.6 g, 95%) as a white solid. LCMS (ESI+): m/z 315.10. 1H NMR (400 MHz, DMSO-d6): δ12.82 (s, 1H), 8.33 (d, J=2.4 Hz, 1H), 7.65 (d, J=2.5 Hz, 1H), 7.21 (d, J=8.8 Hz, 1H), 4.28-4.10 (m, 1H), 3.11 (dd, J=14.3, 4.4 Hz, 1H), 2.81 (dd, J=14.4, 11.0 Hz, 1H), 2.47 (s, 3H), 1.30 (s, 9H).


Step 6: Int. 27



embedded image


NaH (1.19 g, 49.56 mmol, 6 eq.) was added into a stirred mixture of (2S)-2-[(tert-butoxycarbonyl)amino]-3-(5-chloro-2-methylpyridin-3-yl)propanoic acid (2.6 g, 8.26 mmol, 1 eq.) in THF (25 mL) portion wise at 0° C. under nitrogen. Then the mixture was stirred for 1 h at 0° C. MeI (11.72 g, 82.60 mmol, 10 eq.) was added into the above solution at 0° C. under nitrogen atmosphere. The resulting mixture was stirred overnight at rt. The reaction was quenched by the addition of sat. NH4Cl (aq.) (20 mL) at 0° C. The mixture was acidified to pH=6 with diluted HCl (aq. 1N). The resulting mixture was extracted with EtOAc (2×50 mL), and the combined organic layers were dried over anhydrous Na2SO4 and filtered. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 100% gradient in 35 min; detector, UV 210 nm. This resulted in Int. 27 (3.71 g, 133% mass recovery) as a yellow solid. LCMS (ESI+): m/z 329.10. 1H NMR (400 MHz, DMSO-d6): δ 12.98 (s, 1H), 8.34 (dd, J=16.6, 2.4 Hz, 1H), 7.59 (dd, J=21.9, 2.4 Hz, 1H), 4.72 (ddd, J=53.9, 11.2, 4.4 Hz, 1H), 3.27-2.92 (m, 2H), 2.65 (d, J=2.0 Hz, 3H), 2.50 (s, 3H), 1.25 (d, J=34.2 Hz, 9H).


Int. 28: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoic acid



embedded image


Step 1: 5-chloro-3-iodo-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine



embedded image


To a solution of 2,5-dichloro-3-iodopyridine (10 g, 36.51 mmol, 1 eq) in DMF (140 mL) was added Cs2CO3 (23.79 g, 73.02 mmol, 2 eq) and 1-methylpyrazol-4-ol (4.30 g, 43.81 mmol, 1.2 eq). The mixture was stirred at 80° C. for 1 hr. LCMS showed 2,5-dichloro-3-iodopyridine was consumed completely and desired mass was detected. After cooling to rt, the reaction mixture was diluted with water (200 mL), extracted with EtOAc (100 mL×3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The resulting crude product was purified by flash chromatography on silica gel (0-22% EtOAc in PE). 5-chloro-3-iodo-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine (10.9 g, 29.59 mmol, 81% yield, 91% purity) was obtained as a yellow solid.


Step 2: methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoate



embedded image


A mixture of Zn (18.08 g, 276.49 mmol, 3.5 eq) in DMF (150 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 120° C. for 10 min under N2 atmosphere. Then I2 (6.01 g, 23.70 mmol, 4.77 mL, 0.3 eq) in DMF (20 mL) was added dropwise at 20° C., methyl (R)-2-((tert-butoxycarbonyl)amino)-3-iodopropanoate (26 g, 79.00 mmol, 1 eq) in DMF (70 mL) was added dropwise, I2 (6.01 g, 23.70 mmol, 4.77 mL, 0.3 eq) in DMF (20 mL) was added dropwise, the reaction mixture was stirred at 20° C. for 1 hr. The zincate was used directly.


To a solution of 5-chloro-3-iodo-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine (9.9 g, 29.51 mmol, 1 eq) in DMF (260 mL) was added Pd2(dba)3 (1.35 g, 1.48 mmol, 0.05 eq) and sPhos (1.21 g, 2.95 mmol, 0.1 eq) under N2 atmosphere, and then was added dropwise, (R)-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)zinc(II) iodide (24.33 g in DMF, 61.67 mmol, 2.09 eq), under N2 atmosphere and the reaction was stirred at 65° C. for 12 hr under N2. LCMS showed 5-chloro-3-iodo-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine was consumed completely and desired mass was detected. The mixture was quenched by NH4Cl (200 mL) and extracted with EtOAc (200 mL×3). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The resulting crude product was purified by flash chromatography on silica gel (0-21% EtOAc in PE) to give methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoate (5.6 g, 11.97 mmol, 41% yield, 87.8% purity) as a pale-yellow gum.


Step 3: (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoic acid



embedded image


To a solution of methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoate (4.6 g, 11.20 mmol, 1 eq) in THF (40 mL) was added LiOH·H2O (704.75, 16.79 mol, 1499.99 eq) in H2O (20 mL) at 0° C. and the reaction was stirred at 25° C. for 0.5 hr. LCMS showed methyl(S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoate was consumed completely and desired mass was detected. The reaction mixture was washed with MTBE (10 mL×2), then the aqueous phase was adjusted pH to 3-4 with 1M HCl, extracted with EtOAc (30 mL×3). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoic acid (4 g, 9.37 mmol, 84% yield, 93% purity) was obtained as a pale yellow gum without purification.


Step 4: Int. 28



embedded image


To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoic acid (3 g, 7.56 mmol, 1 eq) in THF (90 mL) was added NaH (1.51 g, 37.80 mmol, 60% purity, 5 eq) at 0° C. under N2 atmosphere, the mixture was stirred at 0° C. for 30 min then was added MeI (10.73 g, 75.60 mmol, 4.71 mL, 10 eq). The mixture was warmed to 25° C. and stirred at 25° C. for 12 hr. LCMS showed (S)-2-((tert-butoxycarbonyl)amino)-3-(5-chloro-2-((1-methyl-1H-pyrazol-4-yl)oxy)pyridine-3-yl)propanoic acid was consumed completely and desired mass was detected. The reaction mixture was quenched with NH4Cl (100 mL), adjusted pH to 3-4 with 1M HCl, extracted with EtOAc (30 mL×4). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was diluted with MeOH (50 mL) and washed with PE (20 mL×4). The residue was concentrated under reduced pressure to give pale yellow solid. Int. 28 (2.6 g, 6.10 mmol, 81% yield, 92.8% purity) was obtained as a pale-yellow solid without purification.


Int. 29: (S)-3-(2-((1-acetylazetidin-3-yl)oxy)-5-chloropyridin-3-yl)-2-((tert-butoxycarbonyl)(methyl)amino)propanoic acid



embedded image


This material was synthesized by following the procedure for Int. 13 with 1-(3-hydroxyazetidin-1-yl)ethan-1-one as the starting alcohol. LCMS (ESI+): m/z 428.1 (M+H+)


Int. 30: Preparation of (S)-3-(2-bromo-5-chlorophenyl)-2-((tert-butoxycarbonyl)(methyl)amino)propanoic acid



embedded image


Step 1: (2S,5R)-2-(2-bromo-5-chlorobenzyl)-3,6-diethoxy-5-isopropyl-2,5-dihydropyrazine



embedded image


To a solution of (R)-3,6-diethoxy-2-isopropyl-2,5-dihydropyrazine (151 g, 711 mmol, 1.00 eq) in THF (1500 mL) was added n-BuLi (2.5 M, 285 mL, 1.00 eq) at −70° C. under N2, then to the above solution was added 1-bromo-2-(bromomethyl)-4-chlorobenzene (222.5 g, 782 mmol, 1.10 eq) in THF (220 mL) at −70° C. The mixture was stirred at 25° C. for 12 hours. TLC indicated 1-bromo-2-(bromomethyl)-4-chlorobenzene was remained, and some new spots was detected. The reaction was poured into sat. NH4Cl (aq.) 6000 mL, the reaction mixture was partitioned between water phase 6000 mL and THF 6000 mL. The organic phase was separated and dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE/EtOAc=100/0 to 1/1) to give desired (2S,5R)-2-(2-bromo-5-chlorobenzyl)-3,6-diethoxy-5-isopropyl-2,5-dihydropyrazine (620 g, 1.48 mol, 104% yield, 99.3% purity) as a yellow gum. LCMS (ESI+): m/z 415/417 (M+H+)


Step 2: Ethyl(S)-2-amino-3-(2-bromo-5-chlorophenyl)propanoate



embedded image


To a solution of (2S,5R)-2-(2-bromo-5-chlorobenzyl)-3,6-diethoxy-5-isopropyl-2,5-dihydropyrazine (100 g, 240 mmol, 1.00 eq) in ACN (1500 mL) was added HCl (0.20 M, 1.40 L, 1.16 eq) at 0° C., then was stirred at 25° C. for 48 hours. TLC indicated (2S,5R)-2-(2-bromo-5-chlorobenzyl)-3,6-diethoxy-5-isopropyl-2,5-dihydropyrazine was consumed completely. The reaction mixture was concentrated under reduced pressure to remove ACN and extracted with DCM 4000 mL (2000 mL×2), the organic layers was concentrated under reduced pressure to give a residue. Then the water phase was basified with sat. aq. NaHCO3 to pH=7 and extracted with DCM 4.00 L (2.00 L×2). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give desired ethyl(S)-2-amino-3-(2-bromo-5-chlorophenyl)propanoate (143 g, 466 mmol, 49% yield) as a white solid and to give (2S,5R)-2-(2-bromo-5-chlorobenzyl)-3,6-diethoxy-5-isopropyl-2,5-dihydropyrazine (200 g, crude) was obtained as a yellow gum.


Step 3: (S)-2-amino-3-(2-bromo-5-chlorophenyl)propanoic acid



embedded image


To a solution of ethyl(S)-2-amino-3-(2-bromo-5-chlorophenyl)propanoate (143 g, 466 mmol, 1.00 eq) in THF (715 mL) and EtOH (715 mL) was added LiOH·H2O (58.7 g, 1.40 mol, 3.00 eq) at 0° C., then the reaction was stirred at 25° C. for 12 hours. TLC indicated ethyl(S)-2-amino-3-(2-bromo-5-chlorophenyl)propanoate was consumed completely. The reaction mixture was concentrated under reduced pressure at 30° C. to remove half of solvent. It was added into a saturated solution of citric acid (1.50 L) at 5-10° C., to keep all the progress worked under acid condition. Some solid precipitated, then the suspension was filtered, the filter cake was washed with MTBE (1.00 L). The filter cake was collected and dried in vacuum to give desired (S)-2-amino-3-(2-bromo-5-chlorophenyl)propanoic acid (130 g, crude) as yellow solid which was confirmed by LCMS and SFC, then was used to the next step without further purification.


Step 4: (S)-3-(2-bromo-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoic acid



embedded image


To a mixture of (2S)-2-amino-3-(2-bromo-5-chloro-phenyl)propanoic acid (70.00 g, 251.32 mmol, 1 eq) in THF (750 mL) and NaOH (1 N, 750 mL) was added Boc2O (109.70 g, 502.64 mmol, 115.47 mL, 2 eq) at 0° C., the mixture was stirred at 20° C. for 12 h. The LCMS showed that the reactant 1 was consumed completely, and the desired product was detected. The mixture was poured into sat. Citric acid (4 L) and extracted with EtOAc (1 L×3). The organic layer was washed with water (2 L) and brine (2 L), dried over Na2SO4, filtered, and concentrated. The mixture was suspended in PE (1 L) and stirred at 20° C. for 0.5 h. Then the mixture was filtered, and the white solid was collected and dried to dryness to give compound 2 (2S)-3-(2-bromo-5-chloro-phenyl)-2-(tert-butoxycarbonylamino)propanoic acid (124 g, 320.9 mmol, 64% yield, 98% purity) as a white solid. 1H NMR: ET53616-98-P1Z (400 MHz, MeOD) δ 7.54 (d, J=8.4 Hz, 1H), 7.33 (d, J=2.4 Hz, 1H), 7.17-7.15 (m, 1H), 4.49 (dd, J=4.4, 10.4 Hz, 1H), 3.39 (br dd, J=4.4, 9.6 Hz, 1H), 2.94-2.88 (m, 1H), 1.36-1.28 (m, 9H). LCMS (ESI+): m/z 277.9/279.9 (M+H+)


Step 5: Int. 30



embedded image


To a mixture of (2S)-3-(2-bromo-5-chloro-phenyl)-2-(tert-butoxycarbonylamino)propanoic acid (3.00 g×10, 7.92 mmol, 1 eq) in THF (90 mL×10) was added NaH (1.58 g×10, 39.61 mmol, 60% purity, 5 eq) at 0° C., the mixture was stirred at 0° C. for 0.5 h. Then MeI (11.25 g×10, 79.23 mmol, 4.93 mL, 10 eq) was added at 0° C., the mixture was stirred at 15° C. for 12 h. The LCMS showed that the (2S)-3-(2-bromo-5-chloro-phenyl)-2-(tert-butoxycarbonylamino)propanoic acid was consumed completely, and the desired product was detected. The mixture was quenched by sat. Citric acid (1.5 L), extracted with EtOAc (1.5 L×2). The organic layer was washed with brine (1.5 L), dried over Na2SO4, filtered, and concentrated. The residue was purified by column chromatography (SiO2, PE/EtOAc/THF=20/1/0 to 6/1/1) to give crude product. The mixture was combined with ET53616-100,104. The residue was dissolved in DCM (1 L) and concentrated under reduced pressure to give the desired product Int. 30 (114.0 g, 285.70 mmol, 98% purity) as a yellow solid. 1H NMR: ET53616-105-P1Z2 (400 MHz, CHLOROFORM-d) δ 11.44 (brs, 1H), 7.50-7.47 (m, 1H), 7.20-7.19 (m, 1H), 7.13-7.11 (m, 1H), 4.83-4.71 (m, 1H), 3.48-3.43 (m, 1H), 3.31-3.06 (m, 1H), 2.81-2.70 (m, 3H), 1.44-1.37 (m, 9H). LCMS (ESI+): m/z 291.9/293.9 (M+H+).


Int. 31: (S)-3-(2-bromo-5-fluorophenyl)-2-((tert-butoxycarbonyl)(methyl)amino)propanoic acid



embedded image


This material was synthesized by following the procedure for Int. 30 acid using 1-bromo-2-(bromomethyl)-4-fluorobenzene as the starting material.


Int. 33: ((2S,4R)-4-fluoro-1-(1-(trifluoromethyl)cyclopropane-1-carbonyl)pyrrolidine-2-carbonyl)glycine



embedded image


This material was synthesized in an identical way to Int. 34 using commercially available FMOC protected amino acids. LCMS (ESI+): m/z 327.1 (M+H+).


Int. 34: Preparation of ((S)-2-(1-(trifluoromethyl)cyclopropane-1-carboxamido)butanoyl)-L-alanine



embedded image


Steps 1-4: Int. 34



embedded image


A mix of TEA (7.61 g, 10.5 mL, 6 Eq, 75.2 mmol) and (((9H-fluoren-9-yl)methoxy)carbonyl)-L-alanine (3.90 g, 1 Eq, 12.5 mmol) in DCM (50 mL) was added to a pre-swelled 2-Chlorotrityl chloride resin (5 g). The reaction was agitated for 1 h, then filtered. The loaded resin was washed with DMF (3×50 mL). Next, piperidine 20% in DMF (50 mL) was added to the resin and stirred for 1 h, then washed with DMF (3×50 mL). The freshly deprotected resin was next subjected to a pre-activated solution of Fmoc-protected amino acid consisting of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)butanoic acid (4.08 g, 1 eq, 12.5 mmol), HATU (5.24 g, 1.1 Eq, 13.8 mmol) DIPEA (13.0 g, 17.5 mL, 8 Eq, 100 mmol) in DMF (50 mL). The Resin was agitated for 1 h, then washed with DMF (3×50 mL). The deprotection was repeated with the addition of piperidine 20% in DMF (50 mL). The resin was agitated for 1 h, then washed with DMF (3×50 mL). The final coupling was performed by adding another preactivated carboxylic acid solution, consisting of 1-(trifluoromethyl)cyclopropane-1-carboxylic acid (1.93 g, 1 Eq, 12.5 mmol), HATU (5.24 g, 1.1 Eq, 13.8 mmol), and DIPEA (13.0 g, 17.5 mL, 8 Eq, 100 mmol) in DMF (50 mL), to the resin. This was agitated for 1 h, then washed with DMF (3×50 mL), Methanol (3×50 mL), then DCM (3×50 mL). The tripeptide was then cleaved by agitating the resin with a HFIP/DCM mixture (1:1, 2×50 mL). The combined cleavage filtrate was concentrated and the crude residue was purified by FCC (SiO2, MeOH in DCM 0 to 20% over 10 min.). Int. 34 (500 mg, 80% based on 0.5 mmol/resin loading) was isolated as a white solid. LCMS (ESI+): m/z 311.1 (M+H+).


Int. 35: (S)-2-((2S,4R)-4-fluoro-1-(1-trifluoromethyl)cyclopropane-1-carbonyl)pyrrolidine-2-carboxamido)butanoic acid



embedded image


This material was synthesized in an identical way to Int. 34 using commercially available FMOC protected amino acids. LCMS (ESI+): m/z 355.4 (M+H+).


Int. 39: Preparation of (2S,4R)-4-fluoro-1-(2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylic acid



embedded image


Int. 39 was synthesized in accordance with Int. 148 and 149, but without separating the diastereomers.


Int. 40: Preparation of 1-(4-((4,4,5,5,-tetramethyl-1,3,2-dioxaborolan-2-yl)methylene)piperidin-1-yl)ethan-1-one



embedded image


Step 1: Int. 40



embedded image


tert-butyl 4-((4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)methylene)piperidine-1-carboxylate (1.0 g, 1 Eq, 3.1 mmol) was dissolved in 10 mL of DCM/TFA (3:1) and was stirred until the consumption of starting material (monitored by LCMS). The volatile solvent was completely removed and residual TFA was removed via co-evaporation with toluene. The deprotected amine was used directly without further purification. The crude material from above was dissolved in DCM (20 mL) and cooled to 0° C. and TEA (0.94 g, 1.3 mL, 3 eq, 9.3 mmol) and AcCl (0.29 g, 0.26 mL, 1.2 eq, 3.7 mmol) were added subsequently and stirred at 0° C. Upon completion, the volatile solvent was removed, and the resulting solid was placed under high vacuum overnight. The resulting solid Int. 40 (750 mg, 2.83 mmol, 91%) was used in subsequent reactions with no additional purification. LCMS (ESI+): m/z 266.1 (M+H+).


Int. 41: Preparation of (S)-2-((tert-butoxycarbonyl)(ethyl)amino)pent-4-enoic acid



embedded image


Step 1: (2S)-2-(ethylamino) pent-4-enoic acid



embedded image


Into a stirred solution of (2S)-2-aminopent-4-enoic acid (5 g, 43.43 mmol, 1 eq) in MeOH (50 mL) was added NaBH3CN (3.41 g, 54.29 mmol, 1.25 eq) at 0° C. under nitrogen atmosphere. The resulting solution was stirred for 10 min at 0° C. Acetaldehyde (10.97 mL, 195.43 mmol, 1.5 eq) was added and the resulting solution was stirred for 1 h at 0° C. After warming to rt, the mixture was stirred for 16 h at rt. The resulting mixture was quenched with water at 0° C. and extracted with EtOAc (2×10 mL). This resulted in (2S)-2-(ethylamino) pent-4-enoic acid (15 g, 76%) and the crude resulting mixture was used in the next step directly without further purification. LCMS (ESI+): m/z 144.09 (M+H+).


Step 2: Int. 41



embedded image


Into a stirred solution of (2S)-2-(ethylamino) pent-4-enoic acid (15 g, 104.76 mmol, 1 eq) and NaOH (29.33 g, 733.31 mmol, 7 eq) in dioxane (150 mL)/H2O (150 mL) was added Boc2O (45.73 g, 209.52 mmol, 2 eq) at 0° C. under nitrogen atmosphere. The resulting solution was stirred for 16 h at rt. The reaction mixture was concentrated in vacuo to remove dioxane. The aqueous layer was acidified with 1 N HCl to pH=5. The aqueous layer was extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated in vacuo and the residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 0% to 100% gradient in 40 min; detector, UV 210 nm. This resulted in Int. 41 (11.9 g, 46%) as a white solid. LCMS (ESI+): m/z 244.15 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.05 (dt, J=7.0, 7.0, 14.3 Hz, 3H), 1.38 (d, J=14.8 Hz, 9H), 2.51-2.69 (m, 2H), 3.03 (dq, J=6.8, 6.8, 6.8, 18.6 Hz, 1H), 3.26 (ddq, J=6.9, 7.0, 7.0, 14.0, 28.9 Hz, 1H), 4.18 (ddd, J=5.1, 9.9, 114.9 Hz, 1H), 4.97-5.18 (m, 2H), 5.64-5.90 (m, 1H), 12.52 (s, 1H).


Int. 42: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)hex-5-enoic acid



embedded image


Step 1: (S)-1-[(2R)-1-benzylpyrrolidine-2-carbonyl]-5-(but-3-en-1-yl)-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one



embedded image


A solution of 1-[(2R)-1-benzylpyrrolidine-2-carbonyl]-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one (75.31 g, 151.17 mmol, 1 eq) and NaOH (60.46 g, 1511.67 mmol, 10 eq) in DMF (600 mL) was stirred for 15 min at rt under nitrogen atmosphere. 4-bromo-1-butene (20 g, 148.14 mmol, 0.98 eq) was added to the solution dropwise and stirred for 1.5 h at rt. The mixture was acidified to pH=6 with HOAc (5%) and extracted with EtOAc (3×1 L). The combined organic layers were washed with NH4Cl (aq.) (3×1 L) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum and purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (20:1) to afford crude product (5S)-1-[(2R)-1-benzylpyrrolidine-2-carbonyl]-5-(but-3-en-1-yl)-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one (80 g, 86%) as a red solid. LCMS (ESI+): m/z 552.3 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.19 (t, J=7.1 Hz, 7H), 1.59 (dtd, J=13.3, 6.6, 3.3 Hz, 1H), 1.99 (s, 5H), 2.07 (s, 6H), 2.02-2.25 (m, 3H), 2.48 (s, 1H), 3.33 (s, 4H), 3.34-3.48 (m, 1H), 3.48-3.62 (m, 2H), 3.64 (td, J=10.6, 9.7, 3.7 Hz, 1H), 3.99-4.13 (m, 5H), 4.82 (dd, J=10.2, 1.9 Hz, 1H), 4.85-4.97 (m, 1H), 5.49 (s, 1H), 5.95 (s, 1H), 6.55 (dd, J=8.2, 1.7 Hz, 1H), 6.61-6.74 (m, 1H), 7.03-7.16 (m, 2H), 7.13-7.27 (m, 1H), 7.36 (t, J=7.7 Hz, 2H), 7.39-7.49 (m, 1H), 7.45-7.53 (m, 1H), 7.50-7.64 (m, 3H), 7.98 (dd, J=8.7, 1.2 Hz, 1H), 8.32-8.43 (m, 2H).


Step 2: (2S)-2-aminohex-5-enoic acid



embedded image


A solution of (5S)-1-[(2R)-1-benzylpyrrolidine-2-carbonyl]-5-(but-3-en-1-yl)-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one (100 g, 181.06 mmol, 1 eq) and HCl (600 mL, 3N) in THF (600 mL) was stirred for 2 h at 80° C. The mixture was basified to pH=7 with NaOH/Na2CO3. The resulting mixture was extracted with CH2Cl2 (3×500 mL). (2S)-2-aminohex-5-enoic acid (25 g, 427%) was in water layer was used in the next step directly without further purification. LCMS (ESI+): m/z 130.15 (M+H+).


Step 3: (2S)-2-[(tert-butoxycarbonyl)amino]hex-5-enoic acid



embedded image


To the above water solution (1.5 L) including (2S)-2-aminohex-5-enoic acid (25 g, 193.56 mmol, 1 eq) was added Boc2O (84.50 g, 387.12 mmol, 2 eq) was stirred for 16 h at rt. The mixture was acidified to pH=6 with citric acid and extracted with CH2Cl2 (3×500 mL). The combined organic layers were washed with brine (1×200 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, Acetonitrile in Water, 5% to 100% gradient in 30 min; detector, UV 210 nm. This resulted in (2S)-2-[(tert-butoxycarbonyl)amino]hex-5-enoic acid (12 g, 135%) as a light-yellow oil. LCMS (ESI+): m/z 130.15 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.18 (t, J=7.1 Hz, 1H), 1.35 (s, 1H), 1.39 (s, 8H), 1.57-1.69 (m, 1H), 1.66-1.78 (m, 1H), 1.91 (s, 2H), 1.99 (s, 1H), 2.06 (dq, J=13.9, 7.2 Hz, 2H), 3.88 (ddd, J=9.5, 8.1, 4.7 Hz, 1H), 4.03 (q, J=7.1 Hz, 1H), 4.94-5.06 (m, 2H), 5.78 (ddt, J=16.9, 10.2, 6.6 Hz, 1H), 7.08 (d, J=8.1 Hz, 1H), 12.26 (s, 2H).


Step 4: methyl(S)-2-((tert-butoxycarbonyl)(methyl)amino)hex-5-enoate



embedded image


To a stirred solution of (2S)-2-[(tert-butoxycarbonyl)amino]hex-5-enoic acid (6 g, 26.17 mmol, 1 eq) and Ag2O (24.26 g, 104.68 mmol, 4 eq) in DMF (90 mL) was added CH3I (55.72 g, 392.54 mmol, 15 eq) dropwise at 0° C. The resulting mixture was stirred overnight at rt. The resulting mixture was filtered, the filter cake was washed with DMF (3×50 mL). The filtrate was used in the next step directly without further purification. LCMS (ESI+): m/z 258.20 (M+H+).


Step 5: Int. 42



embedded image


To a stirred solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]hex-5-enoate (20 g, 77.72 mmol, 1 eq) in H2O (500 mL)/DMF (500 mL) was added and NaOH (18.65 g, 466.33 mmol, 6 eq) in portions at 0° C. The resulting mixture was stirred for 16 h at rt and acidified to pH 6 with HCl (3N). The mixture was extracted with CH2Cl2 (3×0.5 L). The combined organic layers were washed with brine (3×300 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum and purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, Acetonitrile in Water, 5% to 50% gradient in 30 min; detector, UV 200 nm. This resulted in racemic product (20 g) which was purified by Prep-CHIRAL-HPLC with the following conditions (Column: CHIRALPAK IC-3, 4.6×50 mm, 3 m; Mobile Phase A: Hex (0.1% TFA): IPA=98:2; Flow rate: 1 mL/min; Gradient: 0% B to 0% B; Injection Volume: 5 ul mL) to afford Int. 42 (15.78 g, 83%) as a light yellow oil. LCMS (ESI+): m/z 242.10 (M+H+). 1H NMR (300 MHz, DMSO-d6) δ 1.38 (d, J=13.4 Hz, 17H), 1.71-1.86 (m, 1H), 1.89 (s, 1H), 1.94 (d, J=4.9 Hz, 1H), 1.94-2.12 (m, 3H), 2.72 (s, 6H), 4.26 (dd, J=10.4, 4.6 Hz, 1H), 4.50 (dd, J=10.8, 4.4 Hz, 1H), 4.93-5.05 (m, 3H), 5.02-5.10 (m, 1H), 5.81 (ddd, J=16.9, 10.2, 3.6 Hz, 1H), 12.73 (s, 2H).


Int. 43: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)hept-6-enoic acid



embedded image


Step 1: (5S)-1-[(2S)-1-benzylpyrrolidine-2-carbonyl]-5-(pent-4-en-1-yl)-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one



embedded image


To a solution of NaOH (80.51 g, 2013.0 mmol, 10 eq.) in DMF (270 mL), 1-[(2S)-1-benzylpyrrolidine-2-carbonyl]-7-phenyl-1H,4H,5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one (102.29 g, 205.33 mmol, 1.02 eq) was added in at 25° C. under nitrogen atmosphere. The reaction was stirred for 15 min at 25° C. 5-bromopent-1-ene (30 g, 201.30 mmol, 1 eq) was dropwise below 25° C. The reaction was stirred for 5 hours at 25° C. The reaction was purified together with Page EB2141275-183 and EB2141275-184. The mixture was acidified to pH=3 with acetic acid (5%), The resulting mixture was extracted with EtOAc (2×250 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (20:1) to afford (5S)-1-[(2S)-1-benzylpyrrolidine-2-carbonyl]-5-(pent-4-en-1-yl)-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one (88 g, 77%) as a yellow oil. LCMS (ESI+): m/z 566 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.54 (dddd, 2H), 1.76-2.04 (m, 3H), 2.12-2.34 (m, 3H), 2.45 (t, 2H), 3.32 (d, 2H), 3.50-3.71 (m, 3H), 4.07 (d, 1H), 4.85-5.09 (m, 2H), 5.76 (ddt, 1H), 6.53 (dd, 1H), 6.66 (ddd, 1H), 7.01-7.24 (m, 3H), 7.36 (t, 2H), 7.45-7.66 (m, 4H), 7.91-8.09 (m, 1H), 8.27-8.46 (m, 2H).


Step 2: (S)-2-aminohept-6-enoic acid



embedded image


To a solution of (5S)-1-[(2S)-1-benzylpyrrolidine-2-carbonyl]-5-(pent-4-en-1-yl)-7-phenyl-5H-benzo[d]1-oxa-3,7-diaza-2-nickelacyclononan-4-one (88 g, 155.39 mmol, 1 eq.) in THF (880.0 g, 12204.1 mmol, 78.54 eq) was added HCl (517.96 mL, 1553.87 mmol, 10 eq) at 25° C. The reaction was stirred for 2 hours at 60° C. The resulting mixture was diluted with water (1500 mL). The mixture was basified to pH=8 with NaOH (aq., 2N). The resulting mixture was extracted with EtOAc (2×500 mL). The aqueous phase (including about 22 g product) was used in next step without further purification. LCMS (ESI+): m/z 144 (M+H+).


Step 3: (2S)-2-[(tert-butoxycarbonyl)amino]hept-6-enoic acid



embedded image


To a solution of (2S)-2-aminohept-6-enoic acid (22 g, 153.65 mmol, 1 eq), Na2CO3 (97.71 g, 921.88 mmol, 6 eq) in water (2000 mL) was added Boc2O (100.60 g, 460.94 mmol, 3 eq) at 25° C. The reaction was stirred for 6 hours at 25° C. The mixture was acidified to pH=6 with diluted citric acid. The resulting mixture was extracted with EtOAc (3×150 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (20:1) to afford (2S)-2-[(tert-butoxycarbonyl)amino]hept-6-enoic acid (30 g, 80%) as a yellow oil. LCMS (ESI+): m/z 266 (M+Na+). 1H NMR (400 MHz, DMSO-d6): δ 1.38 (s, 11H), 1.50-1.59 (m, 1H), 1.66 (dddd, 1H), 2.74 (s, 1H), 2.90 (s, 1H), 3.86 (ddd, 1H), 4.95 (ddt, 1H), 5.00 (dt, 1H), 5.68-5.88 (m, 1H), 7.07 (d, 1H), 12.38 (s, 1H).


Step 4: Methyl(S)-2-((tert-butoxycarbonyl)(methyl)amino)hept-6-enoate



embedded image


MeI (175.02 g, 1233.03 mmol, 10 eq) was added into a solution of (2S)-2-[(tert-butoxycarbonyl)amino]hept-6-enoic acid (30 g, 123.30 mmol, 1 eq) and Ag2O (142.87 g, 616.52 mmol, 5 eq) in DMF (200 mL). The reaction was stirred for 6 hours at 25° C. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with DMF (2×5 mL). The filtrate was used in next step without further purification (about 35 g). LCMS (ESI+): m/z 172 (M+H−Boc+).


Step 5: Int. 43



embedded image


NaOH (25.79 g, 644.91 mmol, 5 eq) in water (350.00 mL, 19428.41 mmol, 150.63 eq) was added into a solution of methyl (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]hept-6-enoate (35 g, 128.98 mmol, 1 eq) in DMF (20 mL) at 25° C. The reaction was stirred for 5 hours at 25° C. Desired product could be detected by LCMS. The reaction was purified with Page EB2206983-001 and EB2206983-002 and EB2206983-003 and EB2206983-004. The resulting mixture was diluted with water (500 mL). The mixture was acidified to pH=6 with HCl (aq. 2N). The resulting mixture was extracted with EtOAc (3×1000 mL). The combined organic layers were washed with saturated ammonium chloride (2×500 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water, 10% to 60% gradient in 20 min; detector, UV 254 nm to afford (2S)-2-[(tert-butoxycarbonyl)(methyl)amino]hept-6-enoic acid (31 g, 93.40%) as a light yellow oil. The compound was further purified by PREP_CHIRAL_HPLC with the following conditions: Column: CHIRAL ART Cellulose-SC, 2*25 cm, 5 m; Mobile Phase A: Hex (0.1% FA)—HPLC, Mobile Phase B: IPA—HPLC; Flow rate: 20 mL/min; Gradient: 2% B to 2% B in 17 min; Wave Length: 220/254 nm; RT1 (min): 6.939; RT2 (min): 12.248; Sample Solvent: EtOH—HPLC to afford Int. 43 (21.4 g, 66%) as a light yellow oil. LCMS (ESI+): m/z 280 (M+Na+). 1H NMR (400 MHz, DMSO-d6): δ 1.39 (d, 11H), 1.56-1.88 (m, 2H), 2.05 (qp, 2H), 2.70 (s, 3H), 4.41 (ddd, 1H), 4.92-5.09 (m, 2H), 5.71-5.88 (m, 1H), 12.65 (s, 1H).


Int. 44: 1-(methylsulfonyl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole



embedded image


This material was synthesized according to the procedure for Int. 40 with tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole-1-carboxylate and methanesulfonyl chloride as starting materials. LCMS (ESI+): m/z 274.2 (M+H+).


Int. 45: 1-(3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-8-azabicyclo[3.2.1]oct-2-en-8-yl)ethan-1-one



embedded image


This material was synthesized according to the procedure for Int. 40 with tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-8-azabicyclo[3.2.1]oct-2-ene-8-carboxylate as the starting material LCMS (ESI+): m/z 278.2 (M+H+).


Int. 122: 1-(3,3-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydropyridin-1 (2H)-yl)ethan-1-one



embedded image


Int. 122 was synthesized according to the procedure Int. 40 with tert-butyl 3,3-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-1 (2H)-carboxylate as starting material. LCMS (ESI+): m/z 280.5 (M+H+).


Int. 127: (S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutene-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)acetic acid



embedded image


This material was synthesized in an identical way to Int. 34 using commercially available FMOC protected amino acids. LCMS (ESI+): m/z 417.1 (M+H+).


Int. 129: (S)-2-cyclopropyl-2-((2S,4R)-4-fluoro-1-(1-(trifluoromethyl)cyclopropane-1-carbonyl)pyrrolidine-2-carboxamido)acetic acid



embedded image


This material was synthesized in an identical way to Int. 34 using commercially available FMOC protected amino acids. LCMS (ESI+): m/z 367.4 (M+H+).


Int. 140: Preparation of (2S,4R)-4-fluoro-1-(1-(trifluoromethyl)cyclopropane-1-carbonyl)pyrrolidine-2-carboxylic acid



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141 With 1-(trifluoromethyl)cyclopropane-1-carboxylic acid as the starting material. LCMS (ESI+): m/z 417.99 (M+H+).


Int. 141: Preparation of (2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


Step 1: Methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate



embedded image


A solution of 1-tert-butyl 2-methyl (2S,4R)-4-fluoropyrrolidine-1,2-dicarboxylate (150 g, 101.1 mmol, 1 eq) and HCl(gas) in 1,4-dioxane (758.29 mL, 505.53 mmol, 5 eq) was stirred for 0.5 h at rt and then concentrated under vacuum to afford methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (110 g, yield, 99%, purity 80%) as a white solid. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 148.15 (M+H+).


Step 2: Methyl (2S,4R)-1-[3,3-difluoro-1-(trifluoromethyl)cyclobutanecarbonyl]-4-fluoropyrrolidine-2-carboxylate



embedded image


To a stirred solution of methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (110 g, 103.3 mmol, 1 eq, 80%), 3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carboxylic acid (122.06 g, 103.3 mmol, 1 eq) and TCFH (251.69 g, 154.95 mmol, 1.5 eq) in ACN (1100 mL) was added NMI (245.51 g, 516.49 mmol, 5 eq) dropwise in 45 min at 0° C. The mixture was slowly warmed up to rt and stirred overnight at rt. The mixture was concentrated under vacuum at 28° C. and diluted with EtOAc (900 mL) and washed with HCl (0.5N, 1400 mL×1). The aqueous layer was extracted again with EtOAc (1×500 mL). The combined organic layers were washed with saturated NaHCO3 (1000 mL×1). The aqueous layer was extracted with EtOAc (300 mL×1) again. The combined organic layers were washed with brine and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum to afford methyl (2S,4R)-1-[3,3-difluoro-1-(trifluoromethyl)cyclobutanecarbonyl]-4-fluoropyrrolidine-2-carboxylate 180 g, 81%) as a light-yellow solid. The crude product was used in the next step directly without further purification. LCMS (ESI+): m/z 334.15 (M+H+).


Step 3: Int. 141



embedded image


To a stirred solution of methyl (2S,4R)-1-[3,3-difluoro-1-(trifluoromethyl)cyclobutanecarbonyl]-4-fluoropyrrolidine-2-carboxylate (180 g, 82.15 mmol, 1 eq, 90%) in MeOH (1400 mL) was added dropwise NaOH (58.33 g, 246.45 mmol, 3 eq) in H2O (400 mL) in 30 min at 0-20° C. The mixture was stirred for 2 h at rt. MeOH was evaporated out under vacuum. The residue was diluted with water (2500 mL) and acidified with HCl (3N, 400 mL) at 0-20° C. Then the precipitated solids were collected by filtration and washed with water (3×200 mL). The filtrate was added HCl (3N, 100 mL). The precipitated solids were collected by filtration and washed with water (3×100 mL). The combined solids were dried in an oven for 16 h at 40° C. This resulted in afford Int. 40 (150.7 g, 97%) as a white solid. LCMS (ESI+): m/z 319.95 (M+H+).


Int. 142: Preparation of (2S,4R)-4-fluoro-1-((R)-3,3,3-trifluoro-2-hydroxy-2-methylpropanoyl)pyrrolidine-2-carboxylic acid



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141. With (R)-3,3,3-trifluoro-2-hydroxy-2-methylpropanoic acid as the starting material. LCMS (ESI+): m/z 421.08


Int. 143: Preparation of (2S,4R)-4-fluoro-1-(2-(trifluoromethyl)spiro[3.3]heptane-2-carbonyl)pyrrolidine-2-carboxylic acid



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141 with 2-(trifluoromethyl)spiro[3.3]heptane-2-carboxylic acid as the starting material. LCMS (ESI+): m/z 323.11 (M+H+).


Int. 144: Preparation of (2S,4R)-4-fluoro-1-(1-(trifluoromethyl)cyclobutane-1-carbonyl)pyrrolidine-2-carboxylic acid



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141 with 1-(trifluoromethyl)cyclobutane-1-carboxylic acid as the starting material. LCMS (ESI+): m/z 283.08 (M+H+).


Int. 145: Preparation of (1-(trifluoromethyl)cyclobutane-1-carbonyl)-L-proline



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141 with (tert-butoxycarbonyl)-L-proline and 1-(trifluoromethyl)cyclobutane-1-carboxylic acid as the starting materials. LCMS (ESI+): m/z 266.1 (M+H+).


Int. 146: (3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-L-proline



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141 with (tert-butoxycarbonyl)-L-proline as the starting material. LCMS (ESI+): m/z 302.2 (M+H+).


Int. 147: Preparation of (2S,4R)-1-(1-cyanocyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


This compound was prepared following the general synthetic sequence described for the preparation of Int. 141 with 1-cyanocyclobutane-1-carboxylic acid as the starting material. LCMS (ESI+): m/z 240.09 (M+H+).


Int. 148 and 149: Preparation of (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylic acid and (2S,4R)-4-fluoro-1-((S)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylic acid



embedded image


Step 1: Methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate



embedded image


A mixture of 1-(tert-butyl) 2-methyl (2S,4R)-4-fluoropyrrolidine-1,2-dicarboxylate (20 g, 80.9 mmol, 1 eq) in HCl/EtOAc (200 mL) (4 M) was stirred at 25° C. for 2 h. LC-MS showed starting material was consumed and one main peak with desired mass was detected. The reaction mixture was concentrated to give methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (15 g, crude, HCl) as a light-yellow solid. This was taken on to the next reaction without further purification.


Step 2: (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate and (2S,4R)-4-fluoro-1-((S)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate



embedded image


To a solution of 2-(trifluoromethyl)tetrahydro-2H-pyran-2-carboxylic acid (13 g, 65.61 mmol, 1 eq) in DCM (200 mL) was added DIEA (8.48 g, 65.61 mmol, 11.43 mL, 1 eq), and the mixture was stirred for 10 min, then BOP—Cl (18.37 g, 72.17 mmol, 1.1 eq), methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate, and DIEA (16.96 g, 131.22 mmol, 22.86 mL, 2 eq) were added. The mixture was stirred at 25° C. for 12 h. LC-MS showed one main peak with desired mass was detected. The reaction mixture was concentrated under reduced pressure to remove DCM, and then EtOAc (30 mL) was added. The solution was washed three times with 5% NaHCO3 solution (15 mL) and once consecutively with water (15 mL), 2M HCl solution (15 mL), water (15 mL) and saturated brine (15 mL). The organic layer was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EtOAc=10:1 to 2:1) to give methyl (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate (10.7 g, 31.7 mmol, 48% yield, 96.8% purity) as a light yellow oil and to give methyl (2S,4R)-4-fluoro-1-((S)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate (11 g, 32.0 mmol, 49% yield, 95.2% purity) as a light yellow oil.


Step 3: (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylic acid



embedded image


To a solution of methyl (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate (10.7 g, 31.66 mmol, 96.83% purity, 1 eq) in THF (100 mL) and H2O (100 mL) was added LiOH·H2O (2.66 g, 63.32 mmol, 2 eq). Then the mixture was stirred at 25° C. for 1 hr. LC-MS showed methyl (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate was consumed and one main peak with desired mass was detected. The mixture was acidified at 0° C. with HCl (1 N) until pH=2-3. Then the mixture was extracted with EtOAc 300 mL (100 mL×3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EtOAc=10:1 to 2:1) to give (2S,4R)-4-fluoro-1-((R)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylic acid (8.9 g, 28.33 mmol, 89% yield, 99% purity) as a white solid. LCMS (ESI+): m/z 314.0 (M+H+).


Step 4: Int. 149



embedded image


The same hydrolysis procedure was performed on methyl (2S,4R)-4-fluoro-1-((S)-2-(trifluoromethyl)tetrahydro-2H-pyran-2-carbonyl)pyrrolidine-2-carboxylate to provide the desired product Int. 149 (9.1 g, 28.97 mmol, 90.5% yield, 99.7% purity) as a white solid. LCMS (ESI+): m/z 314.0 (M+H+).


Int. 150: Preparation of (2S,4R)-4-fluoro-1-(3,3,3-trifluoro-2,2-dimethylpropanoyl)pyrrolidine-2-carboxylic acid



embedded image


Int. 150 was prepared following the general synthetic sequence described for the preparation of Int. 141. LCMS (ESI+): m/z 271.08 (M+H+).


Int. 154: Preparation of tert-butyl(S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)pent-4-enoate



embedded image


Step 1: Int. 154



embedded image


To a solution of Int. 152 (250 g, 1.09 mol, 1 eq) in HC/EtOAc (2500 mL). The mixture was stirred at 25° C. for 12 hour. The reaction mixture was concentrated under reduced pressure to remove solvent to give the deprotected HCl salt (180.5 g, crude, HCl) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 14.33-13.58 (m, 1H), 10.10-8.94 (m, 2H), 5.84-5.69 (m, 1H), 5.31-5.11 (m, 2H), 3.99 (br t, J=5.2 Hz, 1H), 2.68 (br t, J=6.4 Hz, 2H), 2.60-2.52 (m, 3H).


To a solution of the crude HCl salt (170 g, 1.03 mol, 1 eq) in H2O (860 mL) and dioxane (1140 mL) was added FmocOSu (346 g, 1.03 mol, 1 eq) and DIEA (398 g, 3.08 mol, 536 mL, 3 eq). The mixture was stirred until the LCMS showed consumption of starting material. The reaction was quenched with citric acid solution (800 mL) at 25° C., and then extracted with EtOAc (500 mL×4). The combined organic layers were washed with brine (500 mL×2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (Eluent of 0-40% EtOAc/PE) to give (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)pent-4-enoic acid (290 g, 815 mmol, 80% yield, 99% purity) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 13.13-12.72 (m, 1H), 7.89 (dd, J=3.7, 7.2 Hz, 2H), 7.63 (br d, J=7.6 Hz, 2H), 7.45-7.27 (m, 4H), 5.73-5.46 (m, 1H), 5.17-4.95 (m, 2H), 4.67-4.44 (m, 1H), 4.40-4.21 (m, 3H), 2.80-2.70 (m, 3H), 2.53 (br s, 1H), 2.48-2.32 (m, 1H).


Step 2: Int. 154



embedded image


Dissolve (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)pent-4-enoic acid (10 g, 1 Eq, 28 mmol) in DCM (20 mL), bring to 0° C. under argon. Add DCC (6.5 g, 1.1 Eq, 31 mmol) and DMAP (0.35 g, 0.1 Eq, 2.8 mmol) as solids. Add tert-butanol (8.4 g, 11 mL, 4 Eq, 0.11 mol) by syringe. Stir at rt overnight. Dilute in water and DCM, extract 3×DCM. Wash combined organic layers with water and brine, dry over MgSO4, filter, concentrate. Purify by normal phase chromatography (0 to 100% EtOAc in hexanes) to yield Int. 154 (9.8 g, 24 mmol, 85%) as a pale oil.


Int. 162: Preparation of (S)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4,4-difluoropyrrolidine-2-carboxylic acid



embedded image


Step 1: 2-benzyl 1-(tert-butyl) (S)-4,4-difluoropyrrolidine-1,2-dicarboxylate



embedded image


To a solution of (S)-1-(tert-butoxycarbonyl)-4,4-difluoropyrrolidine-2-carboxylic acid (2.50 g, 9.95 mmol, 1.0 Equiv.) in DMF (50 mL) was added Na2CO3 (2.11 g, 19.9 mmol, 2.0 Equiv.) and stirred at room temperature for 0.5 hrs. The reaction mixture was then cooled to 0° C. and BnBr (1.87 g, 10.9 mmol, 1.30 mL, 1.1 Equiv.) was added. The mixture was stirred at 0° C. for another 2.5 hrs. Water (80 mL) was added, and the aqueous phase was extracted with EtOAc (30 mL×3), the combined organic layer was washed by brine (30 mL), dried over Na2SO4, filtered and the filtrate was concentrated to give a residue. The residue was purified by silica gel column chromatography (Pet. Ether/EtOAc, 30:1) to afford 2-benzyl 1-(tert-butyl) (S)-4,4-difluoropyrrolidine-1,2-dicarboxylate (2.80 g, 8.20 mmol, 82.4% yield) as a clear oil. 1H NMR (400 MHz, DMSO-d6): δ7.37 (d, J=4.8 Hz, 5H), 5.23-5.10 (m, 2H), 4.59-4.50 (m, 1H), 3.83-3.70 (m, 2H), 3.02-2.87 (m, 1H), 2.59-2.50 (m, 1H), 1.41-1.25 (m, 9H).


Step 2: benzyl(S)-4,4-difluoropyrrolidine-2-carboxylate



embedded image


To a solution of 2-benzyl 1-(tert-butyl) (S)-4,4-difluoropyrrolidine-1,2-dicarboxylate (2.00 g, 5.86 mmol, 1.0 Equiv.) in EtOAc (10 mL) was added HCl/EtOAc (4 M, 40 mL, 27.3 Equiv.) and stirred at 20° C. for 1 hr. The reaction mixture was concentrated to afford benzyl(S)-4,4-difluoropyrrolidine-2-carboxylate (2.14 g, crude) as a white solid. 1H NMR (400 MHz, DMSO-d6): δ10.78-10.59 (m, 1H), 7.48-7.33 (m, 5H), 5.30-5.22 (m, 2H), 4.86 (t, J=8.8 Hz, 1H), 3.83-3.64 (m, 2H), 2.97-2.85 (m, 1H), 2.75 (dq, J=8.8, 14.8 Hz, 1H).


Step 3: benzyl(S)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4,4-difluoropyrrolidine-2-carboxylate



embedded image


To a solution of benzyl(S)-4,4-difluoropyrrolidine-2-carboxylate (1.90 g, 7.88 mmol, 1.0 Equiv.) in DMF (30 mL) was added DIPEA (3.05 g, 23.6 mmol, 4.12 mL, 3.0 Equiv.), 3,3-difluoro-1-(trifluoromethyl)cyclobutanecarboxylic acid (2.41 g, 11.8 mmol, 1.5 Equiv.) and HATU (4.49 g, 11.8 mmol, 1.5 Equiv.), the mixture was stirred at 20° C. for 2 hrs. Water (60 mL) was added and the aqueous phase was extracted with EtOAc (20 mL×3). Then the organic layers was washed with brine (30 mL), dried over Na2SO4, filtered and the filtrate was concentrated to give a residue. The residue was purified by silica gel column chromatography, eluted with Pet. Ether/EtOAc (10:1 to 3:1) to afford benzyl(S)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4,4-difluoropyrrolidine-2-carboxylate (2.30 g, 5.38 mmol, 68.3% yield) as a yellow oil. 1H NMR (400 MHz, DMSO-d6): δ7.41-7.32 (m, 5H), 5.15 (s, 2H), 4.83 (dd, J=4.4, 10.0 Hz, 1H), 4.13-4.00 (m, 2H), 3.34-3.20 (m, 4H), 2.99-2.83 (m, 1H), 2.59-2.51 (m, 1H).


Step 4: Int. 162



embedded image


To a solution of benzyl(S)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4,4-difluoropyrrolidine-2-carboxylate (2.30 g, 5.38 mmol, 1.0 Equiv.) in MeOH (30 mL) was added 10% Pd/C (572 mg, 538 μmol, 0.1 Equiv.) under Ar atmosphere. The suspension was degassed and purged with H2. The mixture was stirred under H2 (15 Psi) at 20° C. for 2 hrs. The reaction is filtered with diatomaceous earth and the filter cake was rinsed with MeOH (10 mL×3). Then the combined filtrates were concentrated under reduced pressure. The resulting residue was purified by reversed-phase flash chromatography (C18, MeCN in Water (0.2% formic acid), 35%-65% gradient over 10 min) to give Int. 162 (1.08 g, 3.03 mmol, 56.2% yield, 94.5% purity) as a white solid after lyophilization. LCMS: m/z 336.0 (M−H+). 1H NMR (400 MHz, DMSO-d6): δ13.03 (s, 1H), 4.68-4.61 (m, 1H), 3.99 (t, J=11.6 Hz, 2H), 3.33-3.23 (m, 4H), 2.94-2.80 (m, 1H), 2.50-2.40 (m, 1H). 17 (s, 12H), 1.12 (s, 6H).


Int. 172: Preparation of 1-methyl-5′-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3′H-spiro[azetidine-3,1′-isobenzofuran]



embedded image


Step 1: 5′-bromo-3′H-spiro[azetidine-3,1′-isobenzofuran]



embedded image


Tert-butyl 5′-bromo-3′h-spiro[azetidine-3,1′-isobenzofuran]-1-carboxylate (300 mg, 1 Equiv., 882 μmol) was dissolved in 3 mL 3N HCl in MeOH and stirred at 50° C. until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford the crude deprotection product as the HCl salt. The material can be used without any further purification in the next reaction. LCMS (ESI+): m/z 240.37 (M+H).


Step 2: 5′-bromo-1-methyl-3′H-spiro[azetidine-3,1′-isobenzofuran]



embedded image


Crude 5′-bromo-3′H-spiro[azetidine-3,1′-isobenzofuran]hydrochloride, formaldehyde (98.2 mg, 90.1 μL, 37% Wt, 1.5 Equiv., 1.21 mmol) and DIPEA (313 mg, 421 μL, 3 Equiv., 2.42 mmol) were dissolved in THE (8.67 mL) stirred at 25° C. for 15 mins. Then, NaBH(OAc)3 (305 mg, 2 Equiv., 1.61 mmol) was added portion-wise to the reaction mixture. The reaction was stirred at room temperature until the starting material was completely consumed (monitored by LCMS). The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2×20 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The crude mixture was purified by silica gel column chromatography, eluted with DCM/MeOH (95:5). The solvent was removed to afford 5′-bromo-1-methyl-3′H-spiro[azetidine-3,1′-isobenzofuran] (185 mg, 728 mol, 90.3%) as white solid. LCMS (ESI+): m/z 253.96 (M+H).


Step 3: Int. 172



embedded image


In a sealed tube were placed 5′-bromo-1-methyl-3′H-spiro[azetidine-3,1′-isobenzofuran] (185 mg, 1 Equiv., 544 μmol), Potassium acetate (213 mg, 136 μL, 4 Equiv., 2.18 mmol) and Bis(pinacolato)diborane (414 mg, 3 Equiv., 1.63 mmol) in Dioxane (2.72 mL). The reaction was degassed for 15 min with Ar and then [1,1′-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) Dichloro (22.2 mg, 0.05 Equiv., 27.2 mol) was added. The reaction was sealed and heated to 80° C. until the complete consumption of starting material (monitored by LCMS). Then the reaction was cooled to room temperature, quenched with water, and the organic layers were extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (2×10 mL) and brine (10 mL), dried (Na2SO4), concentrated, and purified by flash column chromatography, (DCM:MeOH, 95:5) and concentrated. This resulted in Int. 172 (149 mg, 495 μmol, 68.0%) as brown solid. LCMS (ESI+): m/z 387.52 (M+H).


Int. 174: (2S,4R)-1-(1-(difluoromethyl)-3,3-difluorocyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


Step 1: Isopropyl 1-formyl-3,3-dimethoxycyclobutane-1-carboxylate



embedded image


To a stirred solution of 1,1-diisopropyl 3,3-dimethoxycyclobutane-1,1-dicarboxylate (10 g, 34.681 mmol, 1 Equiv.) in DCM (100 mL) was added DIBAL (69.36 mL, 69.362 mmol, 2 Equiv., 1M in DCM) dropwise at −78° C. under argon atmosphere. The resulting mixture was stirred for 4 hr at −78° C. under argon atmosphere. The desired product could be detected by GCMS. The reaction was quenched with 2 N HCl (aq.) at 0° C. The aqueous layer was extracted with CH2Cl2 (50 mL×2). The combined organics were dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with Pet. Ether/EtOAc (5:1) to afford isopropyl 1-formyl-3,3-dimethoxycyclobutane-1-carboxylate (2.1 g, 25.0%) as a colorless oil. GCMS: (EIC): m/z 230.00 (M).


Step 2: Isopropyl 1-formyl-3-oxocyclobutane-1-carboxylate



embedded image


A mixture of isopropyl 1-formyl-3,3-dimethoxycyclobutane-1-carboxylate (2.1 g, 9.120 mmol, 1 Equiv.) in 6N HCl (25 mL) was stirred overnight at room temperature. Desired product could be detected by GCMS. The aqueous layer was extracted with CH2Cl2 (50 mL). The organic layer was washed with brine, dried over anhydrous Na2SO4 and concentrated under reduced pressure. This resulted in isopropyl 1-formyl-3-oxocyclobutane-1-carboxylate (1.0 g, 53.6%) as a colorless oil which was used in the next reaction without further purification. GCMS (EIC): m/z 184.00 (M).


Step 3: Isopropyl 1-(difluoromethyl)-3,3-difluorocyclobutane-1-carboxylate



embedded image


To a stirred solution of isopropyl 1-formyl-3-oxocyclobutane-1-carboxylate (1 g, 5.429 mmol, 1 Equiv.) in DCM (20 mL) was added DAST (4.81 g, 29.860 mmol, 5.5 Equiv.) dropwise at 0° C. under atmosphere of argon. The resulting mixture was stirred overnight at room temperature. Desired product could be detected by GCMS. The reaction was quenched by the addition of sat. NaHCO3 (aq.) (100 mL) at 0° C. The organics were dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2 to afford isopropyl 1-(difluoromethyl)-3,3-difluorocyclobutane-1-carboxylate (1 g, 72.65%) as a colorless oil. GCMS (EIC): m/z 228.00 (M).


Step 4: 1-(difluoromethyl)-3,3-difluorocyclobutane-1-carboxylic acid



embedded image


To a stirred solution of isopropyl 1-(difluoromethyl)-3,3-difluorocyclobutane-1-carboxylate (1.5 g, 6.574 mmol, 1 Equiv.) in THF (20 mL) was added dropwise NaOH (0.79 g, 19.722 mmol, 3 Equiv.) in H2O (20 mL) at 0° C. The resulting mixture was stirred overnight at room temperature. Desired product could be detected by LCMS. The resulting mixture was diluted with water (20 mL) and acidified with HCl (aq.) to pH ˜4. The aqueous layer was extracted with EtOAc (2×30 mL). The combined organics were dried over anhydrous Na2SO4 and concentrated under reduced pressure. This resulted in 1-(difluoromethyl)-3,3-difluorocyclobutane-1-carboxylic acid (730 mg, 56.69%) as a colorless oil. The material was used directly in the next step without further purification. LCMS (ESI): m/z 185.00 (M−H).


Step 5: Methyl (2S,4R)-1-(1-(difluoromethyl)-3,3-difluorocyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate



embedded image


Into a solution of 1-(difluoromethyl)-3,3-difluorocyclobutane-1-carboxylic acid (1 g, 5.373 mmol, 1 Equiv.), TCFH (2.26 g, 8.06 mmol, 1.5 Equiv.) and methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (0.87 g, 5.910 mmol, 1.1 Equiv.) in MeCN (20 mL) was added NMI (3.31 g, 40.297 mmol, 7.5 Equiv.) dropwise at 0° C. under nitrogen atmosphere. The mixture was stirred for 16 h at room temperature. The reaction solution was concentrated in vacuo. The residue was purified by reverse \chromatography (20 g C18; MeCN in Water +0.1% formic acid; 10% to 50% gradient in 40 min). This resulted in methyl (2S,4R)-1-[1-(difluoromethyl)-3,3-difluorocyclobutanecarbonyl]-4-fluoropyrrolidine-2-carboxylate (300 mg, 15.94%) as a brown solid upon removal of solvents. LCMS (ESI): m/z 315.24 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 2.11 (dddd, J=4.0, 8.9, 14.7, 39.8 Hz, 1H), 2.51-2.67 (m, 2H), 2.85-3.24 (m, 4H), 3.31-3.46 (m, 1H), 3.66-3.99 (m, 3H), 4.55 (t, J=8.7, 8.7 Hz, 1H), 5.41 (dt, J=3.3, 3.3, 52.6 Hz, 1H), 6.47 (t, J=55.2, 55.2 Hz, 1H).


Step 6: Int 174



embedded image


Into a solution of methyl (2S,4R)-1-[1-(difluoromethyl)-3,3-difluorocyclobutanecarbonyl]-4-fluoropyrrolidine-2-carboxylate (600 mg, 1.903 mmol, 1 Equiv.) in THF (10 mL) was added LiOH (136.75 mg, 5.709 mmol, 3 Equiv.) in H2O (10 mL) at 0° C. The resulting solution was stirred for 16 hr at room temperature. The reaction mixture was concentrated in vacuo to remove THF. The aqueous layer was acidified with 1 N HCl to pH ˜4 and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine, dried over Na2SO4 and concentrated in vacuo. This resulted in Int. 174 (0.496 g, 84.8%) as a white solid. LCMS (ESI): m/z 302.21 (M+H+). 1H NMR (400 MHz, CD3OD): δ2.07-2.27 (m, 1H), 2.59-2.75 (m, 1H), 2.91-3.16 (m, 3H), 3.20-3.31 (m, 1H), 3.73 (ddd, J=2.7, 12.6, 36.6 Hz, 1H), 3.95 (ddd, J=2.3, 12.6, 17.7 Hz, 1H), 4.63 (t, J=8.8, 8.8 Hz, 1H), 5.35 (dd, J=3.4, 52.1 Hz, 1H), 6.24 (t, J=55.6, 55.6 Hz, 1H).


Int. 176: 2-((1S,6R)-3-oxabicyclo[4.1.0]heptan-6-yl)-5-(4,4,5,5-tetraethyl-1,3,2-dioxaborolan-2-yl)pyridine



embedded image


Step 1: 5-bromo-2-(3,6-dihydro-2H-pyran-4-yl)pyridine



embedded image


To a stirred solution of 5-bromo-2-iodopyridine (5 g, 17.612 mmol, 1 Equiv.) and 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (4.44 g, 21.134 mmol, 1.2 Equiv.) in dioxane (100 mL) and H2O (50 mL) were added PdCl2(Dppf)·CH2Cl2 (1.43 g, 1.761 mmol, 0.1 Equiv.) and K2CO3 (4.87 g, 35.224 mmol, 2 Equiv.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 6 hr at 80° C. under nitrogen atmosphere. The resulting mixture was diluted with water (50 mL). The aqueous layer was extracted with EtOAc (3×100 mL). The combined organics were dried over anhydrous Na2SO4 and concentrated under vacuum. The residue was purified by flash chromatography (PE/EtOAc 3:1) to afford 5-bromo-2-(3,6-dihydro-2H-pyran-4-yl)pyridine (2.8 g, 66.2%) as a light yellow solid. LCMS: (ESI): m/z 240.10 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 2.61 (dddq, J=5.6, 3.9, 2.8, 1.4 Hz, 2H), 3.94 (t, J=5.5 Hz, 2H), 4.36 (q, J=2.8 Hz, 2H), 6.70 (tt, J=3.1, 1.6 Hz, 1H), 7.28 (d, J=8.4 Hz, 1H), 7.78 (dd, J=8.5, 2.4 Hz, 1H), 8.61 (d, J=2.4 Hz, 1H).


Step 2: 2-(3-oxabicyclo[4.1.0]heptan-6-yl)-5-bromopyridine



embedded image


To a stirred mixture of trimethylsulfoxonium chloride Th(3.00 g, 23.324 mmol, 2.00 Equiv.) and NaH (0.93 g, 23.324 mmol, 2.00 Equiv., 60%) in DMSO (30 mL) was added 5-bromo-2-(3,6-dihydro-2H-pyran-4-yl)pyridine (2.8 g, 11.662 mmol, 1 Equiv.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for overnight at 65° C. under nitrogen atmosphere. The reaction was quenched with sat. NH4Cl (aq.) at 0° C. and extracted with EtOAc (3×100 mL). The combined organics were dried over anhydrous Na2SO4 and concentrated in vacuo. The residue was purified by reversed-phase flash chromatography (C18 silica gel; mobile phase, MeCN in Water with 10 mmol/L NH4HCO3, 5% to 95) This resulted in 2-(3-oxabicyclo[4.1.0]heptan-6-yl)-5-bromopyridine (1.1 g, 37.12%) as a light yellow oil. LCMS (ESI): m/z 254.0 (M+H+). 1H NMR (400 MHz, Chloroform-d): δ 1.08 (dd, J=6.3, 4.2 Hz, 1H), 1.27 (dd, J=9.1, 4.2 Hz, 1H), 1.69 (dddd, J=9.1, 6.1, 4.3, 1.5 Hz, 1H), 2.11 (ddd, J=14.2, 8.9, 5.6 Hz, 1H), 2.44 (dt, J=13.9, 4.9 Hz, 1H), 3.46 (ddd, J=11.6, 8.9, 5.1 Hz, 1H), 3.64 (dt, J=11.6, 5.2 Hz, 1H), 3.91-4.09 (m, 2H), 7.12 (d, J=8.3 Hz, 1H), 7.71 (dd, J=8.5, 2.4 Hz, 1H), 8.53 (d, J=2.4 Hz, 1H).


Step 3: 2-((1S,6R)-3-oxabicyclo[4.1.0]heptan-6-yl)-5-bromopyridine



embedded image


The 2-(3-oxabicyclo[4.1.0]heptan-6-yl)-5-bromopyridine (800 mg, 3.148 mmol, 1 Equiv.) was purified by Prep-Chiral-HPLC with the following conditions (Column: CHIRALPAK IG, 2×25 cm, 5 m; Mobile Phase A: Hex(0.5% 2M NH3-MeOH), Mobile Phase B: MeOH:DCM=1: 1—HPLC; Flow rate: 20 mL/min; Gradient: 10% B) to afford 2-((1S,6R)-3-oxabicyclo[4.1.0]heptan-6-yl)-5-bromopyridine (250 mg, 31.25%) as a colorless oil. LCMS (ESI): m/z 254.00 (M+H+).


Step 4: Int. 176



embedded image


To a stirred solution of 2-((1S,6R)-3-oxabicyclo[4.1.0]heptan-6-yl)-5-bromopyridine (240 mg, 0.944 mmol, 1 Equiv.), KOAc (278.06 mg, 2.83 mmol, 3 Equiv.) and 4,4,5,5-tetraethyl-2-(4,4,5,5-tetraethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (414.97 mg, 1.133 mmol, 1.2 Equiv.) in dioxane (10 mL) was added Pd(dppf)Cl2·CH2Cl2 (76.93 mg, 0.094 mmol, 0.1 Equiv.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 80° C. under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-TLC (Pet. Ether/EtOAc 7:1) to afford Int. 176 as a colorless oil. LCMS (ESI): m/z 358.15 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 0.91 (t, J=7.4 Hz, 12H), 1.03 (dd, J=6.3, 3.9 Hz, 1H), 1.27 (dd, J=9.0, 3.8 Hz, 1H), 1.58 (q, J=7.5 Hz, 2H), 1.63-1.78 (m, 9H), 1.98 (ddd, J=13.8, 8.6, 5.4 Hz, 1H), 2.53-2.62 (m, 1H), 3.35-3.44 (m, 1H), 3.53 (dt, J=11.1, 5.3 Hz, 1H), 3.81 (d, J=11.3 Hz, 1H), 3.92 (dd, J=11.3, 4.6 Hz, 1H), 7.35 (d, J=8.0 Hz, 1H), 7.92 (dd, J=8.1, 1.8 Hz, 1H), 8.66 (s, 1H).


Int. 178: Preparation of 8-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane



embedded image


Step 1: tert-butyl 3-phenyl-3,8-diazabicyclo[3.2.1]octane-8-carboxylate



embedded image


To a solution of bromobenzene (5 g, 31.85 mmol, 3.35 mL, 1 Equiv.) in dioxane (25 mL) was added t-BuONa in dioxane (2 M, 31.85 mL, 2), tert-butyl 3,8-diazabicyclo[3.2.1]octane-8-carboxylate (7.44 g, 35.03 mmol, 1.1 Equiv.) and RuPhos Pd G4 (2.71 g, 3.18 mmol, 0.1 Equiv.) under N2. The mixture was stirred at 90° C. for 12 hr under N2. LCMS showed starting material was consumed desired mass was detected. The reaction mixture was cooled to room temperature and diluted with water (50 mL), extracted with EtOAc (2×50 mL). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced. The resulting residue was purified by flash silica gel chromatography (Pet. Ether/EtOAc, 5:1) to afford tert-butyl 3-phenyl-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (9 g, 31.2 mmol, 97%) as a white solid. LCMS (ESI+): m/z 289.3 (M+H+). 1H NMR (400 MHz, CDCl3): δ 7.32-7.28 (m, 2H), 6.90-6.81 (m, 3H), 4.49-4.30 (m, 2H), 3.44 (dd, J=1.9, 11.1 Hz, 2H), 3.02 (br s, 2H), 2.02-1.85 (m, 4H), 1.51 (s, 9H).


Step 2: tert-butyl 3-(4-bromophenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate



embedded image


To a solution of tert-butyl 3-phenyl-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (9 g, 31.21 mmol, 1 Equiv.) ethanol (540 mL) was added tetrabutylammonium; bromide (10.06 g, 31.21 mmol, 1 Equiv.) and dibromocopper (10.46 g, 46.81 mmol, 2.19 mL, 1.5 Equiv.) under N2. The mixture was stirred at 25° C. for 12 hrs. LCMS showed starting material was consumed and desired mass was detected. The reaction mixture was filtered, and the filter cake was rinsed with EtOAc (3×20 mL). Then the combined filtrates were concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography (Pet. Ether/EtOAc, 5:1) to afford tert-butyl 3-(4-bromophenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (5.0 g, 13.6 mmol, 44%) as a white solid. LCMS (ESI+): m/z 367.1 (M+H+). 1H NMR (400 MHz, CDCl3): δ 7.33 (d, J=9.0 Hz, 2H), 6.70 (br d, J=9.0 Hz, 2H), 4.36 (br s, 2H), 3.40-3.30 (m, 2H), 2.97 (br s, 2H), 2.02-1.79 (m, 4H), 1.48 (s, 9H).


Step 3: tert-butyl 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate



embedded image


To a solution of tert-butyl 3-(4-bromophenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (4 g, 10.89 mmol, 1 Equiv.) dioxane (40 mL) was added bis(pinacolato)diboron (4.15 g, 16.34 mmol, 1.5 Equiv.), potassium acetate (3.21 g, 32.67 mmol, 3 Equiv.) and Pd(dppf)Cl2 (796.89 mg, 1.09 mmol, 0.1 Equiv.) under N2. The mixture was stirred at 80° C. for 12 hrs under N2. LCMS showed starting material was consumed and desired mass was detected. The reaction mixture was filtered, and the filter cake was rinsed with EtOAc (45 mL). Then the combined filtrates were concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography, eluted with Pet. Ether/EtOAc (4:1) to afford a yellow solid. The yellow solid was triturated with Pet. Ether (30 mL) at 25° C. for 30 min, filtered. The filter cake was concentrated to give tert-butyl 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (3.4 g, 8.21 mmol, 75%) as a white solid. LCMS (ESI+): m/z 415.2 (M+H+). 1H NMR (400 MHz, CDCl3): δ 7.71 (d, J=8.5 Hz, 2H), 6.82 (d, J=8.5 Hz, 2H), 4.48-4.27 (m, 2H), 3.49 (dd, J=1.8, 11.3 Hz, 2H), 3.04 (s, 2H), 2.00-1.80 (m, 4H), 1.48 (s, 9H), 1.38-1.30 (m, 12H).


Step 4: 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane



embedded image


To a solution of tert-butyl 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (4.1 g, 9.90 mmol, 1 Equiv.) in EtOAc (20 mL) was added HCl/EtOAc (4 M, 40 mL, 16.17 Equiv.) dropwise. The mixture was stirred at 25° C. untill LCMS showed the consumption of starting material. The mixture was filtered with EtOAc (30 mL) and the filter cake was concentrated under reduced pressure to give 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane (2.6 g, 7.4 mmol, 75%, HCl Salt) as a white solid. LCMS (ESI+): m/z 315.2 (M+H+). 1H NMR (400 MHz, MeOD-d6): δ7.65 (d, J=8.8 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 4.19 (s, 2H), 3.80 (dd, J=2.4, 13.1 Hz, 2H), 3.13 (d, J=12.1 Hz, 2H), 2.13 (s, 4H), 1.33 (s, 12H)


Step 5: Int. 178



embedded image


To a solution of 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-3,8-diazabicyclo[3.2.1]octane (2.4 g, 6.84 mmol, 1 Equiv., HCl) in methanol (24 mL) was added formaldehyde (2.78 g, 34.22 mmol, 2.55 mL, 37% purity, 5 Equiv.). The mixture was stirred for 30 min under N2. Then Na(CN)BH3 (860.14 mg, 13.69 mmol, 2 Equiv.) was added to the reaction. The mixture was stirred at 25° C. until the consumption of starting material was observed (LCMS). The reaction mixture was diluted by water (50 mL), extracted with EtOAc (2×50 mL). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was triturated with methyl tert-butyl ether (20 mL), then filtered. The filter cake was collected to give Int. 178 (1.07 g, 3.26 mmol, 48%) as a white solid. LCMS (ESI+): m/z 329.2 (M+H+). 1H NMR (400 MHz, MeOD-d6): δ7.58 (d, J=8.6 Hz, 2H), 6.80 (d, J=8.6 Hz, 2H), 3.53-3.48 (m, 2H), 3.35-3.32 (m, 2H), 2.99 (br d, J=10.0 Hz, 2H), 2.36 (s, 3H), 2.14-2.06 (m, 2H), 1.81-1.74 (m, 2H), 1.32 (s, 12H)


Int. 179: Preparation of (1S,4S)-2-methyl-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-2,5-diazabicyclo[2.2.1]heptane



embedded image


Step 1: (1S,4S)-2-(4-bromophenyl)-5-methyl-2,5-diazabicyclo[2.2.1]heptane



embedded image


A mixture of (1S,4S)-2-methyl-2,5-diazabicyclo[2.2.1]heptane; dihydrobromide (3.90 g, 14.2 mmol, 1.0 Equiv.), 1-bromo-4-iodo-benzene (4.03 g, 14.2 mmol, 1.0 Equiv.), Pd2(dba)3 (391 mg, 427 μmol, 0.03 Equiv.), t-BuONa (3.42 g, 35.6 mmol, 2.5 Equiv.) and [1-(2-diphenylphosphanyl-1-naphthyl)-2-naphthyl]-diphenyl-phosphane (88.63 mg, 142.34 μmol, 0.01 Equiv.) in toluene (160 mL) was degassed and purged with N2, then the mixture was stirred at 110° C. for 12 hr under N2 atmosphere. After cooling to room temperature, the reaction mixture was concentrated to remove solvent. Water (50 mL) was added and the aqueous phase was extracted with EtOAc (30 mL×3), the combined organic phase was washed with brine (30 mL×3), dried over Na2SO4, filtered, and concentrated. The resulting residue was dissolved in water (50 mL) and acidified to pH of 5-6 with HCl (1 M), the aqueous phase was extracted with MTBE (30 mL×3). Then the aqueous phase was alkalized to pH of 7-8 with saturated NaHCO3 and extracted with ethyl acetate (50 mL×3), the combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated to give (1S,4S)-2-(4-bromophenyl)-5-methyl-2,5-diazabicyclo[2.2.1]heptane (1.70 g, 6.12 mmol, 43.01% yield) as a yellow oil. LCMS (ESI+): m/z 267.2 (M+H+).


Step 2: Int. 179



embedded image


A mixture of (1S,4S)-2-(4-bromophenyl)-5-methyl-2,5-diazabicyclo[2.2.1]heptane (1.70 g, 6.36 mmol, 1.0 Equiv.), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.75 g, 10.8 mmol, 1.7 Equiv.), KOAc (1.56 g, 15.9 mmol, 2.5 Equiv.), Pd(dppf)Cl2 (232 mg, 318 μmol, 0.05 Equiv.) in dioxane (20 mL) was degassed and purged with N2. The mixture was stirred at 100° C. for 12 hr under N2 atmosphere. After cooling to room temperature, the reaction mixture was concentrated. The resulting residue was diluted with water (30 mL) and EtOAc (20 mL), the resultant mixture was filtered, and the filter cake was rinsed with EtOAc (20 mL×3), the resulting biphasic solution was separated, then the aqueous phase was extracted with EtOAc (30 mL×3), the combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The resulting residue was purified by silica gel column chromatography (Pet. Ether/EtOAc, 1:0 to 20:1) to afford Int. 179 (650 mg, 2.02 mmol, 31.74% yield) as a brown solid. LCMS (ESI+): m/z 315.2 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ7.44 (d, J=8.0 Hz, 2H), 6.53 (d, J=8.0 Hz, 2H), 4.32 (s, 1H), 3.30 (d, J=9.2 Hz, 2H), 3.17 (d, J=9.2 Hz, 1H), 2.78 (d, J=8.8 Hz, 1H), 2.42 (d, J=9.2 Hz, 1H), 2.24 (s, 3H), 1.84-1.91 (m, 1H), 1.75 (d, J=8.8 Hz, 1H), 1.25 (s, 12H).


Int. 180: (2S,4R)-1-(3,3-difluoro-1-methylcyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


Step 1: methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate hydrochloride



embedded image


To a solution of 1-(tert-butyl) 2-methyl (2S,4R)-4-fluoropyrrolidine-1,2-dicarboxylate (5 g, 20.22 mmol, 1 Equiv.) in EtOAc (10 mL) was added dropwise HCl/EtOAc (4 M, 200 mL, 39.56 Equiv.). The mixture was stirred until the complete consumption of starting material (LCMS). The reaction was concentrated under reduced pressure to afford methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate hydrochloride (3.5 g, 17.16 mmol, 84.84% yield, HCl salt) as a white solid. 1H NMR (400 MHz, DMSO-d6): δ 10.25 (br d, J=1.1 Hz, 1H), 5.57-5.32 (m, 1H), 4.55 (dd, J=7.3, 11.3 Hz, 1H), 3.66-3.34 (m, 3H), 2.79-2.51 (m, 3H), 2.43-2.21 (m, 1H).


Step 2: methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate hydrochloride



embedded image


To a solution of 3,3-difluoro-1-methyl-cyclobutanecarboxylic acid (2.70 g, 17.97 mmol, 1.1 Equiv.) in DCM (30 mL) was added DIEPA (2.11 g, 16.34 mmol, 2.85 mL, 1 Equiv.). The mixture was stirred at 20° C. for 10 min. 3-[chloro-(2-oxooxazolidin-3-yl)phosphoryl]oxazolidin-2-one (4.58 g, 17.97 mmol, 1.1 Equiv.) was added to the mixture. Methyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (3 g, 16.34 mmol, 1 Equiv., HCl) and DIEPA (4.22 g, 32.68 mmol, 5.69 mL, 2 Equiv.) were added to the mixture. The mixture was stirred at room temperature until the complete consumption of staring material (LCMS). The reaction mixture was quenched by water (30 mL), extracted with DCM (2×30 mL). The combined organics were washed with NaHCO3 (2×20 mL), brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to afford methyl(2S,4R)-1-(3,3-difluoro-1-methyl-cyclobutanecarbonyl)-4-fluoro-pyrrolidine-2-carboxylate (4.5 g, 14.82 mmol, 90.69%) as a yellow gum. LCMS (ESI+): m/z 280.2 (M+H+).


Step 3: Int. 180



embedded image


To a solution of methyl (2S,4R)-1-(3,3-difluoro-1-methyl-cyclobutanecarbonyl)-4-fluoro-pyrrolidine-2-carboxylate (4.5 g, 16.11 mmol, 1 Equiv.) in THF (20 mL) was added LiOH·H2O (2 M, 12.09 mL, 1.5 Equiv.). The reaction was stirred at 20° C. until the complete consumption of starting material was observed (LCMS). The reaction mixture was concentrated under reduced pressure to remove THF, extracted with MTBE (2×20 mL) and acidified by 1N HCl to pH=1˜2 at 0° C. The aqueous phase was extracted with EtOAc (2×20 mL). The combined organics were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to afford Int. 180 (3.85 g, 13.58 mmol, 84.28%) as a white solid. LCMS (ESI+): m/z 266.00 (M+H+). 1H NMR (400 MHz, CDCl3): 5.40-5.20 (m, 1H), 4.79 (t, J=8.6 Hz, 1H), 3.77 (ddd, J=1.9, 12.6, 17.9 Hz, 1H), 3.66-3.47 (m, 1H), 3.23-2.95 (m, 2H), 2.70-2.37 (m, 4H), 1.55 (s, 3H).


Int. 181: 2,2-dimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholine



embedded image


Step 1: 4-(4-bromophenyl)-2,2-dimethylmorpholine



embedded image


A solution of 4-bromoiodobenzene (2 g, 7.069 mmol, 1 Equiv.), 2,2-dimethylmorpholine (1.63 g, 14.14 mmol, 2 Equiv.), XantPhos (409.06 mg, 0.707 mmol, 0.1 Equiv.), t-BuONa (4.24 mL, 8.483 mmol, 1.20 Equiv.) and Pd2dba3 (323.69 mg, 0.353 mmol, 0.05 Equiv.) in Toluene (20 mL) was stirred for 16 hr at 100° C. under N2 atmosphere. The resulting mixture was diluted with water (50 mL) and then extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: C18; mobile phase, MeCN in Water (0.1% FA), 0% to 100% gradient in 20 min. This resulted in 4-(4-bromophenyl)-2,2-dimethylmorpholine (1.4 g, 73.3%) as a brown oil. LCMS: (ESI): m/z 270.01 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.21 (s, 6H), 2.93 (s, 2H), 2.98-3.08 (m, 2H), 3.67-3.77 (m, 2H), 6.84-6.94 (m, 2H), 7.29-7.38 (m, 2H).


Step 2: Int. 181



embedded image


A solution of 4-(4-bromophenyl)-2,2-dimethylmorpholine (1.41 g, 5.219 mmol, 1 Equiv.), bis(pinacolato)diboron (1.46 g, 5.741 mmol, 1.1 Equiv.), AcOK (1.54 g, 15.657 mmol, 3 Equiv.) and Pd(dppf)Cl2CH2Cl2 (425.14 mg, 0.522 mmol, 0.1 Equiv.) in 1,4-dioxane (15 mL) was stirred for 16 h at 80° C. under N2 atmosphere. The resulting mixture was diluted with EtOAc (100 mL) and then filtered. The filter cake was washed with EtOAc (2×50 mL). The combined filtrates were concentrated under vacuum. The residue was purified FCC (PE/EtOAc 4:1) to afford Int. 181 (1.45 g, 87.3%) as an off-white solid. LCMS (ESI): m/z 318.20 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.23 (d, J=20.8 Hz, 18H), 3.03 (s, 2H), 3.08-3.16 (m, 2H), 3.68-3.76 (m, 2H), 6.85-6.92 (m, 2H), 7.47-7.53 (m, 2H).


Int. 182: 4-(2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholine



embedded image


Step 1: 4-(4-bromo-2-methoxyphenyl)morpholine



embedded image


A solution of morpholine (1.11 g, 12.8 mmol, 2 Equiv.), 4-bromo-1-iodo-2-methoxybenzene (2 g, 6.391 mmol, 1 Equiv.), XantPhos (369.81 mg, 0.639 mmol, 0.1 Equiv.), t-BuONa (3.83 mL, 7.669 mmol, 1.2 Equiv.) and Pd2dba3 (292.63 mg, 0.32 mmol, 0.05 Equiv.) in toluene (20 mL) was stirred for 16 hr at 100° C. under N2 atmosphere. The resulting mixture was diluted with water (50 mL) and then extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (3×50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: C18; mobile phase, MeCN in Water (0.1% FA), 0% to 100%. This resulted in 4-(4-bromo-2-methoxyphenyl)morpholine (1.1 g, 63.2%) as a light yellow solid upon the removal of solvent. LCMS: (ESI): m/z 271.95 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 2.88-2.96 (m, 4H), 3.70 (d, J=9.3 Hz, 4H), 3.80 (s, 3H), 6.82 (d, J=8.4 Hz, 1H), 7.03-7.10 (m, 2H).


Step 2: Int. 182



embedded image


A solution of 4-(4-bromo-2-methoxyphenyl)morpholine (1.4 g, 5.1 mmol, 1 Equiv.), bis(pinacolato)diboron (1.44 g, 5.7 mmol, 1.1 Equiv.), AcOK (1.51 g, 15.4 mmol, 3 Equiv.) and Pd(dppf)Cl2·CH2Cl2 (419.07 mg, 0.514 mmol, 0.1 Equiv.) in 1,4-dioxane (20 mL) was stirred for 16 hr at 80° C. under N2 atmosphere. The resulting mixture was diluted with water (50 mL) and then extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under vacuum. The residue was purified by FCC (PE/EtOAc 3:1) to afford Int. 182 (1.28 g, 78.1%) as an off-white solid. LCMS (ESI): m/z 320.20 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.28 (s, 12H), 2.95-3.03 (m, 4H), 3.72 (t, J=4.6 Hz, 4H), 3.79 (s, 3H), 6.87 (d, J=7.8 Hz, 1H), 7.12 (d, J=1.3 Hz, 1H), 7.24 (dd, J=7.8, 1.3 Hz, 1H).


Int. 183: 1-(pyridin-2-ylmethyl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine



embedded image


Step 1: Int. 183



embedded image


To a solution of 1-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]piperazine (8.7 g, 30.19 mmol, 1 Equiv.) in MeCN (200 mL) was added 2-(bromomethyl)pyridine (5.71 g, 33.21 mmol, 1.1 Equiv.) and K2CO3 (16.69 g, 120.75 mmol, 4 eq). The mixture was stirred at 60° C. for 12 hrs. LCMS showed starting material was consumed and desired mass was detected. The reaction mixture was concentrated under reduced pressure to afford a crude residue. The residue was triturated with Pet. Ether (20 mL), then filtered, the filter cake was dried and collected to afford Int. 183 (8.3 g, 21.9 mmol, 73%) as a yellow solid. LCMS (ESI+): m/z 380.20 (M+H+). 1H NMR (400 MHz, CDCl3): δ 8.65-8.55 (m, 1H), 7.78-7.62 (m, 3H), 7.46 (br d, 1H), 7.19 (dd, 1H), 6.89 (d, 2H), 3.74 (s, 2H), 3.31 (br d, 4H), 2.69 (br s, 4H), 1.33 (s, 12H).


Int. 184: Preparation of (S)-3-(methoxymethyl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholine



embedded image


Step 1: (S)-4-(4-bromophenyl)-3-(methoxymethyl)morpholine



embedded image


A mixture of 1-bromo-4-iodo-benzene (3.80 g, 13.4 mmol, 1.0 Equiv.), (3S)-3-(methoxymethyl)morpholine (2.03 g, 12.1 mmol, 0.9 Equiv., HCl), Pd2(dba)3 (123 mg, 134 mol, 0.01 Equiv.), BINAP (83.6 mg, 134 μmol, 0.01 Equiv.) and t-BuONa (3.87 g, 40.3 mmol, 3.0 Equiv.) in toluene (50 mL) was degassed and purged with N2. The mixture was stirred at 80° C. for 10 hr under N2 atmosphere. The toluene was removed in vacuum, then water (50 mL) and saturated NH4Cl (20 mL) were added, and the aqueous phase was extracted with EtOAc (50 mL×3), the combined organic phase was dried with anhydrous Na2SO4, filtered, and concentrated in vacuum. The resulting residue was purified by silica gel column chromatography (Pet. Ether/EtOAc, 1:0 to 10:1) to afford (S)-4-(4-bromophenyl)-3-(methoxymethyl)morpholine (2.80 g, 9.78 mmol, 72.8%) as brown oil. LCMS (ESI+): m/z 286.3/288.3 (M+H+).


Step 2: Int. 184



embedded image


A mixture of (3S)-4-(4-bromophenyl)-3-(methoxymethyl)morpholine (2.60 g, 9.09 mmol, 1.0 Equiv.), Pd(dppf)Cl2 (332 mg, 454 μmol, 0.05 Equiv.), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.54 g, 9.99 mmol, 1.1 Equiv.) and KOAc (2.23 g, 22.7 mmol, 2.5 Equiv.) in dioxane (30 mL) was degassed and purged with N2. The reaction mixture was stirred at 100° C. for 5 hr under N2 atmosphere. The dioxane was removed in vacuum, water (30 mL) was added and the aqueous phase was extracted with EtOAc (30 mL×3), the combined organic phase was dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was subsequently purified by silica gel column chromatography (Pet. Ether/EtOAc, 1:0 to 10:1) to afford Int. 18 (1.30 g, 3.90 mmol, 42.9%) as white solid. LCMS (ESI+): m/z 334.2 (M+H+). 1H NMR (400 MHz, CDCl3): δ7.79-7.68 (m, 2H), 6.85 (d, J=8.4 Hz, 2H), 4.15 (d, J=11.6 Hz, 1H), 4.05-3.96 (m, 1H), 3.84 (d, J=10.0 Hz, 1H), 3.77-3.68 (m, 3H), 3.32 (s, 3H), 3.30 (s, 2H), 3.19-3.09 (m, 1H), 1.34 (s, 12H).


Int. 186: 4-fluoro-1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine



embedded image


Step 1: 4-(4-bromophenyl)-1-methylpiperidin-4-ol



embedded image


To a solution of 4-(4-bromophenyl)piperidin-4-ol (5 g, 19.520 mmol, 1 Equiv.) in acetone (20 mL) was added MeI (3.05 g, 21.5 mmol, 1.1 Equiv.) and K2CO3 (4.05 g, 29.3 mmol, 1.5 Equiv.). The resulting mixture was stirred for 1 h at room temperature under nitrogen atmosphere. The product was detected by LCMS. The reaction was filtered, and the filter cake was washed with EtOAc. The combined organics were dried over anhydrous Na2SO4 and concentrated in vacuo. The residue was purified by reversed-phase flash chromatography (MeCN in Water with 0.1% FA 5% to 100%) to afford 4-(4-bromophenyl)-1-methylpiperidin-4-ol (1 g, 18.96%) as a white solid after removal of solvents. LCMS: (ESI): m/z 270.00 (M+H+).


Step 2: 4-(4-bromophenyl)-4-fluoro-1-methylpiperidine



embedded image


To a solution of 4-(4-bromophenyl)-1-methylpiperidin-4-ol (2 g, 7.403 mmol, 1 Equiv.) in DCM (30 mL) was added dropwise DAST (2.39 g, 14.806 mmol, 2 Equiv.) in DCM (30 mL) at −78° C. under nitrogen atmosphere. The resulting mixture was stirred warmed to RT and stirred overnight. The desired product could be detected by LCMS. The reaction mixture was quenched by the addition of saturated NaHCO3 at −10° C. and extracted with DCM. The combined organics were dried over anhydrous Na2SO4 and concentrated in vacuo. The resulting crude product was purified by Prep-HPLC (Water with 10 mmol/L NH4HCO3+0.05% NH3H2O and MeOHt 58% to 80% over 10 min) to afford 4-(4-bromophenyl)-4-fluoro-1-methylpiperidine (700 mg, 34.74%) as a white solid. LCMS (ESI): m/z 272.00 (M+H+).


Step 3: Int. 186



embedded image


To a mixture of 4-(4-bromophenyl)-4-fluoro-1-methylpiperidine (700 mg, 2.57 mmol, 1 Equiv.) and bis(pinacolato)diboron (979.70 mg, 3.86 mmol, 1.5 Equiv.) in dioxane (20 mL) were added Pd(dppf)Cl2·CH2Cl2 (209.52 mg, 0.257 mmol, 0.1 Equiv.) and KOAc (757.27 mg, 7.72 mmol, 3 Equiv.) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 80° C. under nitrogen atmosphere. The reaction mixture was diluted with water and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4 and concentrated in vacuo. The residue was purified FCC (DCM/MeOH 10:1) to afford Int. 186 (211 mg, 25.70%) as a light yellow solid after the removal of solvent. LCMS (ESI): m/z 320.25 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.29 (s, 11H), 1.88-2.01 (m, 3H), 2.09-2.32 (m, 2H), 2.42 (s, 3H), 2.51 (s, 2H), 2.93 (s, 2H), 7.40 (dd, J=21.2, 7.9 Hz, 2H), 7.76 (dd, J=42.1, 7.8 Hz, 2H).


Int. 187: Preparation of 2-morpholino-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile



embedded image


2-morpholino-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile (Int. 187): Int. 187 was prepared according to the procedure for step 3 of Int. 172 using 5-Bromo-2-morpholinobenzonitrile as starting material. LCMS (ESI+): m/z 313.85 (M+H).


Int. 189: Preparation of (2S,4R)-4-fluoro-1-(2,3, 3,3-tetrafluoro-2-methylpropanoyl) pyrrolidine-2-carboxylic acid



embedded image


Step 1: benzyl (2S,4R)-4-fluoro-1-(3, 3,3-trifluoro-2-hydroxy-2-methylpropanoyl) pyrrolidine-2-carboxylate



embedded image


To a solution of benzyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (4.24 g, 18.9 mmol, 1.0 Equiv.) in DMF (70 mL) was added DIEPA (7.36 g, 56.9 mmol, 9.92 mL, 3.0 Equiv.), then 3,3,3-trifluoro-2-hydroxy-2-methyl-propanoic acid (3.00 g, 18.9 mmol, 1.0 Equiv.) and HATU (10.8 g, 28.4 mmol, 1.5 Equiv.) were added at 0° C. and stirred at 25° C. for 2 h. The reaction mixture was quenched with water (50 mL) and the aqueous phase was extracted with EtOAc (80 ml×3), the combined organic phase was washed with brine (30 mL×2), dried with anhydrous Na2SO4, filtered, and concentrated in vacuum. The residue was purified by silica gel column chromatography (Pet. Ether/EtOAc, 1:0 to 3:2) to afford benzyl (2S,4R)-4-fluoro-1-(3, 3,3-trifluoro-2-hydroxy-2-methyl-propanoyl) pyrrolidine-2-carboxylate (6.14 g, 16.9 mmol, 89.0%) as a yellow oil. LCMS (ESI+): m/z 364.3 (M+H+). 1H NMR (400 MHz, CDCl3-d): 67.39-7.30 (m, 5H), 5.39-5.22 (m, 1H), 5.21-5.10 (m, 2H), 4.76 (dt, J=2.4, 8.8 Hz, 1H), 4.54-4.30 (m, 1H), 3.92-3.70 (m, 1H), 2.84-2.49 (m, 1H), 2.00-1.75 (m, 1H), 1.65 (d, J=5.2 Hz, 3H).


Step 2: benzyl (2S,4R)-4-fluoro-1-(2,3, 3,3-tetrafluoro-2-methylpropanoyl) pyrrolidine-2-carboxylate



embedded image


To a solution of benzyl (2S,4R)-4-fluoro-1-(3, 3,3-trifluoro-2-hydroxy-2-methylpropanoyl) pyrrolidine-2-carboxylate (2.57 g, 7.07 mmol, 1.0 Equiv.) in DCM (25 mL) was added trifluoro-(morpholino)-sulfane (4.96 g, 28.3 mmol, 3.45 mL, 4.0 Equiv.) and stirred at 50° C. for 12 hr under N2. The reaction mixture was poured into ice (50 ml) and the aqueous phase was extracted with DCM (30 ml×3), the combined organic phase was washed with brine (20 mL×2), dried with anhydrous Na2SO4, filtered, and concentrated in vacuum. The residue was purified by silica gel column chromatography (Pet. Ether/EtOAc, 1:0 to 3:2) to afford benzyl (2S,4R)-4-fluoro-1-(2,3, 3,3-tetrafluoro-2-methylpropanoyl) pyrrolidine-2-carboxylate (2.40 g, 6.57 mmol, 46.4%) as a white solid. LCMS (ESI+): m/z 366.3 (M+H+). 1H NMR (400 MHz, CDCl3): δ7.41-7.32 (m, 5H), 5.37-5.26 (m, 1H), 5.23-5.10 (m, 2H), 4.83-4.71 (m, 1H), 4.37-4.22 (m, 1H), 3.99-3.79 (m, 1H), 2.88-2.54 (m, 1H), 2.28-2.06 (m, 1H), 2.04-1.92 (m, 1H), 1.89-1.72 (m, 3H).


Step 3: Int. 189



embedded image


To a solution of benzyl (2S,4R)-4-fluoro-1-(2,3, 3,3-tetrafluoro-2-methylpropanoyl) pyrrolidine-2-carboxylate (2.40 g, 6.57 mmol, 1.0 Equiv.) in MeOH (25 mL) was added 10% Pd/C (500 mg, 469 μmol) and purged with H2. The mixture was stirred at 25° C. for 2 h under H2 (25 psi) atmosphere. The reaction solution was filtered over Diatomaceous earth, and the filtrate was concentrated under reduced pressure to afford Int. 189 (1.60 g, 5.81 mmol, 88.5%) as a colorless oil. LCMS (ESI+): m/z 276.3 (M+H+). 1H NMR (400 MHz, CDCl3): δ8.25 (d, J=1.2 Hz, 1H), 5.45-5.14 (m, 1H), 4.79-4.62 (m, 1H), 4.30 (ddd, J=6.4, 13.2, 20.0 Hz, 1H), 4.00-3.79 (m, 1H), 2.94-2.57 (m, 1H), 2.32-2.04 (m, 1H), 1.90-1.73 (m, 3H).


Int. 190: 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-6,7-dihydro-4H-pyrazolo[5,1-cl[1,4]oxazine



embedded image


Step 1: 3-(4-bromophenyl)-6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazine



embedded image


A solution of 4-bromoiodobenzene (1 g, 3.535 mmol, 1 Equiv.), 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4H,6H,7H-pyrazolo[3,2-c][1,4]oxazine (0.97 g, 3.889 mmol, 1.1 Equiv.), KF (0.41 g, 7.07 mmol, 2 Equiv.) and Pd(dppf)Cl2·CH2Cl2 (0.23 g, 0.283 mmol, 0.08 Equiv.) in dioxane (10 mL) and water (4 mL) was stirred overnight at 80° C. under nitrogen atmosphere. The residue was diluted with water (20 mL) and extracted with EtOAc (3×50 mL). The combined organics were dried over Na2SO4 and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Pet. Ether/EtOAc (50%) to afford 3-(4-bromophenyl)-4H,6H,7H-pyrazolo[3,2-c][1,4]oxazine (750 mg, 76.01%) as a yellow solid. LCMS: (ESI): m/z 279.05 (M+H+).


Step 2: Int. 190



embedded image


A solution of 3-(4-bromophenyl)-4H,6H,7H-pyrazolo[3,2-c][1,4]oxazine (750 mg, 2.687 mmol, 1 Equiv.), bis(pinacolato)diboron (750.53 mg, 2.96 mmol, 1.1 Equiv.), KOAc (527.39 mg, 5.37 mmol, 2 Equiv.) and Pd(dppf)Cl2·CH2Cl2 (175.10 mg, 0.215 mmol, 0.08 Equiv.) in dioxane (10 mL) was stirred for 3 h at 100° C. under nitrogen atmosphere. The residue was purified by Prep-TLC (PE/EtOAc, 50%) to afford Int. 190 (0.7230 g, 82.49%) as a yellow solid. LCMS (ESI): m/z 327.20 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.07 (s, 3H), 1.16 (s, 4H), 1.30 (s, 10H), 4.21-4.08 (m, 4H), 4.21-4.08 (m, 4H), 7.41-7.33 (m, 2H), 5.03 (s, 2H), 7.71-7.63 (m, 2H), 7.93 (d, J=20.4 Hz, 1H).


Int. 191: Preparation of 1-methyl-4-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-1H-pyrazol-1-yl)piperidine



embedded image


Step 1: 4-(4-(4-bromophenyl)-1H-pyrazol-1-yl)-1-methylpiperidine



embedded image


To a solution of 1-bromo-4-iodobenzene (5.13 g, 18.13 mmol, 1.1 Equiv.) in dioxane (60 mL) was added 1-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazol-1-yl)piperidine (4.8 g, 16.48 mmol, 1 Equiv.), K2CO3 (4.56 g, 32.97 mmol, 2 Equiv.) in H2O (15 mL) and Pd(dppf)Cl2·CH2Cl2 (1.35 g, 1.65 mmol, 0.1 Equiv.) under N2. The reaction stirred at 90° C. for 16 hrs under N2. LCMS showed starting material was consumed completely and desired mass was detected. The reaction mixture was cooled to room temperature and quenched by water (100 mL), extracted with EtOAc (2×60 mL). The combined organics were washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a crude residue. The residue was purified by column chromatography (DCM/methanol, 20:1) to afford crude product. The product was triturated with MTBE (50 mL) and filtered. The filter cake was dried and collected to give 4-(4-(4-bromophenyl)-1H-pyrazol-1-yl)-1-methylpiperidine (2.36 g, 4.79 mmol, 29.1%) as a light yellow solid. LCMS (ESI+): m/z 320.2 (M+H+). 1H NMR (400 MHz, MeOD-d4) δ=8.09 (s, 1H), 7.83 (s, 1H), 7.48 (s, 4H), 4.19 (dt, J=5.5, 10.6 Hz, 1H), 3.01 (br d, J=11.9 Hz, 2H), 2.34 (s, 3H), 2.26-2.21 (m, 2H), 2.15-2.08 (m, 4H)


Step 2: Int. 191



embedded image


The boronic ester, Int. 191, was synthesized using the procedure outlined in Step 2 of the synthesis of Int. 179 using 4-(4-(4-bromophenyl)-1H-pyrazol-1-yl)-1-methylpiperidine as starting material. LCMS (ESI+): m/z 368.3 (M+H+). 1H NMR (400 MHz, CDCl3) δ=7.86-7.76 (m, 3H), 7.74 (s, 1H), 7.49 (d, J=8.1 Hz, 2H), 4.26-4.13 (m, 1H), 3.08 (br d, J=11.9 Hz, 2H), 2.39 (s, 3H), 2.32-2.10 (m, 6H), 1.36 (s, 12H)


Int. 192: Preparation of (2R,6R)-2,6-dimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholine



embedded image


Step 1: (2R,6R)-2,6-dimethyl-4-phenylmorpholine



embedded image


To a solution of bromobenzene (2 g, 12.74 mmol, 1.34 mL, 1 Equiv.) in dioxane (30 mL) was added (2R,6R)-2,6-dimethylmorpholine (1.61 g, 14.01 mmol, 1.1 Equiv.), t-BuONa (2.45 g, 25.48 mmol, 2 Equiv.) and RuPhos Pd G4 (1.08 g, 1.27 mmol, 0.1 Equiv.) under N2. The mixture was stirred at 90° C. for 2 hrs under N2. LCMS showed starting material was consumed and desired mass was detected. The residue was diluted with H2O (50 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, then filtered and concentrated under reduced pressure to give a crude residue. The residue was purified by column chromatography (Pet. Ether/EtOAc, 10:1) to afford (2R,6R)-2,6-dimethyl-4-phenyl-morpholine (1.7 g, 8.90 mmol, 64%) as a yellow oil. LCMS (ESI+): m/z 192.10 (M+H+). 1H NMR (400 MHz, CDCl3): δ 7.26-7.13 (m, 2H), 6.89-6.71 (m, 3H), 4.09 (d, 2H), 3.14 (dd, 2H), 2.82 (dd, 2H), 1.25 (d, 6H)).


Step 2: (2R,6R)-4-(4-bromophenyl)-2,6-dimethylmorpholine



embedded image


To a solution of (2R,6R)-2,6-dimethyl-4-phenyl-morpholine (1.5 g, 7.84 mmol, 1 Equiv.) in EtOH (30 mL) was added tetrabutylammoniumbromide (2.53 g, 7.84 mmol, 1 Equiv.) and CuBr2 (2.63 g, 11.76 mmol, 1.5 Equiv.) under N2. The mixture was stirred at 25° C. for 12 hr. LCMS showed starting material was consumed and desired mass was detected. The resulting mixture was filtered and washed with EtOAc (100 mL). The filtrate was concentrated under reduced pressure to remove EtOH, then diluted with H2O (50 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, then filtered and concentrated under reduced pressure to give a crude residue. The residue was purified by column chromatography (Pet. Ether/EtOAc, 10:1) to afford (2R,6R)-4-(4-bromophenyl)-2,6-dimethylmorpholine (1.5 g, 5.6 mmol, 71%) as a pale-yellow solid. LCMS (ESI+): m/z 270.0 (M+H+). 1H NMR (400 MHz, CDCl3): δ 7.45-7.29 (m, 2H), 6.87-6.62 (m, 2H), 4.16 (d, 2H), 3.18 (dd, 2H), 2.86 (dd, 2H), 1.31 (d, 6H)


Step 3: Int. 192



embedded image


The boronic ester, Int. 192, was synthesized using the procedure outlined in Step 2 of the synthesis of Int. 179 using (2R,6R)-4-(4-bromophenyl)-2,6-dimethylmorpholine as the starting material. LCMS (ESI+): m/z 318.20 (M+H+). 1H NMR (400 MHz, CDCl3): δ 7.72 (d, 2H), 6.85 (d, 2H), 4.26-4.03 (m, 2H), 3.30 (dd, 2H), 2.98 (dd, 2H), 1.37-1.28 (m, 18H)


Int. 193: Preparation of tert-butyl 3-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazin-1-yl)azetidine-1-carboxylate



embedded image


Int. 193 was synthesized following the procedure for Int. 172 (step 2) using 1-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine and tert-butyl-3-oxoazetidine-1-carboxylate as starting materials. LCMS (ESI+): m/z 444.53 (M+H).


Int. 195: N-ethylhex-5-en-1-amine



embedded image


Step 1: Isopropyl 1-formyl-3,3-dimethoxycyclobutane-1-carboxylate



embedded image


Into a solution of hex-5-en-1-amine (500 mg, 5.041 mmol, 1 Equiv.) and TEA (1.53 g, 15.123 mmol, 3 Equiv.) in DCM (70 mL) was added Boc2O (1.32 g, 6.049 mmol, 1.2 Equiv.) dropwise at 0° C. under nitrogen atmosphere. The resulting solution was stirred for 16 hr at room temperature. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Pet. Ether/EtOAc (50:1) to afford tert-butyl N-(hex-5-en-1-yl)carbamate (176.5 mg, 17.42%) as a colorless oil. LCMS: (ESI): m/z 200.16 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.37 (s, 13H), 1.94-2.17 (m, 2H), 2.90 (q, J=6.4, 6.4, 6.5 Hz, 2H), 4.81-5.11 (m, 2H), 5.78 (ddt, J=6.6, 6.6, 10.2, 16.9 Hz, 1H), 6.78 (t, J=5.6, 5.6 Hz, 1H).


Step 2: Tert-butyl ethyl(hex-5-en-1-yl)carbamate



embedded image


Into a solution of tert-butyl N-(hex-5-en-1-yl)carbamate (4.2 g, 21.07 mmol, 1 Equiv.) in THF (40 mL) was added portion wise NaH (1.01 g, 25.29 mmol, 1.2 Equiv., 60%) at 0° C. under nitrogen atmosphere. The mixture was stirred for 30 min at 0° C. Iodoethane (4.93 g, 31.61 mmol, 1.5 Equiv.) was added dropwise. The mixture was stirred for 16 hr at room temperature. The reaction was quenched by the addition of sat. NH4Cl (aq.) (100 mL) at 0° C. and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with Pet. Ether/EtOAc (50:1) to afford tert-butyl ethyl(hex-5-en-1-yl)carbamate (3.04 g, 11.35 mmol, 53.9%) as a yellow oil. LCMS (ESI): m/z 228.35 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 1.01 (t, J=7.0, 7.0 Hz, 3H), 1.26-1.34 (m, 2H), 1.38 (s, 9H), 1.45 (t, J=7.4, 7.4 Hz, 2H), 2.02 (p, J=6.7, 6.7, 6.9, 6.9 Hz, 2H), 3.06-3.18 (m, 4H), 4.85-5.09 (m, 2H), 5.67-5.94 (m, 1H).


Step 3: Int 195



embedded image


Tert-butyl ethyl(hex-5-en-1-yl)carbamate was dissolved in TFA:DCM (1:1, 0.1M) and stirred at RT until the complete consumption of starting material was observed by LCMS. The solvent was removed and the residue was co-evaporated with DCE twice. The crude deprotected amine was used in the next reaction without further purification. LCMS (ESI): m/z 128.35 (M+H+).


Int. 196: Preparation of (2R,6S)-1,2,6-trimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine



embedded image


Step 1: (2R,6S)-4-(4-bromophenyl)-1,2,6-trimethylpiperazine



embedded image


A mixture of 1-bromo-4-iodo-benzene (5.00 g, 17.6 mmol, 1.0 Equiv.), (2S,6R)-1,2,6-trimethylpiperazine (2.27 g, 17.6 mmol, 1.0 Equiv.), tBuONa (5.10 g, 53.0 mmol, 3.0 Equiv.), BINAP (110 mg, 176 μmol, 0.01 Equiv.) and Pd2(dba)3 (161 mg, 176 μmol, 0.01 Equiv.) in toluene (50 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 110° C. for 6 hr under N2 atmosphere. The reaction mixture was concentrated to remove toluene, then water (50 mL) was added and the aqueous phase was extracted with EtOAc (50 mL×3), the combined organic layers was washed with brine (50 mL), dried over Na2SO4, filtered and the filtrate was concentrated to give a residue. The residue was purified by prep-HPLC (TFA condition; column: Phenomenex luna C18 (250×70 mm, 15 um); mobile phase: [H2O (0.1% TFA)-ACN]; gradient: 15%-45% B over 20.0 min) to give (2S,6R)-4-(4-bromophenyl)-1,2,6-trimethyl-piperazine (1.8 g, 6.29 mmol, 35.6%) as a brown solid. LCMS (ESI+): m/z 283.2/285.2 (M+H+).


Step 2: Int. 196



embedded image


The boronic ester, Int. 196, was synthesized using the procedure outlined in Step 2 of the synthesis of Int. 179 using (2S,6R)-4-(4-bromophenyl)-1,2,6-trimethyl-piperazine as starting material LCMS (ESI+): m/z 331.4 (M+H+). 1H NMR (400 MHz, DMSO): δ7.50 (d, J=8.0 Hz, 1H), 6.91 (d, J=8.0 Hz, 1H), 3.71-3.69 (m, 1H), 2.34-2.32 (m, 4H), 2.27 (s, 1H), 1.17 (s, 12H), 1.12 (s, 6H).


Int. 199: Preparation of 1-(oxetan-3-yl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine



embedded image


1-(oxetan-3-yl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine (Int. 199) was prepared according to the procedure for Int. 193 using Oxentan-3-on as starting material. LCMS (ESI+): m/z 344.39 (M+H).


Int. 400: Preparation of (3aR,6aS)-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)hexahydro-1H-furo[3,4-c]pyrrole



embedded image


Step 1: (3aR,6aS)-5-(4-bromophenyl)hexahydro-1H-furo[3,4-c]pyrrole



embedded image


In a sealed tube were placed (3aR,6aS)-hexahydro-1H-furo[3,4-c]pyrrole (109 mg, 1 Equiv., 964 μmol), 1-bromo-4-iodobenzene (300 mg, 1.1 Equiv., 1.06 mmol) and t-BuOK (216 mg, 2 Equiv., 1.93 mmol). Toluene (10.6 mL) was added, and the reaction was degassed for 15 min with Ar. Then, 4,5-Bis(diphenylphosphino)-9,9-dimethyl xanthene (83.7 mg, 0.15 Equiv., 145 μmol) and Tris(dibezylideneacetone)dipalladium (44.1 mg, 0.05 Equiv., 48.2 mol) were added. The reaction was heated to 45° C. until the complete consumption of starting material (monitored by LCMS). Then it was cooled to room temperature, quenched with water and the organic layers were extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (2×10 mL) and brine (10 mL), dried (Na2SO4), concentrated, and purified by flash column chromatography (Hexane:EtOAc 2:3) and concentrated. This resulted in (3aR,6aS)-5-(4-bromophenyl)hexahydro-1H-furo[3,4-c]pyrrole (243 mg, 906 μmol, 94.0%) as yellow solid. LCMS (ESI+): m/z 269.35 (M+H).


Step 2: Int. 400



embedded image


(3aR,6aS)-5-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)hexahydro-1H-furo[3,4-c]pyrrole (Int. 400) was prepared according to the procedure for step 3 of synthesis of Int. 172 using (3aR,6aS)-5-(4-bromophenyl)hexahydro-1H-furo[3,4-c]pyrrole as starting material. LCMS (ESI+): m/z 315.34 (M+H).


Int. 401: Preparation of (R)-8-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)octahydropyrazino[2,1-c][1,4]oxazine



embedded image


Step 1: (R)-8-(4-bromophenyl)octahydropyrazino[2,1-c][1,4]oxazine



embedded image


In a sealed tube were placed (R)-octahydropyrazino[2,1-c][1,4]oxazine dihydrochloride (351 mg, 1 Equiv., 1.63 mmol), 1-bromo-4-iodobenzene (600 mg, 1.3 Equiv., 2.12 mmol) t-BuOK (915 mg, 1.03 mL, 5 Equiv., 8.16 mmol) in DMA (10.6 mL). The reaction was degassed for 15 min with Ar and then Pd(OAc)2 (18.3 mg, 0.05 Equiv., 81.6 mol) and BINAP (102 mg, 0.1 Equiv., 163 μmol) were added. The reaction was heated to 70° C. and stirred until the complete consumption of starting material (monitored by LCMS). The reaction was cooled to room temperature, quenched with water and extracted with EtOAc (3×10 mL) The combined organic layers were washed with water (2×10 mL) and brine (10 mL), dried with Na2SO4, concentrated, and purified by flash column chromatography (DCM/MeOH, 9:1) and concentrated. This resulted in (R)-8-(4-bromophenyl)octahydropyrazino[2,1-c][1,4]oxazine (50 mg, 0.17 mmol, 10%) as yellow solid. LCMS (ESI+): m/z 297.08 (M+H).


Step 2: Int. 401



embedded image


(R)-8-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)octahydropyrazino[2,1-c][1,4]oxazine (Int. 401) was prepared according to the procedure for step 3 of Int. 172 using (R)-8-(4-bromophenyl)octahydropyrazino[2,1-c][1,4]oxazine as starting material. LCMS (ESI+): m/z 344.23 (M+H).


Int. 403: Preparation of tert-butyl 3-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazin-1-yl)azetidine-1-carboxylate
Step 1: Int. 403



embedded image


1-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine (200 mg, 1 Equiv., 694 μmol), DIPEA (179 mg, 242 μL, 2 Equiv., 1.39 mmol), and 2-(2-Methoxyethoxy)ethylbromide (254 mg, 188 μL, 2 Equiv., 1.39 mmol) were dissolved in DCM (2.0 mL) and stirred until the starting material was completely consumed (monitored by LCMS). The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (2×10 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The crude mixture was purified by silica gel column chromatography (DCM/MeOH, 95:5) and concentrated. This afforded Int. 403 (230 mg, 589 μmol, 84.9%) as yellow solid. LCMS (ESI+): m/z 390.67 (M+H).


Int. 404: Preparation of 1-(2,2-difluoroethyl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine



embedded image


1-(2,2-difluoroethyl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine (Int. 402) was prepared according to the procedure for Int. 403 using 2,2-difluoroethyl trifluoromethanesulfonate as starting material. LCMS (ESI+): m/z 352.02 (M+H).


Int. 405: Preparation of 1-((1-methyl-1H-pyrazol-4-yl)methyl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine



embedded image


1-((1-methyl-1H-pyrazol-4-yl)methyl)-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine (Int. 405) was prepared according to the procedure for Int. 193 using 1-methyl-1H-pyrazole-4-carbaldehyde as starting material. LCMS (ESI+): m/z 383.16 (M+H).


Int. 406: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl)amino)-4-methoxy-4-oxobutanoic acid



embedded image


Step 1: (9H-fluoren-9-yl)methyl(S)-4-(2-methoxy-2-oxoethyl)-5-oxooxazolidine-3-carboxylate



embedded image


A mixture of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-4-methoxy-4-oxobutanoic acid (100 g, 270.74 mmol, 1 eq), 37% formaldehyde (43 g, 1432.1 mmol, 39.44 mL, 5.29 eq), TsOH (2.34 g, 13.54 mmol, 0.05 eq) in toluene (1800 mL) was degassed for several minutes, and then the mixture was stirred at 110° C. for 12 h under N2 atmosphere. The resultant mixture was filtered over a pad of diatomaceous earth and the filter cake was washed with EtOAc (200 mL×3). The combined filtrates were concentrated under reduced pressure to give a crude residue. The residue was adjusted to pH ˜9 with a saturated Na2CO3 solution. The aqueous phase was diluted with additional H2O (600 mL) and extracted with EtOAc (300 mL×3). The combined organic layers were dried over Na2SO4, filtered and concentrated to give (9H-fluoren-9-yl)methyl(S)-4-(2-methoxy-2-oxoethyl)-5-oxooxazolidine-3-carboxylate (90 g, 207.20 mmol, 76.53%) as a yellow oil. The product was used with no further purification. LCMS (ESI+): m/z 382.2 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 7.89 (d, J=7.2 Hz, 2H), 7.68-7.61 (m, 2H), 7.46-7.39 (m, 2H), 7.37-7.31 (m, 2H), 5.34 (br d, J=2.0 Hz, 1H), 5.24-5.01 (m, 1H), 4.75-4.18 (m, 4H), 3.60 (s, 3H), 3.19-2.51 (m, 2H).


Step 2: (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)-4-methoxy-4-oxobutanoic acid



embedded image


To a solution of (9H-fluoren-9-yl)methyl(S)-4-(2-methoxy-2-oxoethyl)-5-oxooxazolidine-3-carboxylate (85 g, 195.68 mmol, 1 eq) in CH2Cl2 (1700 mL) was added ZnBr2 (66.10 g, 293.52 mmol, 14.68 mL, 1.5 eq) and Et3SiH (68.26 g, 587.06 mmol, 93.76 mL, 3 eq). The mixture was stirred at 25° C. for 12 h. The resulting mixture was filtered over a pad of diatomaceous earth and the filter cake was washed with additional CH2Cl2 (2×200 mL). The filtrate was diluted with H2O (500 mL) and extracted with EtOAc (200 mL×3). The combined organic layers were dried over Na2SO4, filtered and concentrated to give (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)-4-methoxy-4-oxobutanoic acid (100 g, 194.58 mmol, 99.43%) as a yellow oil. The product was used with no further purification. LCMS (ESI+): m/z 384.1 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 13.42 (s, 1H), 7.93-7.85 (m, 2H), 7.69-7.60 (m, 2H), 7.45-7.38 (m, 2H), 7.37-7.28 (m, 2H), 4.93-4.70 (m, 1H), 4.39-4.21 (m, 3H), 3.65-3.55 (m, 3H), 3.00-2.88 (m, 1H), 2.85-2.76 (m, 3H), 2.76-2.57 (m, 1H).


Step 3: 1-benzyl 4-methyl N-(((9H-fluoren-9-yl)methoxy)carbonyl)-N-methyl-L-aspartate



embedded image


To a solution of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)-4-methoxy-4-oxobutanoic acid (80 g, 208.66 mmol, 1 eq) in DMF (700 mL) was added NaHCO3 (35.06 g, 417.32 mmol, 16.24 mL, 2 eq) at 25° C. for 30 min and BnBr (107.06 g, 625.98 mmol, 74.36 mL, 3 eq) was added at 0° C. The mixture was stirred at 0° C. for 12 h. Then, the mixture was quenched by the addition of H2O (1500 mL) and extracted with EtOAc (500 mL×3). The combined organic layers were washed with brine (200 mL×3) and dried over Na2SO4 and filtered. The filtrate was concentrated and the crude product was purified by column chromatography (SiO2, pet. ether/EtOAc, 7:1 to 3:1) to give 1-benzyl 4-methyl N-(((9H-fluoren-9-yl)methoxy)carbonyl)-N-methyl-L-aspartate (75 g, 142.55 mmol, 68.32%) as a yellow oil. LCMS (ESI+): m/z 474.4 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 7.92-7.83 (m, 2H), 7.64-7.56 (m, 2H), 7.45-7.37 (m, 2H), 7.37-7.31 (m, 2H), 7.31-7.22 (m, 5H), 5.20-4.96 (m, 2H), 4.89-4.75 (m, 1H), 4.46-4.16 (m, 3H), 3.58 (s, 3H), 3.01 (dd, J=6.8, 16.4 Hz, 1H), 2.87-2.73 (m, 4H)


Step 4: 1-benzyl 4-methyl methyl-L-aspartate



embedded image


To a solution of 1-benzyl 4-methyl N-(((9H-fluoren-9-yl)methoxy)carbonyl)-N-methyl-L-aspartate (75 g, 158.38 mmol, 1 eq) in CH2Cl2 (750 mL) was added piperidine (40.46 g, 547.16 mmol, 46.92 mL, 3 eq) at 0° C. The mixture was stirred at 25° C. for 12 h. The reaction mixture was concentrated. H2O (1000 mL) was added to the crude product and the aqueous layer was extracted with EtOAc (500 mL×3). The combined organic layers were dried over Na2SO4, filtered, and the solution was concentrated to give a crude residue. 1-benzyl 4-methyl methyl-L-aspartate (32 g, 113.98 mmol, 71.96%) was isolated as a yellow oil after column chromatography (SiO2, pet. ether/EtOAc, 20:1 to 3:1). LCMS (ESI+): m/z 252.3 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 7.40-7.31 (m, 5H), 5.19 (s, 2H), 3.61-3.52 (s, 3H), 3.52-3.29 (m, 1H), 2.83-2.51 (m, 2H), 2.28-2.15 (s, 3H).


Step 5: 1-benzyl 4-methyl N-(tert-butoxycarbonyl)-N-methyl-L-aspartate



embedded image


To a solution of 1-benzyl 4-methyl methyl-L-aspartate (32 g, 127.34 mmol, 1 eq) in THF (320 mL) and H2O (320 mL) was added Na2CO3 (27 g, 254.7 mmol, 2 eq) and Boc2O (111.18 g, 509.4 mmol, 117.02 mL, 4 eq). The mixture was stirred at 25° C. for 12 h. The reaction mix was then extracted with EtOAc (300 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered, and the filtrate was concentrated to give a crude residue. The residue was purified by column chromatography (SiO2, Pet. ether/EtOAc, 20:1 to 3:1). The product was then purified by prep-HPLC to give 1-benzyl 4-methyl N-(tert-butoxycarbonyl)-N-methyl-L-aspartate (27 g, 74.92 mmol, 58.83%) as a yellow oil. LCMS (ESI+): m/z 374.3 (M+Na+), 1H NMR (400 MHz, DMSO-d6): δ 7.36 (d, J=5.6 Hz, 5H), 5.18-5.07 (m, 2H), 4.82-4.59 (m, 1H), 3.59 (d, J=6.0 Hz, 3H), 3.05-2.97 (m, 1H), 2.86-2.76 (m, 4H), 1.38-1.27 (m, 9H).


Step 6: Int. 406



embedded image


To a solution of 1-benzyl 4-methyl N-(tert-butoxycarbonyl)-N-methyl-L-aspartate (27 g, 76.84 mmol, 1 eq) in MeOH (400 mL) was added Pd/C (8.18 g, 7.68 mmol, 10 Wt.%, 0.1 eq) under N2 atmosphere. The suspension was degassed and purged with H2 (×3). The mixture was stirred under H2 (50 Psi) at 25° C. for 12 h. Upon completion, the reaction was filtered over a pad of diatomaceous earth and the filter cake was washed with EtOAc (100 mL×2). Int. 406 (19.4 g, 67.50 mmol, 87.84%) was isolated as white solid after the removal of solvent. LCMS (ESI+): m/z 284 (M+Na+). 1H NMR (400 MHz, DMSO-d6): δ 12.88 (br s, 1H), 4.75-4.39 (m, 1H), 3.60 (d, J=6.8 Hz, 3H), 2.98-2.88 (m, 1H), 2.80-2.66 (m, 4H), 1.37 (d, J=15.2 Hz, 9H).


Int. 408: Preparation of (S)-2-((tert-butoxycarbonyl)(methyl-d3)amino)pent-4-enoic acid



embedded image


Step 1: Int. 408



embedded image


To a solution of(S)-2-((tert-butoxycarbonyl)amino) pent-4-enoic acid (17.5 g, 81.30 mmol, 1 eq) in THF (800 mL) was added 60% NaH (9.76 g, 243.91 mmol, 3 eq) in portions at 0° C. under a nitrogen atmosphere, and the mixture was stirred at 0° C. for 30 min. Then iodomethane-d3 (82.50 g, 569.12 mmol, 35.42 mL, 7 eq) was added at 0° C., then the reaction was warmed to 25° C. and stirred for 4 h The reaction mixture was cooled to 0° C., quenched with saturated NH4Cl (300 mL). The reaction mixture was extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a thick oil. The residue was purified by column chromatography (SiO2, pet. ether/EtOAc. 10-25%) to give a pale yellow solid. The pale-yellow solid was triturated with pet. ether (45 mL) for 30 min, and the mixture was filtered to give Int. 408 (28 g, 117.22 mmol, 72.09%) as a white solid. LCMS (ESI+): m/z 231.1 (M−H). 1H NMR (400 MHZ, DMSO-d6): δ12.73 (s, 1H), 5.81-5.62 (m, 1H), 5.20-4.93 (m, 2H), 4.62-4.30 (m, 1H), 2.63-2.53 (m, 1H), 2.48-2.39 (m, 1H), 1.37 (d, J=11.4 Hz, 9H).


Int. 411: Preparation of 7-bromo-5-chloro-1-methyl-1H-indole



embedded image


Step 1: Int. 411



embedded image


7-bromo-5-chloro-1H-indole (500 mg, 1 Eq, 2.17 mmol) and THF (10 mL) were cooled to 0° C. and 60% NaH in mineral oil (174 mg, 2 Eq, 4.34 mmol) was added in portions. MeI (924 mg, 407 μL, 3 Eq, 6.51 mmol) was added after 15 min. The reaction was warmed to RT and continued until the complete consumption of starting material (monitored by LCMS). The reaction was carefully quenched with water and was partitioned between water (20 mL) and EtOAc (30 mL). the organic layer was washed with brine, dried with MgSO4, concentrated, and purified by flash column chromatography (SiO2, 0 to 30% EtOAc in Hexanes). 7-bromo-5-chloro-1-methyl-1H-indole (394 mg, 1.61 mmol, 74.3%) was isolated as a white solid. LCMS (ESI+): m/z 245.5 (M+H+).


Int. 412: Preparation 4-bromo-6-chloro-1-methyl-1H-indole




embedded image


Int. 412 was synthesized using the procedure outlined in Int. 411 using 4-bromo-6-chloro-1-methyl-1H-indole as starting material. LCMS (ESI+): m/z 245.5 (M+H+).


Int. 414: Preparation of 2-((1-methylpiperidin-4-yl)oxy)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine



embedded image


Step 1: 5-bromo-2-((1-methylpiperidin-4-yl)oxy)pyridine



embedded image


To a solution of 1-methylpiperidin-4-ol (3.31 g, 28.7 mmol, 3.36 mL, 1.1 eq) in THF (50 mL) was added 60% NaH (2.09 g, 52.2 mmol, 2.0 eq) at 0° C. The reaction was stirred at this temperature for 30 min, then 5-bromo-2-fluoropyridine (4.6 g, 26.1 mmol, 2.69 mL, 1.0 eq) was added and the reaction vessel was warmed to 25° C. and stirred for 2.5 h The reaction mixture was quenched with saturated NH4Cl (50 mL), and the aqueous phase was extracted with EtOAc (80 mL×3). The combined organic layers were dried over Na2SO4, filtered, and concentrated to give 5-bromo-2-((1-methylpiperidin-4-yl)oxy)pyridine (7.8 g, crude) as a yellow oil. The product was used without further purification. LCMS: (ESI+): m/z 271.0 (M+H+).


Step Int. 414



embedded image


A mixture of 5-bromo-2-((1-methylpiperidin-4-yl)oxy)pyridine (7.4 g, 27.3 mmol, 1.0 eq), B2Pin2 (8.32 g, 32.7 mmol, 1.2 eq), KOAc (6.70 g, 68.23 mmol, 2.5 eq) and [2-(2-aminophenyl)phenyl]-chloro-palladium; bis(1-adamantyl)-butyl-phosphane (1.82 g, 2.73 mmol, 0.1 eq) in dioxane (74 mL) was degassed with N2 for 15 min. The mixture was stirred at 120° C. for 12 hrs under N2 atmosphere. After cooling to ambient temperature, the reaction mixture was concentrated and the resultant mixture was dissolved in EtOAc (100 mL), filtered, and concentrated under reduced pressure. The crude residue was purified by flash silica gel chromatography (SiO2, 0-2% Methanol/EtOAc) to give Int. 414 (1.8 g, 5.32 mmol, 19.49%) as a yellow solid. LCMS: (ESI+): m/z 319.1 (M+H+). 1H NMR (400 MHz, DMSO-d6): δ 8.36 (s, 1H), 7.85 (dd, J=8.4, 1.6 Hz, 1H), 6.75 (d, J=8.4 Hz, 1H), 5.03 (dt, J=8.4, 4.0 Hz, 1H), 2.58-2.64 (m, 2H), 2.17 (s, 3H), 2.13 (s, 2H), 1.94 (d, J=9.2 Hz, 2H), 1.63-1.69 (m, 2H), 1.28 (s, 12H).


Int. 415: Preparation of (2S,4R)-1-(1-chlorocyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


Step 1: (cyclopropylsulfinyl)benzene



embedded image


To a stirred mixture of cyclopropyl(phenyl)sulfane (4.8 g, 31.95 mmol, 4.59 mL, 1 eq) in DCM (50 mL) was added m-CPBA (7.13 g, 35.14 mmol, 1.1 eq, 85 wt. %) at 0° C. under N2 atmosphere. The resulting mixture was stirred at 0° C. for 0.5 hr under nitrogen atmosphere until the complete consumption of starting material (monitored by LCMS). The reaction mixture was quenched with a Na2SO3 solution (10%, 50 mL), extracted with DCM (2×100 mL). The combined organics were washed with NaOH (5%, 50 mL) and brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (cyclopropylsulfinyl)benzene (5.2 g, crude) as a light-yellow oil. LCMS (ESI+): m/z 167.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.61-7.59 (m, 2H), 7.45-7.44 (m, 3H), 2.22-2.18 (m, 1H), 1.18-0.99 (m, 1H), 0.97-0.87 (m, 3H)


Step 2: ((1-chlorocyclopropyl)sulfinyl)benzene



embedded image


To a stirred mixture of (cyclopropylsulfinyl)benzene (5.2 g, 31.28 mmol, 1 eq) in CHCl3 (50 mL) was added 1-chloropyrrolidine-2,5-dione (5.01 g, 37.54 mmol, 1.2 eq) at 20° C. under nitrogen atmosphere until the complete consumption of starting material (monitored by LCMS). The residue was diluted with H2O (50 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure. The resulting residue was purified by column chromatography (SiO2, 0-50% EtOAc in Pet. ether). ((1-chlorocyclopropyl)sulfinyl)benzene (5.4 g, 23.09 mmol, 73.8%) was isolated as a clear oil after concentration. LCMS (ESI+): m/z 201.1 (M+H+). 1H NMR (400 MHz, CDCl3-d) δ7.71-7.59 (m, 2H), 7.54-7.43 (m, 3H), 1.66-1.55 (m, 2H), 1.34-1.25 (m, 1H), 1.23-1.13 (m, 1H)


Step 3: 1-chlorocyclopropane-1-carboxylic acid



embedded image


To a stirred mixture of ((1-chlorocyclopropyl)sulfinyl)benzene in THF (60 mL) was added cyclohexylmagnesium chloride (1.3 M, 16.87 mL, 1 eq) and purged with Ar −78° C. After 60 min, gaseous carbon dioxide was bubbled into the reaction for 2 h Upon completion (monitored by TLC), the residue was diluted with a saturated solution of NH4Cl (100 mL), and the reaction mixture was concentrated under reduced pressure to remove THF. Then, the pH was adjusted with saturated NaHCO3 (50 mL) and extracted with MTBE (3×100 mL). Next, the aqueous phase was acidified with 1N HCl (200 mL). When the pH was ˜2-3 the aqueous layer was extracted with EtOAc (3×100 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give 1-chlorocyclopropane-1-carboxylic acid (1.2 g, crude) as a white solid. The product was used with no further purification. 1H NMR (400 MHz, CDCl3-d): δ1.78-1.73 (m, 2H), 1.51-1.45 (m, 2H), 7.99 (s, 1H).


Step 4: benzyl (2S,4R)-1-(1-chlorocyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate



embedded image


To a stirred mixture of 1-chlorocyclopropane-1-carboxylic acid (1.2 g, 9.96 mmol, 1 eq) in DCM (30 mL) was added DIPEA (1.29 g, 9.96 mmol, 1.73 mL, 1 eq) and the reaction was stirred for 30 min at ambient temperature. Then, BOPCl (2.79 g, 10.95 mmol, 1.1 eq), benzyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (2.84 g, 10.95 mmol, 1.1 eq, HCl), DIPEA (2.57 g, 19.91 mmol, 3.47 mL, 2 eq) were added sequentially. The resulting mixture was stirred at ambient temperature until the consumption of starting material was observed (monitored by LCMS), the reaction was diluted with H2O (30 mL) and extracted with DCM (3×30 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, then filtered and concentrated. The residue was purified by column chromatography (SiO2, 0-50% EtOAc in pet. ether). The resulting product was further purified by trituration with petroleum ether (15 mL) to give benzyl (2S,4R)-1-(1-chlorocyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate (2.1 g, 6.45 mmol, 64.75%) as a white solid. LCMS (ESI+): m/z 326.1 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.39-7.31 (m, 5H), 5.39-5.10 (m, 3H), 4.70 (t, J=8.6 Hz, 1H), 4.41-4.21 (m, 1H), 4.09-3.85 (m, 1H), 2.75-2.51 (m, 1H), 2.25-1.96 (m, 1H), 1.70-1.49 (m, 2H), 1.39-1.26 (m, 2H)


Step 3: Int. 415



embedded image


To a stirred mixture of 10% Pd/C (620.69 mg, 583.25 μmol, 0.1 eq) in EtOAc (50 mL) was added benzyl (2S,4R)-1-(1-chlorocyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate (1.9 g, 5.83 mmol, 1 eq) at 20° C. under N2 atmosphere. The atmosphere was exchanged with H2, and the resulting mixture was stirred under H2 (50 Psi) at 20° C. until the complete consumption of starting material (monitored by LCMS). The reaction mixture was filtered over celite, and the filter cake rinsed with EtOAc (150 mL). The filtrate was concentrated under reduced pressure to give Int. 415 (1.25 g, 5.27 mmol, 90.39%) as a pale yellow solid. LCMS (ESI+): m/z 236.0 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ8.84 (s, 1H), 5.40-5.27 (d, J=52 Hz 1H), 4.69 (t, J=8.6 Hz, 1H), 4.37 (ddd, J=1.9, 13.1, 19.6 Hz, 1H), 4.04-3.82 (m, 1H), 2.74-2.53 (m, 1H), 2.48-2.19 (m, 1H), 1.70-1.55 (m, 1H), 1.39-1.18 (m, 3H)


Int. 416 and 417: Preparation of (2S,4R)-1-((R)-2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid and (2S,4R)-1-((S)-2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


Step 1: benzyl (2S,4R)-1-(2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate



embedded image


To a solution of 2,2-difluoro-1-methylcyclopropane-1-carboxylic acid (3 g, 22.04 mmol, 1 eq) in DMF (80 mL) was added DIEPA (8.55 g, 66.13 mmol, 11.52 mL, 3 eq), benzyl (2S,4R)-4-fluoropyrrolidine-2-carboxylate (6.30 g, 24.25 mmol, 1.1 eq, HCl) and HATU (16.76 g, 44.09 mmol, 2 eq) sequentially at 0° C. under a N2 atmosphere. The mixture was stirred at 25° C. for 12 h. Water (30 mL) was added, and the aqueous phase was extracted with EtOAc (100 mL×3). The combined organic layers were dried with anhydrous Na2SO4 and concentrated. The resulting residue was purified by flash chromatography (SiO2, 0-30% EtOAc in Pet. ether). This resulted in benzyl (2S,4R)-1-(2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate (7 g, 20.51 mmol, 93.04%) as a yellow oil. LCMS (ESI+): m/z 342.3 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.40-7.29 (m, 5H), 5.37-5.09 (m, 3H), 4.78-4.64 (m, 1H), 4.06-3.73 (m, 2H), 2.73-2.56 (m, 1H), 2.22-2.05 (m, 1H), 2.02-1.77 (m, 1H), 1.52-1.41 (m, 3H), 1.47-1.45 (m, 1H).


Step 2: (2S,4R)-1-(2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid



embedded image


To a solution of benzyl (2S,4R)-1-(2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylate (3 g, 8.79 mmol, 1 eq) in MeOH (50 mL) was added Pd/C (1.8 g, 1.69 mmol, 10% wt. %, 0.19 eq). The mixture was stirred under H2 (50 psi) for 2 h The reaction solution was filtered over celite and concentrated under reduced pressure. (2S,4R)-1-(2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2.2 g, 8.50 mmol, 96.65%) was obtained as a white solid. 1H NMR (400 MHz, CDCl3-d): δ6.53 (s, 1H), 5.38-5.25 (d, J=52 Hz, 1H), 4.69 (q, J=8.8 Hz, 1H), 4.11-3.92 (m, 1H), 3.90-3.69 (m, 1H), 2.76-2.57 (m, 1H), 2.54-2.19 (m, 1H), 2.07-1.80 (m, 1H), 1.49 (d, J=1.1 Hz, 3H), 1.39-1.24 (m, 1H).


Step 3: Int. 416, 417



embedded image


The (2S,4R)-1-(2,2-difluoro-1-methylcyclopropane-1-carbonyl)-4-fluoropyrrolidine-2-carboxylic acid was purified by prep-HPLC (Agela DuraShell C18 250×70 mm×10 um; mobile phase: [H2O (10 mM NH4HCO3)/MeCN]; gradient: 0%-10% B over 17.0 min to afford Int. 416 and Int. 417.


Int. 417 (1.5 g, 5.97 mmol, 50.95%) as a white solid. LCMS (ESI+): m/z 250.1 (M−H+) 1H NMR (400 MHz, DMSO-d6): δ 12.69 (br s, 1H), 5.52-5.31 (d, J=52 Hz, 1H), 4.33 (t, J=8.4 Hz, 1H), 4.02-3.88 (m, 1H), 3.78-3.60 (m, 1H), 2.64-2.53 (m, 1H), 2.25-2.04 (m, 1H), 1.70-1.57 (m, 2H), 1.37 (s, 3H).


Int. 416 (1.3 g, 5.18 mmol, 44.16%) as a white solid. LCMS (ESI+): m/z 250.1 (M−H+), 1H NMR (400 MHz, DMSO-d6): δ 5.44-5.31 (d, J=52 Hz, 1H), 4.34 (t, J=8.4 Hz, 1H), 3.92-3.69 (m, 2H), 2.66-2.53 (m, 1H), 2.21-2.01 (m, 1H), 1.90-1.79 (m, 1H), 1.63-1.52 (m, 1H), 1.40 (s, 3H)


Int. 419: Preparation of 2-(3-fluoro-3-methylazetidin-1-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine



embedded image


Step 1: Int. 419



embedded image


A mixture of 2-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (3 g, 13.45 mmol, 1 eq), 3-fluoro-3-methylazetidine hydrochloride (1.69 g, 13.45 mmol, 1 eq), DIPEA (5.21 g, 40.35 mmol, 7.03 mL, 3 eq) in DMSO (50 mL), was heated to 100° C. under an atmosphere of nitrogen. The reaction continued until the complete consumption of starting material (monitored by LCMS). The reaction mixture was cooled to room temperature and diluted with water (50 mL), extracted with EtOAc (2×50 mL). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated. A second batch of this reaction (using the above conditions and scale) was added at this point. The combined residues were triturated with MTBE (40 mL) and filtered. The filter cake was dried to give Int. 419 (2.29 g, 7.84 mmol, 29.14%) as a yellow solid. LCMS (ESI+): m/z 211.1 (M−80+H+). 1H NMR (400 MHz, MeOD-d4): δ8.33 (s, 1H), 7.82 (dd, J=1.7, 8.4 Hz, 1H), 6.43 (d, J=8.4 Hz, 1H), 4.24-4.00 (m, 4H), 1.76-1.60 (d, J=21 Hz, 3H), 1.33 (s, 12H)


Int. 421: Preparation of 2-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,4-tetrahydroisoquinoline



embedded image


Step 1: 7-bromo-2-methyl-1,2,3,4-tetrahydroisoquinoline



embedded image


To a solution of 7-bromo-1,2,3,4-tetrahydroisoquinoline (2.5 g, 11.79 mmol, 1 eq) in MeOH (50 mL) was added H2CO (37% in H2O, 4.78 g, 58.94 mmol, 4.39 mL, 5 eq) and the suspension was stirred at 25° C. for 1 h. Then, the reaction was cooled to 0° C. and NaBH(OAc)3 (3.00 g, 14.15 mmol, 1.2 eq) was added. The reaction was warmed to ambient temperature and continued until the complete consumption of starting material (monitored by LCMS). The reaction mixture was concentrated under reduced pressure to remove MeOH, and diluted with water (20 mL), extracted with MTBE (25 mL). The aqueous phase was then adjusted to pH 7 with saturated Na2HCO3, extracted with EtOAc (3×20 mL), the combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 7-bromo-2-methyl-1,2,3,4-tetrahydroisoquinoline (4.5 g, 18.51 mmol, 78.51%) as a colorless liquid without purification. The product was used with no further purification. LCMS (ESI+): m/z 226.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.30-7.32 (m, 1H), 7.20 (s, 1H), 7.01 (d, J=8.2 Hz, 1H), 3.58 (s, 2H), 2.93-2.87 (t, 2H), 2.76-2.69 (t, 2H), 2.49 (s, 3H)


Step 2: Int. 421



embedded image


To a solution of 7-bromo-2-methyl-1,2,3,4-tetrahydroisoquinoline (4.5 g, 19.90 mmol, 1 eq) in dioxane (50 mL) was added B2Pin2 (7.58 g, 29.85 mmol, 1.5 eq) and KOAc (4.88 g, 49.75 mmol, 2.5 eq) under nitrogen atmosphere. The suspension was degassed with N2 (15 min) and Pd(dppf)Cl2·CH2Cl2 (1.63 g, 1.99 mmol, 0.1 eq) was added. The reaction mixture was heated to 90° C. until the complete consumption of starting material (monitored by LCMS). The reaction mixture was cooled to ambient temperature and concentrated. The crude residue was diluted with sat. NaHCO3 (30 mL), extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The resulting crude product was purified by flash chromatography (SiO2, 0-6% MeOH in DCM) to give Int. 421 (2.1 g, 7.10 mmol, 35.69%) was obtained as a black solid. LCMS (ESI+): m/z 274.1 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.57 (d, J=7.6 Hz, 1H), 7.48 (s, 1H), 7.12 (d, J=7.5 Hz, 1H), 3.60 (s, 2H), 2.94 (t, J=5.8 Hz, 2H), 2.73-2.66 (m, 2H), 2.45 (d, J=1.1 Hz, 3H), 1.34 (s, 12H).


Int. 424: Preparation of 2-methyl-6-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-2,6-diazaspiro[3.4]octane



embedded image


Step 1: tert-butyl 6-(4-chlorophenyl)-2,6-diazaspiro[3.4]octane-2-carboxylate



embedded image


To a stirred mixture of 4-chloroiodobenzene (5 g, 20.969 mmol, 1 eq) and tert-butyl 2,6-diazaspiro[3.4]octane-2-carboxylate (6.68 g, 31.454 mmol, 1.5 eq) in dioxane (50 mL) were added Pd2(dba)3 (2.17 g, 2.097 mmol, 0.1 eq), XPhos (2.00 g, 4.194 mmol, 0.2 eq) and Cs2CO3 (20.50 g, 62.907 mmol, 3 eq) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for additional overnight at 80° C. The resulting mixture was extracted with EtOAc (2×100 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified RP-HPLC (C18, 10-100% MeCN in water with 0.1% FA). Tert-butyl 6-(4-chlorophenyl)-2,6-diazaspiro[3.4]octane-2-carboxylate (4.6 g, 67.95%) was recovered as a brown solid. LCMS (ESI+): m/z 323.1 (M+H+).


Step 2-3 6-(4-chlorophenyl)-2-methyl-2,6-diazaspiro[3.4]octane



embedded image


A mixture of tert-butyl 6-(4-chlorophenyl)-2,6-diazaspiro[3.4]octane-2-carboxylate (1 g, 3.098 mmol, 1 eq) and TFA (3 mL) in DCM (15 mL) was stirred overnight at room temperature. The resulting mixture was concentrated to afford resulted 6-(4-chlorophenyl)-2,6-diazaspiro[3.4]octane (530 mg, 76.82%) as a brown oil. The product was used in the next step without further purification. LCMS (ESI+): m/z 223.15 (M+H+).


To a stirred mixture of 6-(4-chlorophenyl)-2,6-diazaspiro[3.4]octane (700 mg, 3.143 mmol, 1 eq) and DIEA (812.44 mg, 6.29 mmol, 2 eq) in 1:1 MeOH:THF (4 mL) were added H2CO (0.86 mL, 9.42 mmol, 3 eq, 40% in water) and NaBH3CN (592.5 mg, 9.43 mmol, 3 eq) in portions at room temperature under an atmosphere of nitrogen. The resulting mixture was stirred for an additional 2 h at room temperature. The reaction was directly purified by RP-HPLC (C18, 10-100% MeCN in water with 0.1% FA) to afford 6-(4-chlorophenyl)-2-methyl-2,6-diazaspiro[3.4]octane (150 mg, 20.16%) an oil. LCMS (ESI+): m/z 237.15 (M+H+).


Step 4: Int. 424



embedded image


To a stirred mixture of 6-(4-chlorophenyl)-2-methyl-2,6-diazaspiro[3.4]octane (1.2 g, 5.069 mmol, 1 eq) and bis(pinacolato)diboron (1.93 g, 7.604 mmol, 1.5 eq) in dioxane (20 mL) were added Pd2(dba)3 (0.52 g, 0.507 mmol, 0.1 eq), XPhos (0.48 g, 1.014 mmol, 0.2 eq) and KOAc (1.49 g, 15.207 mmol, 3 eq) under nitrogen. The reaction was heated to 100° C. until consumption of starting material was observed (LCMS). The resulting mixture was extracted with EtOAc (2×100 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Preparative TLC (CH2Cl2/MeOH 10:1) to afford Int. 424 (0.2993 g, 15.00%) as a yellow green solid. LCMS (ESI+): m/z 329.15 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 7.46 (d, J=8.1 Hz, 1H), 7.23-7.05 (m, 1H), 6.48 (t, J=8.9 Hz, 2H), 3.35 (s, 2H), 3.28-3.09 (m, 6H), 2.27 (d, J=2.5 Hz, 3H), 1.16 (d, J=71.8 Hz, 12H).


Int. 425: Preparation of (S)-1,2-dimethyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperazine



embedded image


Step 1: (S)-4-(4-bromophenyl)-1,2-dimethylpiperazine



embedded image


To a solution of (S)-1,2-dimethylpiperazine (3 g, 26.27 mmol, 1 eq) and (4-bromophenyl)boronic acid (7.91 g, 39.41 mmol, 1.5 eq) in CH2Cl2 (100 mL) was added Cu(OAc)2 (954.39 mg, 5.25 mmol, 0.2 eq), Et3N (10.63 g, 105.09 mmol, 14.63 mL, 4 eq), and 4 Å molecular sieves (1 g). The mixture was (C18, 10-100% MeCN in water with 0.1% FA). SiO2, 0-100% EtOAc in pet. ether (C18, 10-100% MeCN in water with 0.1% FA). SiO2, 0-100% EtOAc in pet. ether (C18, 10-100% MeCN in water with 0.1% FA). stirred at ambient temperature under an atmosphere of oxygen (15 psi). The reaction continued until the consumption of starting materials (monitored by LCMS). The resulting suspension was filtered through a pad of silica gel and the filter cake was washed with EtOAc (20 mL×3). The combined filtrates were concentrated to dryness and the residue was purified by flash silica gel chromatography (SiO2, 0-100% EtOAc in pet. ether). (S)-4-(4-bromophenyl)-1,2-dimethylpiperazine (3 g, 11.14 mmol, 42.42%) was obtained as a gray solid. LCMS: (ESI+): m/z 269.3 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.33 (d, J=8.8 Hz, 2H), 6.77 (d, J=8.8 Hz, 2H), 3.51-3.36 (m, 2H), 3.01-2.87 (m, 2H), 2.62-2.43 (m, 2H), 2.42-2.29 (m, 4H), 1.17 (d, J=6.4 Hz, 3H).


Step 2: Int. 425



embedded image


A mixture of (S)-4-(4-bromophenyl)-1,2-dimethylpiperazine (2.9 g, 10.77 mmol, 1 eq), B2Pin2 (3.28 g, 12.93 mmol, 1.2 eq), KOAc (2.64 g, 26.93 mmol, 2.5 eq) and [2-(2-aminophenyl)phenyl]-chloro-palladium; bis(1-adamantyl)-butyl-phosphane (720.34 mg, 1.08 mmol, 0.1 eq) in dioxane (50 mL) was degassed with N2 for 20 min. The reaction was heated to 95° C. for 4 h. The suspension was filtered through a pad of silica gel and the filter cake was washed with EtOAc (20 mL×3). The combined filtrates were concentrated to dryness and the residue was purified by flash silica gel chromatography (SiO2, 0-100% EtOAc in Pet. ether). Int. 425 (2 g, 6.32 mmol, 58.70%) was obtained as a gray solid. LCMS: (ESI+): m/z 317.2 (M+H+), 1H NMR (400 MHz, CDCl3-d) δ7.71 (d, J=8.4 Hz, 2H), 6.89 (d, J=8.4 Hz, 2H), 3.69-3.50 (m, 2H), 3.02-2.87 (m, 2H), 2.65-2.55 (m, 1H), 2.41 (dt, J=3.2, 11.4 Hz, 1H), 2.37-2.32 (m, 3H), 2.27 (ddd, J=3.2, 6.4, 9.7 Hz, 1H), 1.33 (s, 12H), 1.15 (d, J=6.4 Hz, 3H).


Int. 426: Preparation of (S)—N,N-dimethyl-1-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methanamine
Step 1: (R)-(4-(4-bromophenyl)morpholin-2-yl)methanol



embedded image


Step 1 was performed using the procedure outline in step 1 for the synthesis of Int. 425 using (R)-morpholin-2-ylmethanol as starting material. LCMS (ESI+): m/z 272.0 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.42-7.34 (m, 2H), 6.83 (d, J=8.6 Hz, 2H), 4.12-4.03 (m, 1H), 3.95-3.63 (m, 4H), 3.46-3.34 (m, 2H), 2.86 (dt, J=3.4, 11.9 Hz, 1H), 2.69 (t, J=11.1 Hz, 1H), 1.93 (s, 0.5H), 1.51 (s, 0.5H)


Step 2: (R)-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methanol



embedded image


To a solution of (R)-(4-(4-bromophenyl)morpholin-2-yl)methanol (2.5 g, 9.19 mmol, 1 eq), bis(pinacolato)diboron (3.50 g, 13.78 mmol, 1.5 eq), KOAc (2.70 g, 27.56 mmol, 3 eq) in dioxane (25 mL) was degassed with N2 for 10 min. Pd(dppf)Cl2 (750.21 mg, 918.65 μmol, 0.1 eq) was added to the reaction. The mixture was heated to 100° C. until the complete consumption of starting material (monitored by LCMS). The reaction mixture was diluted with water (50 mL), extracted with EtOAc (2×50 mL). The combined organics were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (SiO2, 0-40% EtOAc in pet. ether) to give (R)-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methanol (1.7 g, 5.06 mmol, 55.08%) as a yellow solid. LCMS (ESI+): m/z 320.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.74 (d, J=8.6 Hz, 2H), 6.94 (d, J=7.1 Hz, 2H), 4.11-4.04 (m, 1H), 3.95-3.65 (m, 4H), 3.60-3.50 (m, 2H), 2.99-2.69 (m, 2H), 2.04-1.91 (m, 1H), 1.34 (s, 12H).


Step 3: (R)-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methyl methanesulfonate



embedded image


To a solution of (R)-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methanol (1.6 g, 5.01 mmol, 1 eq) in dichloromethane (80 mL) was added DIPEA (3.24 g, 25.06 mmol, 4.37 mL, 5 eq) at 0° C. Then MsCl (918.70 mg, 8.02 mmol, 620.74 μL, 1.6 eq) was added to the reaction dropwise under a N2 atmosphere at 0° C. The reaction was warmed to ambient temperature and stirred until the consumption of starting material (monitored by LCMS). The reaction mixture was cooled, diluted with water (50 mL), and extracted with EtOAc (2×50 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give (R)-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methyl methanesulfonate (2 g, 4.53 mmol, 90.39%) as a yellow oil. LCMS (ESI+): m/z 398.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.74 (d, J=8.6 Hz, 2H), 6.89 (d, J=8.6 Hz, 2H), 4.37-4.30 (m, 2H), 4.10-4.04 (m, 1H), 4.01-3.91 (m, 1H), 3.81 (dt, J=2.7, 11.5 Hz, 1H), 3.64-3.48 (m, 2H), 3.10 (s, 3H), 2.91 (dt, J=3.4, 11.9 Hz, 1H), 2.77-2.68 (m, 1H), 1.34 (s, 12H)


Step 4: Int. 426



embedded image


A solution of (R)-(4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholin-2-yl)methyl methanesulfonate (2 g, 5.03 mmol, 1 eq) in dimethylamine (2 M in THF, 42.79 mL, 17 eq) was added to 100 mL Teflon tank. The reaction was stirred at 75° C. for 12 h. The reaction mixture was diluted with water (50 mL), extracted with EtOAc (2×50 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (heptane/EtOH with 0.1% iso-propylamine) to give Int. 426 (0.85 g, 2.30 mmol, 45.68%) as a white solid. LCMS (ESI+): m/z 347.2 (M+H+). 1H NMR (400 MHz, MeOD-d4): δ7.62 (d, J=8.6 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 4.01 (td, J=1.8, 9.8 Hz, 1H), 3.81-3.68 (m, 2H), 3.67-3.52 (m, 2H), 2.79 (dt, J=3.4, 11.9 Hz, 1H), 2.59-2.40 (m, 3H), 2.31 (s, 6H), 1.32 (s, 12H).


Int. 427 and Int. 428: Preparation of (R)-1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine and (S)-1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine



embedded image


Step 1: tert-butyl 5-(4-bromophenyl)-3,6-dihydropyridine-1 (2H)-carboxylate



embedded image


To a solution of tert-butyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-1 (2H)-carboxylate (10.0 g, 32.34 mmol, 1 eq) in dioxane (200 mL) and H2O (35 mL) was added Pd(dppf)Cl2 (236.64 mg, 323.41 μmol, 0.01 eq) and Na2CO3 (6.86 g, 64.68 mmol, 2 eq) and 1-bromo-4-iodo-benzene (9.15 g, 32.34 mmol, 1 eq). The mixture was stirred at 80° C. for 1 h. The reaction mixture was diluted with water (100 mL) and extracted with EtOAc (80 mL×3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Pet. ether/EtOAc 0-15%). tert-butyl 5-(4-bromophenyl)-3,6-dihydropyridine-1 (2H)-carboxylate (14 g, 41.39 mmol, 63.99%) was obtained as a clear oil. LCMS (ESI+): m/z 338.1 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.46 (d, J=8.4 Hz, 2H), 7.24 (d, J=8.4 Hz, 2H), 6.20 (td, J=2.0, 4.0 Hz, 1H), 4.23 (s, 2H), 3.55 (t, J=6.0 Hz, 2H), 2.31 (d, J=3.6 Hz, 2H), 1.50 (s, 9H).


Step 2: tert-butyl 3-(4-bromophenyl)piperidine-1-carboxylate



embedded image


To a suspension of PtO2 (0.5 g) of in EtOAc (80 mL) was added and tert-butyl 5-(4-bromophenyl)-3,6-dihydropyridine-1 (2H)-carboxylate (5 g, 14.78 mmol, 1 eq) and the mixture was stirred at 20° C. for 2 h under H2 atmosphere. The reaction mixture was filtered through a pad of silica gel and the filter cake was washed with EtOAc (50 mL×3). The combined filtrates were concentrated to dryness and the residue was purified by column chromatography (SiO2, EtOAc:PE 0-50%) to afford tert-butyl 3-(4-bromophenyl)piperidine-1-carboxylate (5 g, 14.69 mmol, 99.41%) as a white oil. LCMS (ESI+): m/z 340.0 (M+H+) 1H NMR (400 MHz, CDCl3-d): δ7.44 (d, J=8.4 Hz, 2H), 7.11 (d, J=8.4 Hz, 2H), 4.20-4.12 (m, 2H), 2.73 (d, J=11.2 Hz, 2H), 2.00 (d, J=9.2 Hz, 1H), 1.80-1.71 (m, 1H), 1.66-1.54 (m, 3H), 1.47 (s, 9H).


Step 3: tert-butyl 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine-1-carboxylate



embedded image


To a solution of tert-butyl 3-(4-bromophenyl)piperidine-1-carboxylate (2 g, 5.88 mmol, 1 eq) in dioxane (40 mL) was added B2Pin2 (2.24 g, 8.82 mmol, 1.5 eq), KOAc (1.15 g, 11.76 mmol, 2 eq) and Pd(dppf)Cl2 (430.10 mg, 587.80 μmol, 0.1 eq). The solution was degassed with a stream of N2 and heated to 100° C. for 1 h. Upon the consumption of the starting material (monitored by LCMS) the reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL×3), the combined organic layers were dried over Na2SO4, filtered, and concentrated. The resulting residue was purified by column chromatography (SiO2, 0 to 10% EtOAc in pet. ether) to afford tert-butyl 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine-1-carboxylate (2.2 g, 5.68 mmol, 96.63%) as a clear oil. LCMS (ESI+): m/z 388.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.77 (d, J=7.6 Hz, 2H), 7.25 (d, J=8.0 Hz, 2H), 4.24-4.09 (m, 2H), 2.81-2.64 (m, 3H), 1.79-1.55 (m, 4H), 1.34 (s, 9H), 1.27 (s, 12H).


Step 4: 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine



embedded image


To a solution of tert-butyl 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine-1-carboxylate (1.05 g, 2.71 mmol, 1 eq) in DCM (20 mL) was added TFA (4.2 mL). The mixture was stirred at 25° C. for 1 h The solution was basified to pH=7 with NaHCO3 (1 mol/L), and extracted with EtOAc (10 mL×3), dried over Na2SO4, filtered, and concentrated under reduced pressure to give 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine (1.7 g, crude) as a white solid. The material was used directly in the next reaction. LCMS (ESI+): m/z 288.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.78 (d, J=8.0 Hz, 2H), 7.20 (d, J=8.0 Hz, 2H), 3.43 (d, J=12.4 Hz, 2H), 3.16-3.04 (m, 1H), 2.96-2.82 (m, 2H), 2.14-1.95 (m, 3H), 1.76-1.62 (m, 1H), 1.34 (s, 12H).


Step 5: 1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine



embedded image


To a solution of 3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine (1 g, 3.48 mmol, 1 eq) in MeOH (10 mL) was added a 37% formaldehyde solution (1.41 g, 17.41 mmol, 1.30 mL, 5 eq) and AcOH (20.91 mg, 348.18 mol, 19.93 μL, 0.1 eq) at 0° C. The reaction was stirred for 30 minutes, then, NaBH3CN (656.42 mg, 10.45 mmol, 3 eq) was added. The reaction continued for an additional 30 min. The reaction mixture was then diluted with water (20 mL) and extracted with EtOAc (15 mL×3), the combined organic layers were dried over Na2SO4, filtered, and concentrated. The residue was triturated with MTBE (10 mL) to give 1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine (0.2 g, 663 mol, 19.0%) as a white solid. LCMS (ESI+): m/z 302.4 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.79 (d, J=8.0 Hz, 2H), 7.25 (d, J=8.0 Hz, 2H), 3.56-3.39 (m, 2H), 3.30-3.16 (m, 1H), 2.89 (t, J=12.4 Hz, 2H), 2.84 (s, 3H), 2.20-2.02 (m, 3H), 1.78-1.64 (m, 1H), 1.33 (s, 12H).


Step 6: Int. 427, Int. 428



embedded image


The 1-methyl-3-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine was further separated by SFC (condition: column: Daicel Chiralcel OZ 250×50 mm; 10 m; mobile phase: [CO2-MeOH (0.1% IPAm)]; B %: 25%, isocratic elution) to afford (R)-stereochemistry Int. 428 (0.5 g, 1.56 mmol, 23.50%), LCMS (ESI+): m/z 302.4 (M+H+) 1H NMR (400 MHz, CDCl3-d): 67.76 (d, J=7.6 Hz, 2H), 7.25 (d, J=7.6 Hz, 2H), 3.01-2.81 (m, 3H), 2.31 (s, 3H), 2.02-1.89 (m, 3H), 1.83-1.68 (m, 2H), 1.43 (dd, J=4.4, 12.4 Hz, 1H), 1.34 (s, 12H), and (S)-stereochemistry Int. 427 (0.46 g, 1.39 mmol, 20.93%) as white solid. LCMS (ESI+): m/z 302.4 (M+H+) 1H NMR (400 MHz, CDCl3-d): δ7.77 (d, J=7.6 Hz, 2H), 7.24 (d, J=7.6 Hz, 2H), 3.15 (d, J=11.6 Hz, 2H), 3.03 (tt, J=3.4, 12.0 Hz, 1H), 2.49 (s, 3H), 2.32-2.20 (m, 2H), 2.02-1.87 (m, 3H), 1.57-1.48 (m, 1H), 1.34 (s, 12H).


Int. 429: Preparation of 1-(2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-4-isopropylpiperazine



embedded image


Step 1: 1-(4-bromo-2-fluorophenyl)-4-isopropylpiperazine



embedded image


A mixture of (4-bromo-2-fluorophenyl)boronic acid (4 g, 13.29 mmol, 1 eq), 1-isopropylpiperazine (1.70 g, 13.29 mmol, 1.90 mL, 1 eq), Pd2(dba)3 (304.33 mg, 332.34 mol, 0.025 eq), Xantphos (769.20 mg, 1.33 mmol, 0.1 eq) and NaOt-Bu (2.56 g, 26.59 mmol, 2 eq) in Tol. (80 mL) was degassed and purged with N2 (20 min). The mixture was stirred at 80° C. for 1 h. The reaction mixture was concentrated to give a residue and the residue was purified by flash chromatography (SiO2, 30-35% EtOAc in pet. ether) to give 1-(4-bromo-2-fluorophenyl)-4-isopropylpiperazine (2.8 g, 7.41 mmol, 55.74%) as a brown oil. LCMS (ESI+): m/z 301.0 (M+H+), 1H NMR (CDCl3-d, 400 MHz): δ 7.15-7.22 (m, 2H), 6.82 (t, J=8.8 Hz, 1H), 3.07-3.12 (m, 4H), 2.73-2.78 (m, 1H), 2.69-2.72 (m, 4H), 1.10 (d, J=6.4 Hz, 6H).


Step 2: Int. 429



embedded image


A mixture of 1-(4-bromo-2-fluorophenyl)-4-isopropylpiperazine (2.7 g, 8.96 mmol, 1 eq), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (3.41 g, 13.45 mmol, 1.5 eq), KOAc (2.20 g, 22.41 mmol, 2.5 eq), Pd(dppf)Cl2 (327.96 mg, 448.21 μmol, 0.05 eq) in dioxane (50 mL) was degassed with N2 for 20 min. The mixture was heated to 100° C. for 5 h. The reaction mixture was concentrated and the resulting residue was purified by flash chromatography (SiO2, 0-5% MeOH in EtOAc) to afford Int. 429. The product further purified by salt formation with HCl in EtOAc. Int. 429 (2.3 g, 5.86 mmol, 65.32%, HCl) as a white solid. LCMS: (ESI+): m/z 349.2 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 7.44 (dd, J=7.6, 1.2 Hz, 1H), 7.31 (dd, J=13.6, 1.2 Hz, 1H), 7.09 (t, J=8.4 Hz, 1H), 3.58 (br d, J=12.8 Hz, 2H), 3.47 (d, J=11.2 Hz, 3H), 3.32 (t, J=11.6 Hz, 2H), 3.10-3.23 (m, 2H), 1.32 (d, J=6.4 Hz, 6H), 1.27 (s, 12H).


Int. 430: Preparation of ((R)-2-methyl-4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)morpholine



embedded image


Step 1: (R)-4-(4-bromophenyl)-2-methylmorpholine



embedded image


A mixture of (R)-2-methylmorpholine (3 g, 29.66 mmol, 1 eq), 1-bromo-4-iodo-benzene (10.91 g, 38.56 mmol, 1.3 eq), NaOtBu (8.55 g, 88.98 mmol, 3 eq), Xantphos (2.15 g, 3.71 mmol, 0.125 eq) and Pd2(dba)3 (1.36 g, 1.48 mmol, 0.05 eq) in toluene (50 mL) was degassed with N2 for 10 min, then heated to 85° C. for 1 h. After cooling to room temperature, the reaction mixture was concentrated. The resulting residue was diluted with water (50 mL) and extracted with EtOAc (80 mL×3). The combined organic layers were dried over Na2SO4, filtered, and concentrated. The residue was purified by flash chromatography (SiO2, 0-5% EtOAc in Pet. ether) to give (R)-4-(4-bromophenyl)-2-methylmorpholine (7 g, 23.27 mmol, 78.44%) as a yellow solid. LCMS: (ESI+): m/z 256.0 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 7.35 (d, J=9.2 Hz, 2H), 6.89 (d, J=9.2 Hz, 2H), 3.89 (dd, J=11.6, 2.4 Hz, 1H), 3.54-3.64 (m, 3H), 3.46 (br d, J=11.6 Hz, 1H), 2.61 (td, J=11.6, 3.6 Hz, 1H), 2.29 (t, J=10.8 Hz, 1H), 1.14 (d, J=6.4 Hz, 3H).


Step 2: Int. 430



embedded image


A mixture of (R)-4-(4-bromophenyl)-2-methylmorpholine (4 g, 15.62 mmol, 1 eq), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (7.93 g, 31.23 mmol, 2 eq), KOAc (3.83 g, 39.04 mmol, 2.5 eq), cataCXium A Pd G2 (1.04 g, 1.56 mmol, 0.1 eq) in dioxane (50 mL) was degassed with N2 for 15 min. The reaction was heated to 90° C. for 2 h. After cooling to room temperature, the reaction mixture was concentrated. EtOAc (30 mL) was added, solids were filtered off, and the filtrate was concentrated under reduced pressure. The residue was purified by flash chromatography (SiO2, 0-5% EtOAc in Pet. ether) to give Int. 430 (2.06 g, 6.20 mmol, 39.68%) as a gray solid. LCMS: (ESI+): m/z 304.2 (M+H+), 1H NMR (400 MHz, DMSO-d6): δ 7.52 (d, J=8.4 Hz, 2H), 6.90 (d, J=8.4 Hz, 2H), 3.90 (d, J=10.8 Hz, 1H), 3.54-3.69 (m, 4H), 2.66 (td, J=12.0, 3.2 Hz, 1H), 2.34 (t, J=10.8 Hz, 1H), 1.26 (s, 12H), 1.15 (d, J=6.4 Hz, 3H).


Int. 431: Preparation of (S)-1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine



embedded image


Step 1: 2-(4-bromophenyl)-1-methylpiperidine



embedded image


To a solution of 2-(4-bromophenyl)piperidine (2 g, 8.32 mmol, 1.0 eq) in MeOH (40 mL) was added 37% formaldehyde (3.38 g, 41.64 mmol, 3.10 mL, 5 eq) and AcOH (50.02 mg, 832.86 μmol, 47.68 μL, 0.1 eq) and stirred at 0° C. for 30 min. Then, NaBH3CN (1.57 g, 24.98 mmol, 3 eq) was added and stirred at 0° C. for 30 min. Upon the consumption of starting material, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (40 mL×3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure to give 2-(4-bromophenyl)-1-methylpiperidine (2.2 g, crude) as a yellow oil. The product was used without further purification. LCMS (ESI+): m/z 254.2 (M+H+). 1H NMR (400 MHz, CDCl3-d): δ7.48 (d, J=8.4 Hz, 2H), 7.22 (d, J=8.4 Hz, 2H), 3.20-3.10 (m, 1H), 2.92 (dd, J=2.8, 11.2 Hz, 1H), 2.32-2.22 (m, 1H), 2.08 (s, 3H), 1.91-1.84 (m, 1H), 1.80-1.76 (m, 2H), 1.75-1.70 (m, 1H), 1.70-1.60 (m, 1H), 1.48-1.35 (m, 1H).


Step 2: 1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine



embedded image


To a solution of 2-(4-bromophenyl)-1-methylpiperidine (1.1 g, 4.33 mmol, 1 eq) in dioxane (40 mL) was added B2Pin2 (1.65 g, 6.49 mmol, 1.5 eq), Pd(dppf)Cl2 (316.67 mg, 432.79 μmol, 0.1 eq) and KOAc (849.50 mg, 8.66 mmol, 2 eq). The mixture was degassed with N2 then stirred at 100° C. for 1 h under N2. Then, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (40 mL×3). The combined organic layers were dried over Na2SO4, filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC (C18, H2O+10 mM NH4HCO3 and MeCN) to afford 1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine (117 mg, 388.41 μmol, 8.97%) as a white solid. LCMS (ESI+): m/z 302.1 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 7.62 (d, J=7.6 Hz, 2H), 7.31 (d, J=7.6 Hz, 2H), 2.93 (d, J=11.2 Hz, 1H), 2.76 (d, J=9.2 Hz, 1H), 2.02 (t, J=10.4 Hz, 1H), 1.87 (s, 3H), 1.72 (d, J=12.4 Hz, 1H), 1.67-1.52 (m, 3H), 1.45-1.30 (m, 2H), 1.28 (s, 9H).


Step 3: Int. 431



embedded image


1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidine was further separated by SFC (DAICEL CHIRALPAK AD, CO2/MeOH+0.1% IPAm 10% isocratic) to afford Int. 431 (0.1 g, 331.97). LCMS (ESI+): m/z 302.1 (M+H+). 1H NMR (400 MHz, CDCl3-d) δ7.77 (d, J=8.0 Hz, 2H), 7.34 (d, J=7.6 Hz, 2H), 3.04 (br d, J=11.6 Hz, 1H), 2.78 (dd, J=2.4, 11.2 Hz, 1H), 2.17-2.06 (m, 1H), 2.00 (s, 3H), 1.83-1.68 (m, 4H), 1.64-1.52 (m, 1H), 1.38 (br d, J=4.4 Hz, 1H), 1.34 (s, 12H).


Int. 433: Preparation of 4-bromo-6-chloro-1-methyl-1H-indazole



embedded image


Int. 433 was synthesized using the procedure outlined in Int. 411. LCMS (ESI+): m/z 293.5 (M+H+).


Int. 438: Preparation of (S)-3-(2-bromo-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoic acid



embedded image


Step 1: methyl(S)-3-(2-bromo-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoate



embedded image


To a vial was added zinc (4.8 g, 3 Eq, 73 mmol). The vial was evacuated and backfilled with argon. DMA (10 mL) was added by syringe. Iodine (0.37 g, 0.06 Eq, 1.5 mmol) was added as a solid, and the mixture was stirred until exotherm and color change had subsided. Separately, methyl (R)-2-((tert-butoxycarbonyl)amino)-3-iodopropanoate (18 g, 2.3 Eq, 55 mmol) was dissolved in 10 mL DMA, and this solution was added by syringe to zinc/iodine mixture while stirring, forming the zincate reagent in situ.


Separately, 1-bromo-4-chloro-2-iodobenzene (7.7 g, 1 Eq, 24 mmol) was added to a flask, and the flask was evacuated and backfilled with argon. DMA (10 mL) was added to this flask by syringe. To this solution was added Pd2(dba)3 (0.67 g, 0.03 Eq, 0.73 mmol) and tris-2-furanyl-phosphine (1.0 g, 0.18 Eq, 4.4 mmol) as solids, and the mixture was stirred to dissolve. The zincate solution was added to the reaction flask by syringe while stirring. The reaction was heated to 55° C. and stirred overnight. The reaction was allowed to cool to room temperature, diluted with water, and extracted three times with EtOAc. The combined organics were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. The crude product was purified by silica gel chromatography (0 to 100% EtOAc in hexanes) to yield methyl(S)-3-(2-bromo-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoate (3.93 g, 10.02 mmol, 41%). LCMS (ESI+): m/z 338.1 (M−55+H+).


Step 2: Int. 438



embedded image


Methyl(S)-3-(2-bromo-5-chlorophenyl)-2-((tert-butoxycarbonyl)amino)propanoate (3.93 g, 1 Eq, 10 mmol) was dissolved in THF (4 mL), water (2 mL), and MeOH (1 mL). Lithium hydroxide monohydrate (480 mg, 2 Eq, 20 mmol) was added and the reaction mixture was stirred for 1 hour. The reaction was stirred for 1 hour, acidified with 1M HCl, and extracted three times with EtOAc. The combined organics were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. Int. 438 (3.78 g, 10 mmol, quant.) was used without further purification. LCMS (ESI+): m/z 378.2 (M−55+H+).


A. Representative Procedures for Preparing Compounds of Formula (I)

The following subsections describe procedures for preparing compounds of Formula (I). It is understood that compounds of Formula (I) can be prepared using variety of synthetic routes. The following procedures are meant to be illustrative, not limiting. Generally, compounds of Formula (I) are prepared by synthesizing a macrocyclic portion of the compound and completing final synthetic steps to obtain compounds of Formula (I). The final synthetic steps include optional modifications (e.g., reduction, Phe Suzuki coupling, acylation) and tail addition (comprising the R3, R4a/b/c, and sometimes R5a/b/c variable positions of Formula (I)). The macrocyclic synthesis procedures described herein include Methods A, B, C, and D. The final synthesis procedures described herein include Methods 1-14.


1. Macrocycle Synthesis Procedures

a. Method A


Generic Procedure for Method A



embedded image


Representative Procedure for Method A



embedded image


embedded image


Representative Experimental for Method A:
Step 1: (S)-3-(2-bromo-5-fluorophenyl)-2-((tert-butoxycarbonyl)(methyl)amino)propanoic acid (201)



embedded image


To a solution of Int. 31 (32 g, 85 mmol, 1 eq) and Int. 3 (11.5 g, 112 mmol, 1.3 eq) in DMF (1000 mL) was added HATU (48.5 g, 127.5 mmol, 1.5 eq) and DIPEA (55.0 g, 425.3 mmol, 74.1 mL, 5 eq) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 2000 mL, then extracted with EtOAc (2000 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-60%) to yield 201: (37 g, 78.5 mmol, 92.3% yield) as a yellow oil. LCMS (ESI+): m/z 493.1 (M+Na+)


Step 2: (S)-3-(2-bromo-5-fluorophenyl)-N-(hex-5-en-1-yl)-N-methyl-2-(methylamino)propenamide TFA salt (202)



embedded image


To a solution of 201 (37 g, 78.5 mmol, 1 eq) in DCM (400 mL) was added TFA (80 mL) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give 202 (38 g, crude, TFA) as a light-yellow oil. LCMS (ESI+): m/z 371.1 (M+H+)


Step 3: Tert-butyl ((S)-1-(((S)-3-(2-bromo-5-fluorophenyl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)(methyl)amino)-4-methyl-1-oxopentan-2-yl)carbamate (203)



embedded image


To a solution of 202 (38 g, 78.3 mmol, 1 eq, TFA salt) and Int. 151 (19.9 g, 86. mmol, 1.1 eq) in DMF (1000 mL) was added HATU (44.7 g, 117.5 mmol, 1.5 eq) and DIEPA (50.6 g, 391.5 mmol, 68.2 mL, 5 eq) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 2000 mL, then extracted with EtOAc (2000 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes 0-50%) to give 203 (45 g) as a yellow oil. LCMS (ESI+): m/z 606.2 (M+Na+)


Step 4: (S)-2-amino-N—((S)-3-(2-bromo-5-fluorophenyl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)-N,4-dimethylpentanamide TFA salt (204)



embedded image


To a solution of 203 (45 g, 76.9 mmol, 1 eq) in DCM (500 mL) was added TFA (100 mL) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give 204 (46 g, crude) as a light-yellow oil. LCMS (ESI+): m/z 484.1 (M+H+)


Step 5: Tert-butyl ((S)-1-(((S)-1-(((S)-3-(2-bromo-5-fluorophenyl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)(methyl)amino)-4-methyl-1-oxopentan-2-yl)amino)-1-oxopent-4-en-2-yl)(methyl)carbamate (205)



embedded image


To a solution of 204 (46 g, 76.9 mmol, 1 eq, TFA) and Int. 152 (19.38 g, 84.6 mmol, 1.1 eq) in DMF (1300 mL) was added HATU (43.8 g, 115.3 mmol, 1.5 eq) and DIEPA (49.7 g, 384.32 mmol, 66.94 mL, 5 eq) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 2000 mL, then extracted with EtOAc (2000 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-60%) to give 205 (46 g, 66.1 mmol, 78% yield) as a yellow oil. LCMS (ESI+): m/z 717.4 (M+Na+)


Step 6: (S)—N—((S)-1-(((S)-3-(2-bromo-5-fluorophenyl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)(methyl)amino)-4-methyl-1-oxopentan-2-yl)-2-(methylamino)pent-4-enamide TFA salt (206)



embedded image


To a solution of 205 (46 g, 66.1 mmol, 1 eq) in DCM (500 mL) was added TFA (100 mL) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give 206 (47 g) as a yellow oil. LCMS (ESI+): m/z 595.1 (M+H+) Step 7: Tert-butyl((1S,4S,7S,10S)-4-allyl-10-(2-bromo-5-fluorobenzyl)-1-cyclopropyl-7-isobutyl-3,9,12-trimethyl-2,5,8,11-tetraoxo-3,6,9,12-tetraazaoctadec-17-en-1-yl)carbamate (207)




embedded image


To a solution 206 (47 g, 66.2 mmol, 1 eq, TFA) and Int. 153 (16.4 g, 76.2 mmol, 1.15 eq) in DMF (1300 mL) was added HATU (37.8 g, 99.4 mmol, 1.5 eq) and DIEPA (85.6 g, 662.3 mmol, 115.4 mL, 10 eq) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 2000 mL, then extracted with EtOAc (2000 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-60%) to give 207 (44 g, 55.5 mmol, 70% yield) as a yellow oil. LCMS (ESI+): m/z 814.5 (M+Na+). 1H NMR (400 MHz, DMSO-d6) δ 0.34 (br s, 2H) 0.42 (br d, J=7.87 Hz, 2H) 0.85 (br d, J=6.32 Hz, 6H) 1.11 (br d, J=7.15 Hz, 1H) 1.29 (br d, J=6.44 Hz, 3H) 1.38 (s, 9H) 1.45 (br s, 3H) 1.52-1.63 (m, 1H) 2.02 (br d, J=6.68 Hz, 2H) 2.30-2.40 (m, 1H) 2.83 (s, 3H) 2.94 (br s, 2H) 3.00 (s, 3H) 3.03 (br d, J=9.06 Hz, 1H) 3.06 (s, 3H) 3.09-3.24 (m, 2H) 3.26-3.47 (m, 1H) 4.14 (br s, 1H) 4.66-4.76 (m, 1H) 4.81-5.23 (m, 5H) 5.67 (br s, 1H) 5.70-5.85 (m, 2H) 6.21-6.69 (m, 1H) 6.92-7.01 (m, 1H) 7.03-7.15 (m, 1H) 7.54 (br dd, J=8.52, 5.54 Hz, 1H)


Step 8: Tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-bromo-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (208)



embedded image


A mixture the pre-cyclized peptide 207 (8 g, 10.1 mmol, 1 eq), [1,3-bis(2,4,6-trimethylphenyl)imidazolidin-2-ylidene]-dichloro-[(2-isopropoxyphenyl)methylene]ruthenium (695.5 mg, 1.11 mmol, 0.11 eq) in DCE (1600 mL) was degassed and purged with N2 for 0.5 hr. The mixture was capped and stirred at 70° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give a thick residue. The crude product was purified by reversed phase HPLC flash C18 gel chromatography (ISCO; 330 g SepaFlash C18 Flash Column, eluent of 50-100% MeCN in H2O buffered with 0.1% TFA) 208 (5.41 g, 6.8 mmol, 67% yield) as an off white solid. Compound was isolated as a mix of cis/trans isomers LCMS (ESI+): m/z 764.4 (M+H). 1H NMR of major isomer (400 MHz, DMSO-d6) δ 0.25-0.47 (m, 4H) 0.70-0.86 (m, 6H) 1.05-1.13 (m, 1H) 1.29 (br s, 3H) 1.40 (br s, 9H) 1.43-1.62 (m, 3H) 1.78-2.16 (m, 3H) 2.47-2.49 (m, 1H) 2.51-2.56 (m, 1H) 2.60-3.03 (m, 11H) 3.09-3.29 (m, 2H) 4.10-4.32 (m, 2H) 4.53-5.07 (m, 2H) 5.19-5.40 (m, 1H) 5.41-5.58 (m, 1H) 6.25-6.60 (m, 1H) 6.95-7.06 (m, 1H) 7.07-7.16 (m, 1H) 7.53-7.63 (m, 1H) 7.74-7.98 (m, 1H)


b. Method B


Generic Procedure for Method B: Z-Selective Macrocycle Formation



embedded image


Representative Procedure for Method B



embedded image


embedded image


Representative Experimental for Method B:
Step 1: Tert-butyl (S)-(3-(5-chloro-2-cyclopropoxypyridin-3-yl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)(methyl)carbamate (209)



embedded image


To a solution of Int. 7 (7.0 g, 1 Eq, 19 mmol) and Int. 3 (2.8 g, 1.3 Eq, 25 mmol) in DMF (94 mL) was added HATU (9.3 g, 1.3 Eq, 25 mmol) and DIPEA (9.8 g, 13 mL, 4 Eq, 76 mmol) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 400 mL, then extracted with EtOAc (300 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-60%) to yield 209 (8.0 g, 17 mmol, 91% yield) as a yellow oil. LCMS (ESI+): m/z 466.4 (M+H+)


Step 2: (S)-3-(5-chloro-2-cyclopropoxypyridin-3-yl)-N-(hex-5-en-1-yl)-N-methyl-2-(methylamino)propanamide TFA salt (210)



embedded image


To a solution of 209 (8.0 g, 17 mmol, 1 eq) in DCM (80 mL) was added TFA (16 mL) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give 210 (8.8 g, crude, TFA) as a light-yellow oil. LCMS (ESI+): m/z 366.2 (M+H+)


Step 3: Tert-butyl ((S)-1-(((S)-3-(5-chloro-2-cyclopropoxypyridin-3-yl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)(methyl)amino)-4-methyl-1-oxopentan-2-yl)carbamate (211)



embedded image


To a solution of 210 (8.0 g, 17 mmol, 1 eq, TFA) and Int. 151 (5.1 g, 1.2 Eq, 21 mmol) in DMF (86 mL) was added DIPEA (8.9 g, 12 mL, 4 Eq, 69 mmol) and HATU (7.8 g, 1.2 Eq, 21 mmol) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction was added into ice water 400 mL, then extracted with EtOAc (300 mL×2), the combined organic layers were washed with brine, dried over MgSO4 and filtered. The filtrate was concentrated under reduced pressure to give crude product which was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-100%) to yield 211 (9.2 g, 16 mmol, 93% yield) as a yellow oil. LCMS (ESI+): m/z 579.7 (M+H+)


Step 4: Tert-butyl (6S,9S,19S,Z)-19-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)-9-((5-chloro-2-cyclopropoxypyridin-3-yl)methyl)-6-isobutyl-2,2,8,11-tetramethyl-4,7,10-trioxo-3-oxa-5,8,11-triazaicos-16-en-20-oate (212)



embedded image


A mixture of Int. 154 (2.5 g, 1 Eq, 4.3 mmol) 211 (5.3 g, 3 Eq, 13 mmol) in DCE (22 mL) was degassed and purged with Ar for 0.5 hr, and Hoveyda-Grubbs Catalyst® M2001 (0.41 g, 0.15 Eq, 0.65 mmol) was added to the mixture. The mixture was stirred at 70° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give a thick residue. The crude product was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-100%) to give 212 (1.8 g, 1.9 mmol, 44% yield, crude, Trans/Cis=1:4) as brown syrup. LCMS (ESI+): m/z 958.8 (M+H).


Step 5: (S,Z)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)-9-((S)-2-((S)-2-(12-azaneyl)-N,4-dimethylpentanamido)-3-(5-chloro-2-cyclopropoxypyridin-3-yl)-N-methylpropanamido)non-4-enoic acid TFA salt (213)



embedded image


To a solution of peptide 212 (1.8 g, 1.9 mmol, 1 eq) in DCM (15 mL) was added TFA (7 mL) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give 213 (2 g, crude, TFA) as a brown oil. LCMS (ESI+): m/z 858.6 (M+H+)


Step 6: (9H-fluoren-9-yl)methyl((3S,6S,9S,Z)-3-((5-chloro-2-cyclopropoxypyridin-3-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)carbamate (214)



embedded image


To a solution of the deprotected peptide 213 (1.8 g, 1.9 mmol, 1 eq, TFA) in DCM (0.37 L), add DIEPA (0.87 g, 1.2 mL, 3 Eq, 6.7 mmol). Separately, dissolve 1-Hydroxy-7-azabenzotriazole (0.37 g, 0.38 mL, 1.2 Eq, 2.7 mmol) and PyBOP (1.4 g, 1.2 Eq, 2.7 mmol) in DCM (0.37 L) and add DIEPA (0.87 g, 1.2 mL, 3 Eq, 6.7 mmol). Add the second solution into the first solution dropwise by additional funnel. The mixture was stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give a thick residue. The crude product was purified by column chromatography (SiO2, EtOAc:Hexanes; 0-100%) to give 214 (0.5 g, 0.6 mmol, 30% yield, 99% purity) as brown solid. LCMS (ESI+): m/z 784.5 (M+H+)


Step 7: (3S,6S,9S,Z)-3-((5-chloro-2-cyclopropoxypyridin-3-yl)methyl)-6-isobutyl-1,4-dimethyl-9-(methylamino)-1,4,7-triazacyclohexadec-11-ene-2,5,8-trione (215)



embedded image


To peptide 214 (0.5 g, 0.6 mmol, 1 eq) was added dimethylamine in THF (6.76 g, 10.4 mL, 2.0 molar, 32.6 Eq, 20.8 mmol) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The reaction mixture was concentrated under reduced pressure to give a thick residue. The crude product was purified by reversed phase HPLC flash C18 gel chromatography (ISCO; 12 g C18 Flash Column, eluent of 20-100% MeCN in H2O buffered with 0.1% TFA) to give 215 (0.35 g, 0.64 mmol, 100% yield) LCMS (ESI+): m/z 562.5 (M+H+)


Step 8: Tert-butyl ((S)-2-(((3S,6S,9S,Z)-3-((5-chloro-2-cyclopropoxypyridin-3-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (216)



embedded image


To a solution of deprotected starting material 215 (350 mg, 0.64 mmol, 1 eq) and Int. 153 (164 mg, 1.2 Eq, 764 μmol) in DMF (3.18 mL) was added DIPEA (494 mg, 666 μL, 6 Eq, 3.82 mmol) and HATU (291 mg, 1.2 Eq, 764 μmol) at 0° C. The mixture was removed from the cooling bath and stirred at 25° C. until LCMS analysis showed the complete consumption of starting material. The mixture was neutralized to pH˜7 with 1N HCl and directly purified by RP-HPLC (C18, 50% to 80% MeCN/H2O with 0.5% formic acid) to yield 216 (120 mg, 158 μmol, 25%) as off-white solid. LCMS (ESI+): m/z 759.5 (M+H+)


c. Method C


Generic Procedure for Method C



embedded image


As seen when comparing Method A and Method C, Steps 1-5 follow similar synthetic procedures.


Representative Experimental for Method C:
(9H-fluoren-9-yl)methyl((S)-1-(((S)-1-(((S)-3-(1,4-dichloro-115-chlorinin-3-yl)-1-(hex-5-en-1-ylamino)-1-oxopropan-2-yl)(methyl)amino)-4-methyl-1-oxopentan-2-yl)amino)-1-oxopent-4-en-2-yl)carbamate (217)



embedded image


Compound 217 was prepared according to Steps 1-5 of Method A with starting materials Int. 1, 6, 151 and 21. LCMS (ESI+): m/z 761.1 (M+H).


Step 6: (9H-fluoren-9-yl)methyl((3S,6S,9S)-3-(2,5-dichlorobenzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)carbamate (218)



embedded image


217 (200 mg, 1 Eq, 263 μmol) was dissolved in DCE (80 mL) and degassed for 30 min with a stream of nitrogen. Dichloro(1,3-dimesityl-2-imidazolidinylidene)(2-isopropoxybenzylidene)ruthenium (32 mg, 0.19 Eq, 51 μmol) was added rapidly and the reaction was degassed for an additional 10 min. reaction was then heated to 50° C. and continued until the complete consumption of starting material (monitored by LCMS). The reaction was cooled, concentrated, and purified by normal phase purification Methanol/DCM (0 to 15%, over 10 min.). 218 (121 mg, 165 μmol, 63%) was isolated as a brown solid. It was used as a mixture of E/Z isomers (˜7:3). LCMS (ESI+): m/z 733.4 (M+H).


d. Method D


Generic Procedure for Method D



embedded image


Representative Procedure for Method D



embedded image


embedded image


Representative Experimental for Method D:
Step 1: tert-butyl (S)-(3-(2-bromo-5-chlorophenyl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)carbamate (249)



embedded image


Int. 238 (3.73 g, 1 Eq, 10.02 mmol) was dissolved in DMF (10 mL) along with Int. 3 (1.36 g, 1.8 mL, 1.2 Eq, 12.03 mmol), HATU (4.5 g, 1.2 Eq, 12.03 mmol), and DIPEA (4.53 g, 6.11 mL, 3.5 Eq, 35.08 mmol). The pH was checked by indicator paper and DIPEA was added until the pH was confirmed to be basic. The reaction was stirred for 1 hour, diluted with water, and extracted with EtOAc (3×100 mL). The combined organics were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. The crude product was purified by silica gel chromatography (0 to 100% ethyl acetate in hexanes) to yield 249 (1.195 g, 2.52 mmol, 25% yield). LCMS (ESI+): m/z 473.2 (M+H+).


Steps 2-3: (S)-3-(2-bromo-5-chlorophenyl)-2-(ethylamino)-N-(hex-5-en-1-yl)-N-methylpropanamide (250)



embedded image


249 (1.195 g, 1 Eq, 2.522 mmol) was dissolved in 4M HCl/EtOAc and stirred for 1 hour. The reaction was diluted in saturated sodium bicarbonate EtOAc (3×30 mL). The combined organics were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. The residue was carried on without further purification. LCMS (ESI+): m/z 373.5 (M+H+).


The residue in was dissolved in MeOH (20 mL) and Water (2.0 mL). To this solution was added DIPEA (2.1 g, 2.8 mL, 6.4 Eq, 16 mmol), followed by a solution of acetaldehyde (433 mg, 550 μL, 3.90 Eq, 9.84 mmol) in ethanol. Sodium borohydride (400 mg, 4.19 Eq, 10.6 mmol) was added and the reaction was stirred overnight. Additional DIPEA (2.1 g, 2.8 mL, 6.4 Eq, 16 mmol), solution of acetaldehyde (433 mg, 550 μL, 3.90 Eq, 9.84 mmol) in ethanol, and sodium borohydride (400 mg, 4.19 Eq, 10.6 mmol) were added and the reaction was stirred for 2 hr. The reaction was concentrated by rotary evaporation and the crude product, 250 (1.01 g, 2.522 mmol, quant.), was used without further purification. LCMS (ESI+): m/z 401.2 (M+H+).


Step 4: tert-butyl ((S)-1-(((S)-3-(2-bromo-5-chlorophenyl)-1-(hex-5-en-1-yl(methyl)amino)-1-oxopropan-2-yl)(ethyl)amino)-4-methyl-1-oxopentan-2-yl)carbamate (251)



embedded image


250 (1.01 g, 1 Eq, 2.51 mmol) was dissolved in MeCN (25 mL) along with COMU (8 g, 7.4 Eq, 19 mmol), Boc-L-Leucine monohydrate (4.5 g, 7.2 Eq, 18 mmol), and DIPEA (5.6 g, 7.5 mL, 17 Eq, 43 mmol). The reaction was refluxed at 85° C. for 24 hours and allowed to cool to room temperature before being concentrated by rotary evaporation. The residue was partitioned between water (20 mL) and EtOAc (20 mL), and the aqueous layer was extracted with EtOAc (3×20 mL). The combined organics were washed with brine, dried over MgSO4, filtered, and concentrated by rotary evaporation. The crude product was purified by silica gel chromatography (0 to 100% EtOAc in hexanes) to yield 251 (440 mg, 2.51 mmol, 25%). LCMS (ESI+): m/z 617.8 (M+H+).


Steps 5-9: tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-bromo-5-chlorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (252)



embedded image


embedded image


Steps 5-9 for the synthesis of 252 were carried out using reaction sequences and procedures outlined in Method A, Steps 4-8. LCMS (ESI): m/z 780.4 (M+H+).


2. Final Synthesis Procedures to Prepare Compounds of Formula (I)

a. Method 1




embedded image


Representative Procedure for Method 1: Example 118



embedded image


Representative Experimental for Method 1: Example 118
Tert-butyl ((S)-2-(((3S,6S,9S)-3-(5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)benzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (219)



embedded image


The macrocyclic starting material for Method 1 was synthesized Using Method A with Int. 1, and 8. It was used as a mixture of E/Z isomers. LCMS (ESI+): m/z 770.4 (M+H).


Step 1: (S)-2-amino-N-((3S,6S,9S)-3-(5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)benzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)-2-cyclopropyl-N-methylacetamide (220)



embedded image


219 (112 mg, 1 Eq, 145 μmol) was dissolved in DCM/TFA (3:1) and stirred until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford a quantitative yield of 220 (97.4 mg, 145 μmol, 100%) as the TFA salt. LCMS (ESI+): m/z 732.4 (M+H).


Step 2: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-(5-chloro-2-(5-methyl-1,3,4-oxadiazol-2-yl)benzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 118)



embedded image


220 (48.7 mg, 1 Eq, 72.7 μmol) and Int. 141 (27.8 mg, 1.2 Eq, 87.2 μmol) were dissolved in DMF (2.0 mL). DIPEA (56.3 mg, 75.9 μL, 6 equiv., 436 μmol) and HATU (33.2 mg, 1.2 Eq, 87.2 μmol) were added sequentially and the reaction was continued until the consumption of starting material (monitored via LCMS). The reaction mixture was neutralized to pH ˜7 with 1N HCl and directly purified by RP-HPLC (C18, 40% to 75% MeCN/H2O with 0.5% formic acid). Example 118 was isolated as two peaks. The first eluting (28.5 mg, 29.3 μmol, 40.4%) and the second eluting peak (18.2 mg, 18.7 μmol, 25.7%) were isolated as a white solid after lyophilization. Stereochemistry of isolated peaks was not assigned. LCMS data was identical for both peaks. LCMS (ESI+): m/z 971.3 (M+H).


b. Method 2




embedded image


Representative Procedure for Method 2: Example 273



embedded image


Representative Experimental for Method 2: Example 273
Tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-bromo-5-chlorobenzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-12-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (221)



embedded image


The macrocyclic Starting material for Method 2 was synthesized Using Method A with Int. 2, 30, and 42. It was used as a mixture of E/Z isomers. LCMS (ESI+): m/z 766.2 (M+H).


Step 1: tert-butyl ((S)-2-(((3S,6S,9S)-3-(5-chloro-2-(1-(methylsulfonyl)-1H-pyrazol-4-yl)benzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-12-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (222)



embedded image


221 (124 mg, 2 Eq, 456 μmol), PdCl2(dppf)-DCM adduct (186 mg, 1 Eq, 228 mol), Int. 64, and sodium carbonate (96.7 mg, 4 equiv., 912 μmol) were dissolved in 4:1 Dioxane/Water (6.3 mL). The reaction was degassed for 10 min with Ar and then heated to 80° C. until the complete consumption of starting material monitored by LCMS. The vial was then cooled and adjusted to pH ˜7 with 1N HCl. The solution was filtered and directly purified by RP-HPLC (C18, 50% to 80% MeCN/H2O with 0.5% formic acid) to obtain 222 (150 mg, 180 μmol, 79%) as a white solid after lyophilization. LCMS (ESI+): m/z 832.4 (M+H).


Step 2: (S)-2-amino-N-((3S,6S,9S)-3-(5-chloro-2-(1-(methylsulfonyl)-1H-pyrazol-4-yl)benzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-12-en-9-yl)-2-cyclopropyl-N-methylacetamide (223)



embedded image


222 (126.6 mg, 1 Eq, 152.1 μmol) was dissolved in DCM/TFA (3:1) and stirred until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford a quantitative yield of 223 (111.4 mg, 152.1 μmol, 100.0%) as the TFA salt. LCMS (ESI+): m/z 732.4 (M+H).


Step 3: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-(5-chloro-2-(1-(methylsulfonyl)-1H-pyrazol-4-yl)benzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-12-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 273)



embedded image


223 as the TFA salt (111.4 mg, 1 Eq, 152.1 μmol) and Int. 141 (58.27 mg, 1.2 Eq, 182.5 μmol) were dissolved in DMF (5.0 mL). DIPEA (118.0 mg, 159 μL, 6 Eq, 912.7 μmol) and HATU (69.41 mg, 1.2 Eq, 182.5 μmol) was added sequentially and the reaction was continued until consumption of starting material (monitored by LCMS). The reaction mixture was neutralized to pH ˜7 with 1N HCl and directly purified by RP-HPLC (C18, 40% to 70% MeCN/H2O with 0.5% formic acid). Example 273 (31.72 mg, 30.69 μmol, 20.18%) was obtained as a white solid after lyophilization. LCMS (ESI+): m/z 1033.4 (M+H).


c. Method 3




embedded image


Representative Procedure for Method 3: Example 53



embedded image


Representative Experimental for Method 3: Example 53
Tert-butyl ((S)-2-(((3S,6S,16S)-16-(2-bromo-5-chlorobenzyl)-3-isobutyl-1-methyl-2,5,15-trioxo-113-chlora-4,14-diazacyclohexadec-8-en-6-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (224)



embedded image


The starting material for Method 3, 224, was synthesized according to method A using Int. 1 and 30. LCMS (ESI+): 766.2 m/z (M+H).


Steps 1-2: Ethyl 4-(2-(((3S,6S,9S)-9-((S)-2-((tert-butoxycarbonyl)amino)-2-cyclopropyl-N-methylacetamido)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-3-yl)methyl)-4-chlorophenyl)-3,6-dihydropyridine-1 (2H)-carboxylat (225)



embedded image


224 (100 mg, 1 Eq, 127 μmol), Int. 67 (31.1 mg, 1 Eq, 127 μmol), Na2CO3 (40.3 mg, 3 Eq, 380 μmol) and PdCl2(dppf) (10.4 mg, 0.1 Eq, 12.7 μmol) were dissolved in 4:1 Dioxane/Water (4 mL). The reaction was degassed for 10 min with Ar and then heated to 80° C. The reaction was stopped by the addition of 5 mL water upon the consumption of starting material (monitored by LCMS). The aqueous layer was extracted EtOAc (2×10 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The material could either be purified by RP-HPLC (C18, 10% to 50% MeCN/H2O with 0.5% formic acid) or used directly in the next reaction.


The crude reaction product was dissolved in DCM (2 mL), cooled in an ice bath before the addition of TEA (64.2 mg, 88.4 μL, 5 Eq, 634 μmol). ethyl chloroformate (20.6 mg, 18.3 μL, 1.5 Eq, 190 μmol) was added slowly to the cooled solution. The reaction was stirred at 0° C. until the complete consumption of starting material. Once complete the volatile solvents were completely removed and the crude residue was purified directly by RP-HPLC (C18, 50% to 80% MeCN/H2O with 0.5% formic acid) which afforded 225 (54 mg, 64 μmol, 51%) as an off-white solid. LCMS (ESI+): 841.3 m/z (M+H).


Steps 3-4: Ethyl 4-(4-chloro-2-(((3S,6S,9S)-9-((S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-N-methylacetamido)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-3-yl)methyl)phenyl)-3,6-dihydropyridine-1 (2H)-carboxylate (Example 53)



embedded image


225 (107 mg, 1 Eq, 127 μmol) was dissolved in DCM/TFA (3:1) and stirred until the deprotection was complete (ministered by LCMS). The volatile solvent was completely removed to afford a quantitative yield of the free amine.


The deprotected amine and Int. 141 (44.8 mg, 1.1 Eq, 140 μmol) were dissolved in DMF (1 mL). DIPEA (98.9 mg, 133 μL, 6 Eq, 765 μmol) and HATU (53.3 mg, 1.1 Eq, 140 mol) were added sequentially. and the reaction continued until the consumption of my starting material by LCMS. The reaction mixture was neutralized to pH ˜7 with 1 N HCl and directly purified by RP-HPLC(C18, 40% to 70% MeCN/H2O with 0.5% formic acid). Example 53 (48.8 mg, 46.8 μmol, 37%) was obtained as a white solid after lyophilization. LCMS (ESI+): m/z 1043.6 (M+H).


d. Method 4




embedded image


Representative Procedure for Method 4: Example 50



embedded image


embedded image


Representative Experimental for Method 4: Example 50
Tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-bromo-5-chlorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (226)



embedded image


The macrocycle starting material for method 4 (226), was synthesized using Method A with Int. 30. It was used as a mixture of E/Z isomers (˜9:1) LCMS: m/z [M+H]. LCMS (ESI+): 780.3 m/z (M+H).


Step 1: ((S)-2-(((3S,6S,9S)-3-(2-bromo-5-chlorobenzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (227)



embedded image


226 (12.0 g, 1 Eq, 15.6 mmol) was dissolved in EtOAc (80 ml) by stirring at ambient temperature. platinum(IV) oxide (284 mg, 0.08 Eq, 1.25 mmol) was added to the stirring reaction mixture. The reaction was then placed under an atmosphere of hydrogen and stirred vigorously until LCMS analysis showed complete consumption of starting material. The reaction mixture was filtered over a pad of Celite. The Filter was washed with EtOAc (2×50 mL). The organic solvent was concentrated to afford 227 (11.8 g, 15.3 mmol, 98%) as an off white solid. The reaction was used without further purification. LCMS: m/z 782.2 [M+H]+


Step 2: tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-(1-acetyl-1,2,3,6-tetrahydropyridin-4-yl)-5-chlorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (228)



embedded image


227 (34.5 mg, 1.0 Eq, 44.0 μmol), Na2CO3 (14.0 mg, 3 Eq, 132 μmol), Int. 47 (22.1 mg, 2.0 Eq, 88.1 μmol) and PdCl2(dppf)-CH2Cl2 adduct (3.60 mg, 0.1 Eq, 4.40 μmol) were dissolved in 4:1 Dioxane/Water (4 mL). The reaction was degassed for 10 min with Ar and then heated to 80° C. until the complete consumption of starting material. The vial was then cooled and adjusted to pH ˜7 with 1 N HCl. The homogeneous solution was directly purified by RP-HPLC (C18, 50% to 85% MeCN/H2O with 0.5% formic acid) to obtain 228 (31.6 mg, 44.0 μmol, 86.7%) as a white solid after lyophilization. LCMS (ESI+): m/z 827.4 (M+H).


Step 3: (S)—N-((3S,6S,9S)-3-(2-(1-acetyl-1,2,3,6-tetrahydropyridin-4-yl)-5-chlorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)-2-amino-2-cyclopropyl-N-methylacetamide TFA salt (229)



embedded image


228 (31.6 mg, 1 Eq, 38.2 μmol) was dissolved in 6 mL DCM/TFA (3:1) and stirred until the deprotection was complete (monitored by LCMS). The volatile solvent was completely removed to afford a quantitative yield of 229. LCMS (ESI+): m/z 727.4 (M+H).


Step 4: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-(2-(1-acetyl-1,2,3,6-tetrahydropyridin-4-yl)-5-chlorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 50)



embedded image


The deprotected amine and Int. 141 (13.4 mg, 1.1 Eq, 42.1 μmol) were dissolved in DMF (1 mL). DIPEA (29.7 mg, 40.0 μL, 6 Eq, 229 μmol) and HATU (16.0 mg, 1.1 Eq, 42.1 mol) were added sequentially and the reaction was continued until the consumption of starting material (monitored by LCMS). The reaction mixture was neutralized to pH ˜7 with 1N HCl and directly purified by RP-HPLC (C18, 45% to 75% MeCN/H2O with 0.5% formic acid). Example 50 (25.0 mg, 24.3 μmol, 64%) was obtained as a white solid after lyophilization. LCMS (ESI+): m/z 1028.6.4 (M+H).


e. Method 5




embedded image


Representative Procedure for Method 5: Example 45



embedded image


embedded image


Representative Experimental for Method 5: Example 45
Step 1: Tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-(1-acetyl-1,2,3,6-tetrahydropyridin-4-yl)-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (230)



embedded image


208 (300 mg, 1 Eq, 392 μmol), Int. 47 (128 mg, 1.3 Eq, 510 μmol), Na2CO3 (125 mg, 3 Eq, 1.18 mmol) and PdCl2(dppf)-DCM adduct (32.0 mg, 0.1 Eq, 39.2 μmol) were dissolved in 4:1 Dioxane/Water (10 mL). The reaction was degassed for 10 min with Ar and then heated to 80° C. until the complete consumption of starting material monitored by LCMS. The vial was then cooled and adjusted to pH ˜7 with 1N HCl. The solution was concentrated to ˜4 mL, filtered, then directly purified by RP-HPLC (C18, 50% to 80% MeCN/H2O with 0.5% formic acid) to obtain Int. 430 (200 mg, 247 μmol, 63%). LCMS: m/z 809.4 [M+H].


Step 2: Tert-butyl ((S)-2-(((3S,6S,9S)-3-(2-(1-acetyl-1,2,3,6-tetrahydropyridin-4-yl)-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (231)



embedded image


230 (100 mg, 1 Eq, 124 μmol) was dissolved in MeOH (2 mL). Pd/C (100 mg) was carefully added, and the reaction was stirred under an atmosphere of H2. Upon the consumption of starting material (monitored by LCMS), the reaction was filtered over a pad of celite. The filter cake was washed with EtOAc (2×20 mL). the filtrate was concentrated to afford 231 (85.7 mg, 105 μmol, 85%) d as an off-white solid. It was used without any further purification. LCMS: m/z 813.5 [M+H].


Step 3: (S)—N-((3S,6S,9S)-3-(2-(1-acetylpiperidin-4-yl)-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)-2-amino-2-cyclopropyl-N-methylacetamide (232)



embedded image


231 (40.0 mg, 1 Eq, 49.2 μmol) was dissolved in 2 ml DCM/TFA (3:1) and stirred until the deprotection was complete (monitored by LCMS). The volatile solvent was completely removed to afford a quantitative yield of 232 as the TFA salt. LCMS: m/z 713.4 [M+H].


Step 4: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-(2-(1-acetylpiperidin-4-yl)-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 45)



embedded image


232 as the TFA salt (40.0 mg), Int. 141 (17.3 mg, 1.1 Eq, 54.1 μmol) and HATU (20.6 mg, 1.1 Eq, 54.1 μmol) were dissolved in DMF (2 mL). The reaction was stirred and DIPEA (38.2 mg, 51.4 μL, 6 Eq, 295 μmol) was added slowly. The reaction continued until the consumption of starting material (monitored by LCMS). The reaction mixture was neutralized to pH ˜7 with 1N HCl and directly purified by RP-HPLC (C18, 40% to 70% MeCN/H2O with 0.5% formic acid). Example 45 (22.0 mg, 21.7 μmol, 44%) was isolated as a white solid upon lyophilization. LCMS: m/z 1015.5[M+H].


f. Method 6




embedded image


Representative Procedure for Method 6: Example 331



embedded image


Representative Experimental For Method 6: Example 331
Tert-butyl ((S)-2-(((3S,6S,9S)-3-((5-chloro-2-methylpyridin-3-yl)methyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (233)



embedded image


The macrocycle starting material for Method 6, 233, was synthesized using Method A with Int. 1 and 27. It was used as a mixture of E/Z isomers. LCMS (ESI+): 703.3 m/z (M+H).


Step 1: tert-butyl ((S)-2-(((3S,6S,9S)-3-((5-chloro-2-methylpyridin-3-yl)methyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (234)



embedded image


233 (230.6 mg, 1 Eq, 327.9 μmol) was dissolved in EtOAc (5.0 mL) by stirring at ambient temperature. Platinum(IV) oxide (11.17 mg, 0.15 equiv., 49.18 μmol) was added to the stirring reaction mixture. The reaction was then placed under an atmosphere of hydrogen and stirred vigorously until LCMS analysis showed complete consumption of starting material. The reaction mixture was filtered over a pad of Celite. The filter was washed with EtOAc (2×50 mL). The organic solvent was concentrated to afford 234 (220 mg, 312 μmol, 95%) as an off white solid. The reaction was used without further purification. LCMS: m/z 705.3 [M+H].


Step 2: (S)-2-amino-N-((3S,6S,9S)-3-((5-chloro-2-methylpyridin-3-yl)methyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)-2-cyclopropyl-N-methylacetamide (235)



embedded image


234 (231.3 mg, 1 Eq, 327.9 μmol) was dissolved in 6 mL DCM/TFA (3:1) and stirred until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford a quantitative yield of 235 as the TFA salt. LCMS (ESI+): m/z 605.7 (M+H).


Step 3: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-((5-chloro-2-methylpyridin-3-yl)methyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 331)



embedded image


235 as the TFA salt (198.5 mg, 1 Equiv., 328.0 μmol) and Int. 141 (125.6 mg, 1.2 Equiv., 393.6 μmol) were dissolved in DMF (2.0 mL). DIPEA (254.3 mg, 343 μL, 6 Equiv., 1.968 mmol) and HATU (149.7 mg, 1.2 Equiv., 393.6 μmol) were added sequentially and the reaction was continued until the consumption of starting material (monitored via LCMS). The reaction mixture was neutralized to pH ˜7 with 1N HCl and directly purified by RP-HPLC (C18, 40% to 80% MeCN/H2O with 0.5% formic acid). Example 331 (83.25 mg, 91.85 μmol, 28%)) was obtained as a white solid after lyophilization. LCMS (ESI+): m/z 906.6 (M+H).


g. Method 7




embedded image


Representative Procedure for Method 7: Example 165 and Example 58



embedded image


embedded image


Representative Experimental for Method 7: Example 165 and Example 58
Step 1: tert-butyl((1S)-2-(((3S,6S,9S)-4-acetyl-3-(2-bromo-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,414,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (236)



embedded image


208 (14 g, 1 Equiv., 18 mmol) was dissolved in EtOAc (180 mL) and platinum (IV) oxide (0.33 g, 0.08 Equiv., 1.5 mmol) was added. The reaction was placed under a hydrogen atmosphere and vigorously stirred. The reaction was run rt overnight. The reaction was filtered over a bed of celite and the solvent was reduced and 236 was isolated as an off-white solid (Quant. 14.0 g). The reaction was used in the next step without any further purification. LCMS (ESI+): m/z 766.4 (M+H).


Step 2-3: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-(2-bromo-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (237)



embedded image


236 (300 mg, 1 Equiv., 391 μmol) was dissolved in 6 mL DCM/TFA (3:1) and stirred until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford a quantitative yield of free amine. The crude deprotected amine was used in the next step without any further purification.


The deprotected amine, Int. 141 (137 mg, 1.1 Equiv., 430 μmol) and HATU (164 mg, 1.1 Equiv., 430 μmol) were dissolved in DMF (4 mL) and DIPEA (253 mg, 341 μL, 5 Equiv., 1.96 mmol) was added slowly. The reaction was stopped by the addition of 15 mL water upon the consumption of starting material (monitored by LCMS). The aqueous layer was extracted with EtOAc (2×10 mL). The combined organic layer was washed with brine, dried with MgSO4, filtered, and concentrated. 237 (300 mg, 310 μmol, 79%) was isolated as an off-white solid and used without further purification. LCMS (ESI+): m/z 967.3 (M+H).


Step 4: Tert-butyl 3-(2-(((3S,6S,9S)-9-((S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-N-methylacetamido)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-3-yl)methyl)-4-fluorophenyl)-8-azabicyclo[3.2.1]oct-2-ene-8-carboxylate (238)



embedded image


237 (54.0 mg, 1.2 Equiv., 161 μmol), Na2CO3 (47.0 mg, 222 p L, 2.0 molar, 3.3 Equiv., 443 μmol), Int. 134 (31.2 mg, 2 Equiv., 93.0 μmol) and PdCl2(dppf)-DCM adduct (16.5 mg, 0.15 Equiv., 20.1 μmol) were dissolved in 4:1 Dioxane/Water (4 mL). The reaction was degassed for 10 min with Ar and then heated to 80° C. until the complete consumption of starting material (monitored by LCMS). The reaction mixture was diluted with water (5 mL) and extracted with EtOAc (2×10 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. 208 (as a mixture of diastereomers) was used in the next reaction without any additional purification. LCMS (ESI+): m/z 1097.5 (M+H).


Step 5: (2S,4R)—N-((1S)-2-(((3S,6S,9S)-3-(2-(8-azabicyclo[3.2.1]oct-2-en-3-yl)-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Exampl 165)



embedded image


The crude Suzuki product was dissolved in 2 mL DCM/TFA (3:1) and stirred until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford the crude deprotection product. The remaining TFA was removed by co-evaporation with toluene. The material can be used without any further purification in the next reaction. A portion of the crude reaction mixture was isolated via RP-HPLC (C18, 15% to 50% MeCN/H2O with 0.5% formic acid) to afford Example 165 (as a mixture of diastereomers, Formic Acid Salt) as a white powder after lyophilization. The remaining unpurified material was used in the next reaction directly. LCMS (ESI+): m/z 996.47 (M+H).


Step 6: (2S,4R)—N-((1S)-2-(((3S,6S,9S)-3-(2-(8-azabicyclo[3.2.1]oct-2-en-3-yl)-5-fluorobenzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 58)



embedded image


Example 165 was dissolved in DCM (4 mL), TEA was added to adjust the pH to ˜8.0 (˜4 equiv.). Next, the reaction was cooled with an ice bath and AcCl (2.4 mg, 2.1 μL, 1 Equiv., 30 μmol) was added. Upon the complete consumption of starting material, the reaction was concentrated and directly purified by RP-HPLC (C18, 50% to 80% MeCN/H2O with 0.5% formic acid) to afford Example 58 as a mixture of diastereomers (13 mg, 12 μmol, 40%) as a white solid after lyophilization. LCMS (ESI+): m/z 1038.59 (M+H).


h. Method 8




embedded image


Representative Experimental for Method 8: Example 1
Step 1: N—((S)-1-(((S)-1-(((3S,6S,9S)-3-(2,5-dichlorobenzyl)-6-isobutyl-4-methyl-2,5,8-trioxo-1,4,7-triazacyclohexadec-11-en-9-yl)amino)-1-oxopropan-2-yl)amino)-1-oxobutan-2-yl)-1-(trifluoromethyl)cyclopropane-1-carboxamide (Example 1)



embedded image


218 (30 mg, 1 Equiv., 41 μmol) was dissolved in MeOH (2 ml), and K2CO3 (7.0 mg, 1.2 Equiv., 51 μmol) was added while stirring. The reaction continued until the complete removal of the FMOC protecting group (monitored by LCMS). Upon completion, the reaction was concentrated and DCE (2×4 mL) was used to dissolve the deprotected amine. The DCE was filtered and concentrated. The crude amine was used directly in the next reaction.


The deprotected amine from above, Int. 34 (15 mg, 1.2 Equiv., 48 μmol), and HATU (16 mg, 1 Equiv., 41 μmol) were dissolved in DMF (2 mL). DIPEA (35.5 μL, 1 equiv., 205 μmol) was added and the reaction was continued until the complete consumption of starting material (monitored by LCMS). Upon completion, the reaction mixture was neutralized with minimal 1N HCl and directly purified by RP-HPLC (C18, 40% to 70% MeCN/H2O with 0.5% formic acid). Example 1 (4.5 mg, 5.6 μmol, 14%) was isolated after lyophilization as a white solid. LCMS (ESI+): m/z 803.4 (M+H).


i. Method 9




embedded image


Representative Procedure for Method 9: Example 359



embedded image


Representative Expiremental for Method 9: Example 359



embedded image


The starting material for Method 9, 239, was synthesized using identical procedures for steps 1-3 of Method 7 using 226 and Int. 141 LCMS (ESI): m/z 985.5 [M+H].


Step 4: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-((4-chloro-4′-(morpholinomethyl)-[1,1′-biphenyl]-2-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 359)



embedded image


239 (125 mg, 1 Equiv. 127 mol), 4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)morpholine (38.5 mg, 1 Equiv. 127 μmol), and Chloro[(di(1-adamantyl)-N-butylphosphine)-2-(2-aminobiphenyl)]palladium(II) (8.49 mg, 0.1 Equiv. 12.7 μmol) were dissolved in 1.0 mL THF and Na3PO4 (267 μL, 1.0 molar, 2.1 Equiv. 267 μmol). The mixture was degassed with a stream of Argon, then sealed and stirred at 70° C. until the complete consumption of starting material (monitored by LCMS). The reaction was then cooled to room temperature and adjusted to pH ˜7 with 1N HCl. The reaction mixture was filtered and directly purified by RP-HPLC (C18, 30% to 60% MeCN/H2O with 0.5% formic acid). Example 359 (48.8 mg, 45.2 μmol, 35.6%) was isolated as a white solid after lyophilization. LCMS (ESI): m/z 1080.3 [M+H].


j. Method 10




embedded image


Representative Procedure for Method 10: Example 382



embedded image


Step 1: tert-butyl ((S)-2-(((3S,6S,9S)-3-(5-chloro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (240)



embedded image


To a solution of macrocycle 227 (300 mg, 383.03 μmol, 1 eq) and B2Pin2 (194.53 mg, 766.06 μmol, 2 eq) in Toluene (5 mL) was added KOAc (75.18 mg, 766.06 μmol, 2 eq) and ditert-butyl(cyclopentyl)phosphane; dichloropalladium; iron (24.96 mg, 38.30 μmol, 0.1 eq) under N2 atmosphere. The reaction mixture was heated to 80° C. and stirred for 28 hr. The reaction mixture was cooled to room temperature and quenched by water (10 mL), extracted with EtOAc (10 mL×2). The combined organic layer was washed with brine (15 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was first purified by flash column chromatography (SiO2, 0 to 50% EtOAc/Pet. Ether). Then by prep-HPLC (C18, using H2O (10 mM NH4HCO3) with MeCN 50 to 80% over 8.0 min). The product was lyophilized to give 240 (20 mg, 5.78 μmol, 1.51% yield) as a white solid. LCMS (ESI): 730.6 (M−Boc)


Step 2: tert-butyl ((S)-2-(((3S,6S,9S)-3-(5-chloro-2-(5-morpholinopyridin-2-yl)benzyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)carbamate (241)



embedded image


240 200 mg, 1 Equiv. 241 μmol), 4-(6-bromopyridin-3-yl)morpholine (82.0 mg, 1.4 Equiv. 337 μmol), and PdCl2(dppf) (19.7 mg, 0.1 Equiv. 24.1 μmol) were dissolved in mixture of 1,4-Dioxane (1.3 mL) and 2M Na2CO3 (58.7 mg, 277 μL, 2.3 Equiv. 554 μmol). The reaction was degassed with a stream of Ar then heated to 100° C. for 6 hr. The reaction was then cooled and adjusted to pH˜7 with Formic Acid. The crude reaction mixture was then diluted in MeOH (2 mL), filtered, and directly purified by RP-HPLC (C18, 50% to 100% MeCN/H2O with 0.5% formic acid) to obtain 241 (95 mg, 0.11 mmol, 46%) as an off-white powder after lyophilization. LCMS (ESI): m/z 966.3 [M+H].


Steps 3-4: Example 382



embedded image


Example 382 was synthesized using identical procedures for Method 4 (Steps 3-4) using 241 and Int. 141. LCMS (ESI): m/z 1068.2 [M+H].


k. Method 11




embedded image


Representative Procedure for Method 11: Example 383



embedded image


embedded image


Step 1: tert-butyl 3-(4-(4′-chloro-2′-(((3S,6S,9S)-9-((S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-N-methylacetamido)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-3-yl)methyl)-[1,1′-biphenyl]-4-yl)piperazin-1-yl)azetidine-1-carboxylate (242)



embedded image


239 (550 mg, 1 Equiv. 559 μmol), Int. 193 (496 mg, 2 Equiv. 1.12 mmol), Na2CO3 (237 mg, 4 Equiv. 2.24 mmol) and PdCl2(dppf) (45.6 mg, 0.1 Equiv. 55.9 μmol) were dissolved in Dioxane:Water (4:1) (5.59 mL). The reaction was degassed for 10 min with Ar and then heated to 80° C. until the complete consumption of starting material (monitored by LCMS). The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (2×10 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The crude reaction mixture was isolated via RP-HPLC (C18, 30% to 80% MeCN/H2O with 0.5% formic acid) to afford 242 (400 mg, 328 μmol, 58.6%) as a white powder after lyophilization. LCMS (ESI+): m/z 1220.7 (M+H).


Step 2: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-((4′-(4-(azetidin-3-yl)piperazin-1-yl)-4-chloro-[1,1′-biphenyl]-2-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (243)



embedded image


242 (400 mg, 328 μmol, 1 Eq) was dissolved in 2 mL 3N HCl in MeOH and stirred at 50° C. until the deprotection was complete (monitored via LCMS). The volatile solvent was completely removed to afford the crude deprotection product, 243, as the HCl salt. The material can be used without any further purification in the next reaction. LCMS 1120.7 (ESI+): m/z (M+H).


Step 3: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-((4-chloro-4′-(4-(1-methylazetidin-3-yl)piperazin-1-yl)-[1,1′-biphenyl]-2-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide Example 383



embedded image


243 (300 mg, 1 Equiv. 268 μmol), formaldehyde (43.5 mg, 39.9 μL, 37% Wt, 2 Equiv. 535 μmol) and DIPEA (138 mg, 187 μL, 4 Equiv. 1.07 mmol) was dissolved in THF (2.68 mL) stirred at 25° C. for 15 mins. Then, sodium triacetoxyborohydride (113 mg, 2 Equiv. 535 μmol) was added portion-wise to the reaction mixture. The reaction was carried on at 25° C. for 1 hour. The starting material was completely consumed (monitored by LCMS). The reaction mixture was diluted with water (15 mL) and extracted with EtOAc (2×15 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The crude reaction mixture was purified via RP-HPLC (C18, 30% to 70% MeCN/H2O with 0.5% formic acid) to afford Example 383 (150 mg, 132 μmol, 49.4%) as a white powder after lyophilization. LCMS (ESI+): m/z 1135.25 (M+H).


l. Method 12


Generic Procedure for Method 12



embedded image


Representative Procedure for Method 12: Example 392



embedded image


embedded image


Representative Experimental for Method 12: Example 392



embedded image


The starting material for Method 12, 244, was synthesized using Method A with int. 206. LCMS (ESI): m/z 652.4 (M+H+).


Steps 1-3: Methyl 2-((3S,6S,9S)-9-((S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-N-methylacetamido)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-3-yl)acetate (245)



embedded image


245 was synthesized using identical procedures for steps 1-3 of Method 7 using 244 and Int. 141. LCMS (ESI): m/z 854.1 (M+H+).


Steps 4-5: 1,3-Dioxoisoindolin-2-yl 2-((3S,6S,9S)-9-((S)-2-cyclopropyl-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-N-methylacetamido)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-3-yl)acetate (246)



embedded image


245 (2.0 g, 1 Eq, 2.345 mmol), was dissolved in THF (16.4 mL). In a separate vial, LiOH (112.3 mg, 2 Eq, 4.690 mmol) was dissolved in Water (10.9 mL) and the LiOH solution was added to the stirring THF solution. MeOH (˜1 mL) was added until the solution became homogeneous. The reaction was stirred at 25° C. until the complete consumption of starting material (monitored by LCMS). upon completion, the solution was acidified to pH˜3 with 1N HCl. The acidified mixture was diluted with water (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The crude carboxylic acid product was used directly in the next reaction. LCMS (ESI): m/z 839.3 (M+H+).


The crude carboxylic acid (590 mg, 1 Eq, 703 μmol), 2-hydroxyisoindoline-1,3-dione (126 mg, 1.1 Eq, 774 μmol) and DMAP (8.59 mg, 0.1 Eq, 70.3 mo) were dissolved in DCM (4.0 mL) and DIC (97.6 mg, 121 μL, 1.1 Eq, 774 μmol) was added. the reaction was stirred until the consumption of starting material was observed (TLC 50% EtOAc in hexane). The reaction was filtered over a pad of SiO2 which was washed with EtOAc (2×10 mL). The organic solvent was removed and the product, 246 (692 mg, 703 μmol, quant.), was used without further purification. Product decomposed to the carboxylic acid on LCMS.


Step 6: (2S,4R)—N—((S)-2-(((3S,6S,9S)-3-((5-chloro-1-methyl-1H-indol-7-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-1-cyclopropyl-2-oxoethyl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 392)



embedded image


DMA (2.0 mL) was added to a vial containing 246 (175 mg, 1 Eq, 178 μmol), 7-bromo-5-chloro-1-methyl-1H-indole (87.0 mg, 2 Eq, 356 μmol). Then, TMS-C1 (4.3 mg, 5.0 μL, 0.22 Eq, 39 μmol) was added and the mixture was degassed with a stream of Ar for 5 min. (Dtpby)NiBr2 (21.6 mg, 0.25 Eq, 44.5 μmol) was added, while degassing was continued for an additional 5 min. Next, the vial was quickly capped, and the mixture was stirred overnight at ambient temperature. LCMS showed the consumption of starting material, and the reaction was filtered and directly purified by RP-HPLC. Example 392 was isolated as a fluffy white solid (15.7 mg, 16.4 μmol, 9.21%). LCMS (ESI): m/z 958.48 (M+H+).


m. Method 13




embedded image


Representative Procedure for Method 13: Example 419



embedded image


embedded image


Representative Experimental for Method 13: Example 419



embedded image


The starting material for Method 13, 247, Method A with Int. 30, 207. LCMS (ESI): m/z 826.4 (M+H+).


Steps 1-4: ethyl(S)-5-(((3S,6S,9S)-3-((4-chloro-4′-morpholino-[1,1′-biphenyl]-2-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-4-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-5-oxopentanoate (248)



embedded image


Example 415 was synthesized using identical procedures from Method 7 Steps 1-4 using 247, Int. 117 and 141 as starting materials. LCMS (ESI): m/z 1113.11 (M+H+).


Steps 5-6: (S)—N1-((3S,6S,9S)-3-((4-chloro-4′-morpholino-[1,1′-biphenyl]-2-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)-2-((2S,4R)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamido)-N1,N5,N5-trimethylpentanediamide (Example 419)



embedded image


Example 415 (300 mg, 1 Eq, 270 μmol) was dissolved in THE (4.0 mL). In a separate vial, LiOH·water (23.8 mg, 2.1 Eq, 566 μmol) was dissolved in Water (1.0 mL). The LiOH solution was added into the stirring THF solution. MeOH (˜0.1 mL) was added dropwise to ensure a homogeneous reaction mixture. The reaction was quenched with 1N HCl (4 mL) upon the consumption of starting material (monitored by LCMS). The reaction was extracted with EtOAc (10 mL×2). The combined organic layers were washed with brine, dried with MgSO4, filtered, and concentrated to afford Example 418 (296 mg, 270 mol, 100%) as a white solid. The reaction was carried on without further purification. A small portion (˜15 mg) was purified by RP-HPLC for biological evaluation. LCMS (ESI): m/z 1090.02 (M+H+).


Example 418 (35 mg, 1 Eq, 32 μmol) was dissolved in DMF (1.0 mL). HATU (13 mg, 1.1 Eq, 35 μmol) and DIPEA (25 mg, 33 μL, 6 Eq, 0.19 mmol) were subsequently added. The reaction was stirred for 5 min. Then, 1M dimethylamine in THF (14 mg, 0.32 mL, 10 Eq, 0.32 mmol) was added. The reaction was continued until the complete consumption of starting material (monitored by LCMS). The reaction mixture was neutralized to pH ˜4, with 1N HCl filtered, and directly purified by RP-HPLC. Example 419 (20.9 mg, 18.6 μmol, 58%) was isolated as white solid after lyophilization. LCMS (ESI): m/z 1126.12 (M+H+).


n. Method 14




embedded image


Representative Procedure for Method 14: Example 420



embedded image


embedded image


Representative Experimental for Method 14: Example 420



embedded image


The intermediate for Method 14, Example 418, was synthesized using Method 13 steps 1-5.


Step 6: (2S,4R)—N—((S)-1-(((3S,6S,9S)-3-((4-chloro-4′-morpholino-[1,1′-biphenyl]-2-yl)methyl)-6-isobutyl-1,4-dimethyl-2,5,8-trioxo-1,4,7-triazacyclohexadecan-9-yl)(methyl)amino)-5-hydroxy-1-oxopentan-2-yl)-1-(3,3-difluoro-1-(trifluoromethyl)cyclobutane-1-carbonyl)-4-fluoropyrrolidine-2-carboxamide (Example 420)



embedded image


Example 418 (35 mg, 1 Eq, 32 μmol) was dissolved in THF 2 mL and was cooled to 0° C. BH3·THF (8.2 mg, 96 μL, 1.0 molar in THF, 3 Eq, 96 μmol) was added. The reaction was slowly warmed to RT and stirred overnight. The reaction mixture was quenched with the addition of sat. NaHCO3, filtered, and directly purified by RP-HPLC. Example 420 (7.8 mg, 7.2 μmol, 23%) was isolated as a white solid after lyophilization. LCMS (ESI): m/z 1085.25 (M+H+).


3. Exemplary Compounds Preparation Summary Tables

Table 2, below, provides information on the methods used to prepare the exemplified compounds in the current application. This table includes a column listing the Example Number, Macrocyclic Synthesis procedure, and Final Synthesis procedure. Chemical structures for the exemplified compounds are shown in Table 4, while analytical data for these compounds are shown in Table 3.


Certain examples in the following tables include a P1 or a P2 after the example number. This indicates that two isomers of an exemplary compound were isolated using HPLC purification, but the absolute stereochemistry for each isolated isomer was not determined. P1 refers to the first eluting peak, and P2 refers to the second eluting peak.









TABLE 2







Summary Table Procedures for Compound Preparation









Example




Number
Macrocycle Synthesis
Final Synthesis





 1
Method C
Method 8 with Int. 34


 2
Method C
Method 8 with Int. 35


 3
Method C
Method 8 with Int. 33


 4
Method C
Method 8 with Int. 127


 5-P1
Method A with Int. 1, Int. 6
Method 1 with Int. 140


 5-P2
Method A with Int. 1, Int. 6
Method 1 with Int. 140


 6-P1
Method A with Int. 1, Int. 6
Method 1


 6-P2
Method A with Int. 1, Int. 6
Method 1


 7
Method A with Int. 1, Int. 6
Method 6


 8
Method C
Method 8 with Int. 127


 9
Method A with Int. 1, Int. 6
Method 6 with Int. 140


 10
Method A with Int. 1, Int. 6
Method 6 with Int. 142


 11
Method A with Int. 6
Method 1 with Int. 140


 12
Method C with Int. 3
Method 8 with Int. 127


 13
Method C with Int. 3
Method 8 with Int. 35


 14
Method A with Int. 6
Method 1


 15
Method A with Int. 6
Method 1 with Int. 143


 16-P1
Method A with Int. 1, Int. 6
Method 1 with Int. 143


 16-P2
Method A with Int. 1, Int. 6
Method 1 with Int. 143


 17
Method A with Int. 1, Int. 30
Method 2 with Int. 46


 18
Method A with Int. 1, Int. 30
Method 2 with Int. 47


 19
Method A with Int. 1, Int. 30
Method 2 with Int. 48


 20
Method A with Int. 1, Int. 30
Method 4


 21
Method A with Int. 1, Int. 30
Method 4 with Int. 46


 22
Method A with Int. 1, Int. 30
Method 2 with Int. 49


 23
Method A with Int. 1, Int. 30
Method 2 with Int. 50


 24
Method A with Int. 1, Int. 30
Method 2 with Int. 51


 25
Method A with Int. 6
Method 6


 26
Method A with Int. 1, Int. 30
Method 2 with Int. 52


 27
Method A with Int. 1, Int. 30
Method 2 with Int. 53


 28
Method A with Int. 1, Int. 30
Method 2 with Int. 54


 29
Method A with Int. 30
Method 2 with Int. 52


 30
Method A with Int. 30
Method 2 with Int. 55


 31
Method A with Int. 30
Method 2 with Int. 47


 32
Method A with Int. 1, Int. 30
Method 2 with Int. 144, Int. 53


 33
Method A with Int. 1, Int. 30
Method 2 with Int. 144, Int. 54


 34
Method A with Int. 30
Method 2 with Int. 144, Int. 52


 35
Method A with Int. 30
Method 2 with Int. 144, Int. 55


 36
Method A with Int. 30
Method 2 with Int. 144, Int. 47


 37
Method A
Method 2 with Int. 52


 38
Method A
Method 2 with Int. 55


 39
Method A
Method 2 with Int. 47


 40
Method A
Method 2 With Int. 144, Int. 52


 41
Method A
Method 2 with Int. 144, Int. 55


 42
Method A
Method 2 with Int. 144, Int. 47


 43
Method A with Int. 1, Int. 30
Method 2 with Int. 56


 44
Method A with Int. 30
Method 2 with Int. 56


 45
Method A
Method 5


 46
Method A
Method 2 with Int. 57


 47
Method A
Method 2 with Int. 51


 48
Method A with Int. 6
Method 1 with Int. 144


 49
Method A with Int. 30
Method 4 with Int. 52


 50
Method A with Int. 30
Method 4


 51
Method A with Int. 30
Method 4 with Int. 46


 52
Method A with Int. 30
Method 5


 53
Method A with Int. 1, Int. 30
Method 3 with Int. 67, Int. 36


 54
Method A with Int. 1, Int. 6, Int. 32
Method 1


 55
Method A with Int. 1, Int. 6, Int. 32
Method 6


 56-P1
Method A with Int. 1, Int. 30
Method 2 with Int. 58


 56-P2
Method A with Int. 1, Int. 30
Method 2 with Int. 58


 57-P1
Method A with Int. 30
Method 7 with Int. 134


 57-P2
Method A with Int. 30
Method 7 with Int. 134


 58
Method A
Method 7 with Int. 134


 59-P1
Method A
Method 5 With Int. 45


 59-P2
Method A
Method 5 With Int. 45


 60
Method A with Int. 30
Method 7 with Int. 137


 61
Method A with Int. 1, Int. 30
Method 4 with Int. 68


 62
Method A with Int. 1, Int. 30
Method 4 with Int. 69


 63
Method A with Int. 1, Int. 30
Method 5 with Int. 68


 64
Method A with Int. 1, Int. 30
Method 5 with Int. 69


 65
Method A
Method 4 with Int. 52


 66
Method A with Int. 1, Int. 30
Method 4 with Int. 52


 67
Method A with Int. 1, Int. 30
Method 4 with Int. 59


 68
Method A
Method 4 with Int. 59


 69
Method A
Method 4 with Int. 144, Int. 52


 70
Method A with Int. 1, Int. 30
Method 4 with Int. 144, Int. 52


 71
Method A with Int. 4, Int. 30
Method 4 with Int. 46


 72
Method A with Int. 4, Int. 30
Method 4 with Int. 52


 73
Method A with Int. 4, Int. 30
Method 4 with Int. 59


 74
Method A with Int. 4, Int. 30
Method 4


 75
Method A with Int. 30
Method 4 with Int. 70


 76
Method A with Int. 30
Method 4 with Int. 145, Int. 52


 77
Method A with Int. 30
Method 4 with Int. 146, Int. 52


 78
Method A with Int. 30
Method 2 with Int. 59


 79
Method A with Int. 30
Method 2 with Int. 60


 80
Method A with Int. 30
Method 2 with Int. 144, Int. 60


 81
Method A with Int. 30
Method 2 with Int. 144, Int. 59


 82
Method A with Int. 30
Method 4 with Int. 147, Int. 52


 83
Method A
Method 2 with Int. 144, Int. 61


 84
Method A
Method 2 with Int. 62


 85
Method A
Method 2 with Int. 63


 86
Method A
Method 2 with Int. 59


 87
Method A
Method 2 with Int. 64


 88
Method A
Method 2 with Int. 65


 89
Method A with Int. 1, Int. 30
Method 2 with Int. 66


 90
Method A with Int. 1, Int. 30
Method 3 with Int. 67, Int. 37


 91
Method A with Int. 1, Int. 30
Method 3 with Int. 67, Int. 38


 92
Method A
Method 4 with Int. 144, Int. 61


 93
Method A
Method 4 with Int. 62


 94
Method A
Method 4 with Int. 63


 95
Method A
Method 4 with Int. 64


 96
Method A
Method 4 with Int. 65


 97
Method A with Int. 1, Int. 30
Method 4 with Int. 66


 98
Method A with Int. 1, Int. 30
Method 7 with Int. 138


 99
Method A with Int. 1, Int. 30
Method 7 with Int. 138


100
Method A
Method 4 with Int. 50


101
Method A
Method 5 with Int. 66


102-P1
Method A with Int. 1, Int. 7
Method 1


102-P2
Method A with Int. 1, Int. 7
Method 1


103
Method A with Int. 1, Int. 7
Method 6


104
Method A with Int. 7
Method 1


105
Method A with Int. 7
Method 6


106
Method A with Int. 7
Method 1 with Int. 144


107-P1
Method A with Int. 1, Int. 7
Method 1 with Int. 144


107-P2
Method A with Int. 1, Int. 7
Method 1 with Int. 144


108
Method A with Int. 1, Int. 7
Method 6 with Int. 144


109
Method A with Int. 7
Method 6 with Int. 144


110
Method A with Int. 30
Method 4 with Int. 59


111
Method A with Int. 30
Method 4 with Int. 71


112
Method A with Int. 30
Method 4 with Int. 65


113
Method A with Int. 30
Method 4 with Int. 63


114
Method A with Int. 30
Method 4 with Int. 62


115
Method A with Int. 8
Method 1 with Int. 144


116
Method A with Int. 8
Method 1


117-P1
Method A with Int. 1, Int. 8
Method 1 with Int. 144


117-P2
Method A with Int. 1, Int. 8
Method 1 with Int. 144


118-P1
Method A with Int. 1, Int. 8
Method 1


118-P2
Method A with Int. 1, Int. 8
Method 1


119
Method A with Int. 10
Method 6 with Int. 144


120
Method A with Int. 10
Method 6


121
Method A
Method 4 with Int. 72


122
Method A
Method 4 with Int. 147, Int. 59


123
Method A
Method 4 with Int. 130


124
Method A
Method 7 with Int. 135


125
Method A with Int. 1, Int. 9
Method 1


126
Method A with Int. 9
Method 1


127
Method A with Int. 1, Int. 9
Method 6


128
Method A with Int. 9
Method 6


129
Method A with Int. 1, Int. 10
Method 1


130
Method A with Int. 10
Method 1


131
Method A with Int. 1, Int. 10
Method 6


132
Method A with Int. 1, Int. 9
Method 1 with Int. 144


133
Method A with Int. 9
Method 1 with Int. 144


134
Method A with Int. 1, Int. 9
Method 6 with Int. 144


135
Method A with Int. 9
Method 6 with Int. 144


136-P1
Method A with Int. 1, Int. 10
Method 1 with Int. 144


136-P2
Method A with Int. 1, Int. 10
Method 1 with Int. 144


137
Method A with Int. 10
Method 1 with Int. 144


138
Method A with Int. 1, Int. 10
Method 6 with Int. 144


139
Method A with Int. 12
Method 6 with Int. 144


140
Method A with Int. 12
Method 6


141
Method A with Int. 1, Int. 12
Method 6 with Int. 144


142
Method A with Int. 1, Int. 12
Method 6


143
Method A with Int. 30
Method 7 with Int. 138, Int. 37


144
Method A with Int. 30
Method 7 with Int. 138, Int. 38


145
Method A with Int. 30
Method 7 with Int. 138, Int. 36


146
Method A
Method 4 with Int. 148, Int. 59


147
Method A
Method 4 with Int. 149, Int. 59


148
Method A
Method 4 with Int. 73


149
Method A
Method 7 with Int. 134


150
Method A
Method 7 with Int. 134


151
Method A
Method 7 with Int. 135


152
Method A with Int. 1, Int. 11
Method 1


153
Method A with Int. 11
Method 1


154
Method A with Int. 1, Int. 11
Method 6


155
Method A with Int. 11
Method 6


156-P1
Method A with Int. 1, Int. 11
Method 1 with Int. 144


156-P2
Method A with Int. 1, Int. 11
Method 1 with Int. 144


157
Method A with Int. 11
Method 1 with Int. 144


158
Method A with Int. 1, Int. 11
Method 6 with Int. 144


159
Method A with Int. 11
Method 6 with Int. 144


160
Method A with Int. 1, Int. 8
Method 6


161
Method A with Int. 1, Int. 8
Method 6 with Int. 144


162
Method A with Int. 8
Method 6


163
Method A with Int. 8
Method 6 with Int. 144


164
Method A
Method 4 with Int. 74


165
Method A
Method 7


166
Method A
Method 7 with Int. 135


167
Method A with Int. 30
Method 4 with Int. 75


168
Method B with Int. 30
Method 2 with Int. 52


169
Method A with Int. 30
Method 4 with Int. 131


170
Method A with Int. 30
Method 4 with Int. 76


171
Method A with Int. 30
Method 4 with Int. 77


172
Method A with Int. 30
Method 4 with Int. 78


173
Method B with Int. 30
Method 2 with Int. 144, Int. 52


174
Method B with Int. 30
Method 2 with Int. 149, Int. 52


175
Method B with Int. 30
Method 2 with Int. 59


176
Method B with Int. 30
Method 2 with Int. 144, Int. 59


177
Method A with Int. 30
Method 4 with Int. 79


178
Method A with Int. 30
Method 4 with Int. 80


179
Method A with Int. 30
Method 4 with Int. 81


180
Method A with Int. 30
Method 4 with Int. 132


181
Method A with Int. 30
Method 4 with Int. 82


182
Method A with Int. 30
Method 4 with Int. 83


183
Method A with Int. 30
Method 4 with Int. 148, Int. 52


184
Method A with Int. 30
Method 4 with Int. 149, Int. 52


185
Method A with Int. 30
Method 4 with Int. 144, Int. 52


186
Method A
Method 4 with Int. 144, Int. 59


187
Method A with Int. 30
Method 4 with Int. 84


188
Method A with Int. 1, Int. 30
Method 4 with Int. 144, Int. 59


189
Method A with Int. 1, Int. 30
Method 2 with Int. 59


190
Method A with Int. 30
Method 4 with Int. 73


191
Method A with Int. 30
Method 4 with Int. 85


192
Method A with Int. 30
Method 4 with Int. 86


193
Method A with Int. 1, Int. 30
Method 7 with Int. 134


194
Method A with Int. 1, Int. 30
Method 7 with Int. 135


195
Method A with Int. 1
Method 2 with Int. 52


196
Method A with Int. 30
Method 4 with Int. 121


197
Method A with Int. 30
Method 4 with Int. 87


198
Method A with Int. 30
Method 4 with Int. 88


199
Method A with Int. 30
Method 4 with Int. 87


200
Method A with Int. 30
Method 4 with Int. 90


201
Method A with Int. 30
Method 4 with Int. 91


202
Method A with Int. 30
Method 4 with Int. 92


203
Method A with Int. 30
Method 4 with Int. 143, Int. 52


204
Method A with Int. 30
Method 4 with Int. 150, Int. 52


205
Method A with Int. 30
Method 4 with Int. 93


206
Method A with Int. 30
Method 4 with Int. 94


207
Method A with Int. 30
Method 4 with Int. 95


208
Method A with Int. 30
Method 4 with Int. 96


209
Method A with Int. 30
Method 4 with Int. 97


210
Method A with Int. 30
Method 4 with Int. 98


211
Method A with Int. 30
Method 4 with Int. 99


212
Method A with Int. 30
Method 4 with Int. 100


213
Method A with Int. 1
Method 4 with Int. 59


214
Method A with Int. 1
Method 4 with Int. 73


215
Method A with Int. 1
Method 4 with Int. 52


216
Method A with Int. 30
Method 4 with Int. 101


217
Method A with Int. 1
Method 4


218
Method A with Int. 30
Method 4 with Int. 52


219
Method A with Int. 7, Int. 41
Method 6


220
Method A with Int. 7, Int. 41
Method 6 with Int. 144


221
Method A with Int. 7, Int. 41
Method 6 with Int. 149


222
Method A with Int. 13
Method 6


223
Method A with Int. 13
Method 6 with Int. 149


224
Method A with Int. 13
Method 6 with Int. 144


225
Method A with Int. 14
Method 6


226
Method A with Int. 14
Method 6 with Int. 149


227
Method A with Int. 14
Method 6 with Int. 144


228
Method A with Int. 15
Method 6


229
Method A with Int. 15
Method 6 with Int. 149


230
Method A with Int. 15
Method 6 with Int. 144


231
Method A with Int. 30
Method 4 with Int. 102


232
Method A with Int. 30
Method 4 with Int. 103


233
Method A with Int. 30
Method 4 with Int. 85


234
Method A with Int. 30
Method 4 with Int. 104


235
Method A with Int. 30
Method 4 with Int. 105


236
Method A with Int. 30
Method 4 with Int. 106


237
Method A with Int. 30
Method 4 with Int. 107


238
Method A with Int. 30
Method 4 with Int. 64


239
Method A with Int. 30
Method 4 with Int. 108


240
Method A with Int. 16
Method 6


241
Method A with Int. 1
Method 7 with Int. 135


242
Method A with Int. 30
Method 4 with Int. 74


243
Method A with Int. 17
Method 6


244
Method A with Int. 18
Method 6


245
Method A with Int. 18
Method 6 with Int. 144


246
Method A with Int. 19
Method 6


247
Method A with Int. 19
Method 6 with Int. 144


248
Method A with Int. 19
Method 6 with Int. 148


249
Method A with Int. 30
Method 4 with Int. 109


250
Method A with Int. 30
Method 4 with Int. 110


251
Method A with Int. 30
Method 4 with Int. 111


252
Method A
Method 4 with Int. 112


253
Method A with Int. 30
Method 4 with Int. 113


254
Method A with Int. 30
Method 4 with Int. 112


255
Method A with Int. 30
Method 4 with Int. 114


256
Method A with Int. 30
Method 4 with Int. 115


257
Method A with Int. 30
Method 4 with Int. 116


258
Method B
Method 1


259
Method A with Int. 20
Method 6


260
Method A with Int. 20
Method 6 with Int. 148


261
Method A with Int. 30
Method 4 with Int. 117


262
Method A with Int. 30
Method 4 with Int. 118


263
Method A with Int. 30
Method 4 with Int. 130


264
Method A with Int. 1
Method 4 with Int. 130


265
Method A with Int. 30
Method 4 with Int. 119


266
Method A with Int. 30
Method 4 with Int. 120


267
Method A with Int. 2, Int. 30, Int. 42
Method 2 with Int. 47


268
Method A with Int. 13
Method 6 with Int. 150


269
Method A with Int. 14
Method 6 with Int. 150


270
Method A with Int. 1, Int. 30
Method 4 with Int.130


27
Method A
Method 4 with Int. 121


272
Method A with Int. 30
Method 4 with Int. 89


273
Method A with Int. 2, Int. 30, Int. 42
Method 2 with Int. 64


274
Method A with Int. 1, Int. 13
Method 6 with Int. 148


275
Method A with Int. 1, Int. 13
Method 6


276
Method A with Int. 1, Int. 30
Method 4 with Int. 133


277
Method A with Int. 1, Int. 30
Method 4 with Int. 122


278
Method B with Int. 31
Method 2 with Int. 64


279
Method A
Method 4 with Int. 123


280
Method A with Int. 1
Method 4 with Int. 133


281
Method A with Int. 1
Method 4 with Int. 112


282
Method A
Method 4 with Int. 112, Int. 39


283
Method A with Int. 30
Method 4 with Int. 112, Int. 39


284
Method A with Int. 1, Int. 30
Method 4 with Int. 50


285
Method A with Int. 5, Int. 30, Int. 43
Method 2 with Int. 52


286
Method A with Int. 5, Int. 30, Int. 43
Method 2 with Int. 47


287
Method A with Int. 5, Int. 30, Int. 43
Method 2 with Int. 59


288
Method A with Int. 1, Int. 30
Method 4 with Int. 40


289
Method A with Int. 30
Method 4 with Int. 40


290
Method A with Int. 1
Method 4 with Int. 123


291
Method A with Int. 1, Int. 30
Method 4 with Int. 123


292
Method A with Int. 30
Method 4 with Int. 123


293
Method A
Method 4 with Int. 124


294
Method A with Int. 1
Method 4 with Int. 124


295
Method A with Int. 1, Int. 30
Method 4 with Int. 124


296
Method A with Int. 30
Method 4 with Int. 124


297
Method A with Int. 30
Method 4 with Int. 125


298
Method B with Int. 30
Method 2 with Int. 64


299
Method A with Int. 30
Method 4 with Int. 148, Int. 64


300
Method A with Int. 1, Int. 30
Method 4 with Int. 112


301
Method A with Int. 30
Method 4 with Int. 150, Int. 64


302
Method A with Int. 1, Int. 22
Method 6


303
Method A
Method 4 with Int. 148, Int. 64


304
Method A
Method 4 with Int. 150, Int. 64


305
Method A with Int. 30
Method 4 with Int. 126


306
Method A
Method 4 with Int. 64


307
Method A with Int. 30
Method 4 with Int. 64


308
Method A with Int. 30
Method 7 with Int. 128


309
Method A with Int. 30
Method 4 with Int. 44


310
Method A
Method 4 with Int. 126


311
Method A with Int. 1, Int. 23
Method 6


312
Method A with Int. 1, Int. 30
Method 7 with Int. 134


313
Method A with Int. 1, Int. 30
Method 7 with Int. 134


314
Method A with Int. 23
Method 6


315
Method A with Int. 1
Method 7 with Int. 134


316
Method A with Int. 1
Method 7 with Int. 134


317
Method A
Method 7 with Int. 134


318
Method A
Method 7 with Int. 134


319
Method A with Int. 24
Method 6


320
Method A with Int. 25
Method 6


321
Method A with Int. 26
Method 6


322
Method A with Int. 27
Method 6


323
Method A with Int. 28
Method 6


324
Method A with Int. 29
Method 6


325
Method A with Int. 1
Method 7 with Int. 135


326
Method A
Method 7 with Int. 135


327
Method A with Int. 1, Int. 30
Method 7 with Int. 135


328
Method A
Method 7 with Int. 135


329
Method A
Method 7 with Int. 135


330
Method A with Int. 1, Int. 26
Method 6


331
Method A with Int. 1, Int. 27
Method 6


332
Method A with Int. 28
Method 6 with Int. 148


333
Method A with Int. 1, Int. 30
Method 4 with Int. 117


334
Method A with Int. 1
Method 4 with Int. 117


335
Method A with Int. 30
Method 4 with Int. 117, Int. 150


336
Method A with Int. 30
Method 4 with Int. 157


337
Method A
Method 9 with Int. 117


338
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 161


339
Method A with Int. 30
Method 9 with Int. 164


340
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 167


341
Method A with Int. 30
Method 4 with Int. 168


342
Method A with Int. 30
Method 9 with Int. 165


343
Method A with Int. 30
Method 4 with Int. 52, Int. 162


344
Method A with Int. 30
Method 9 with Int. 163


345
Method A with Int. 30
Method 9 with Int. 166


346
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 199


347
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 169


348
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 171


349
Method A with Int. 30
Method 9 with Int. 173


350
Method A with Int. 30
Method 4 with Int. 117, Int. 174


351
Method A with Int. 30
Method 9 with Int. 177


352
Method A with Int. 30
Method 7 with Int. 404


353
Method A
Method 9 with Int. 159


354
Method A with Int. 30
Method 9 with Int. 160


355
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 172


356
Method A with Int. 30
Method 9 with Int. 403


357
Method A with Int. 30
Method 9 with Int. 179


358
Method A with Int. 30
Method 9 with Int. 170


359
Method A with Int. 30
Method 9 with Int. 155


360
Method A with Int. 30
Method 9 (Steps 1-4) with Int. 156


361
Method A with Int. 30
Method 9 with Int. 402


362
Method A with Int. 30
Method 9 with Int. 178


363
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 400


364
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 401


365
Method A with Int. 30
Method 4 with Int. 117, Int. 180


366
Method A with Int. 30
Method 9 with Int. 181


367
Method A with Int. 30
Method 9 with Int. 182


368
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 183


369
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 176


370
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 186


371
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 175


372
Method A with Int. 30
Method 7 with Int. 405


373
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 184


374
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 185


375
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 158


376
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 188


377
Method A with Int. 30
Method 4 with Int. 117, Int. 189


378
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 190


379
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 187


380
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 191


381
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 194


382
Method A with Int. 30
Method 10


383
Method A with Int. 30
Method 11


384
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 196


385
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 197


386
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 198


387
Method A with Int. 30, Int. 195
Method 7 (Steps 1-4) with Int. 195, Int. 157


388
Method A with Int. 30, Int. 41
Method 7 (Steps 1-4) with Int. 157


389
Method A with Int. 30
Method 7 (Steps 1-4) with Int. 192


390
Method A with Int. 30
Method 9 with Int. 209


391
Method A
Method 9 with Int. 210


392
Method A with int. 206
Method 12 with Int. 211


393
Method A with int. 206
Method 12 with Int. 212


394
Method A with int. 206
Method 12 with Int. 213


395
Method A with Int. 1, 30
Method 9 with Int. 172


396
Method A with Int. 30
Method 9 with Int. 214


397
Method B with Int. 3, 30
Method 9 with Int. 159


398
Method A with Int. 30
Method 4 with Int. 117, Int. 215


399
Method A with Int. 30
Method 4 with Int. 117, Int. 215


400
Method A with Int. 30
Method 4 with Int. 117, Int. 217


401
Method A
Method 9 with Int. 218


402
Method A with Int. 30
Method 9 with Int. 219


403
Method A with Int. 30
Method 9 With Int. 220


404
Method A with Int. 30, 195
Method 4 with Int. 117


405
Method D
Method 4 with Int. 159


406
Method A with Int. 30
Method 9 with Int. 221


407
Method A with Int. 30
Method 9 with Int. 222


408
Method A with Int. 30
Method 9 with Int. 223


409
Method A with Int. 30
Method 9 with Int. 225


410
Method A with Int. 30
Method 9 with Int. 226


411
Method A with Int. 30
Method 9 with Int. 227


412
Method A with Int. 30
Method 9 with Int. 228


413
Method A with Int. 30
Method 9 with Int. 229


414
Method A with Int. 30
Method 9 with int. 230


415
Method A with Int. 30, 207
Method 4 with int. 117


416
Method A with Int. 30, 207
Method 12 with Int. 236


417
Method A with Int. 30
Method 4 with Int. 231


418
Method A with Int. 30, 207
Method 13 steps 1-5


419
Method A with Int. 30, 207
Method 13


420
Method A with Int. 30, 207
Method 13 with Int. 237


421
Method A with Int. 30, 207
Method 14


422
Method A with Int. 30, 208
Method 4 with Int. 159


423
Method A with int. 206
Method 12 with Int. 232


424
Method A with int. 206
Method 12 with Int. 233


425
Method A with int. 206
Method 12 with Int. 234


426
Method A with int. 206
Method 12 with Int. 235









Table 3, below, provides the expected (Exact Mass) and observed molecular weight for each exemplary compound listed in Table 2.









TABLE 3







Analytical Data for Exemplary Compounds of Table 2











Ex. Number
Exact Mass
Observed m/z















 1
802.32
803.33



 2
846.33
847.49



 3
818.29
819.44



 4
908.32
909.43



 5-P1
872.34
873.46



 5-P2
872.34
873.46



 6-P1
922.34
923.42



 6-P2
922.34
923.42



 7
924.35
925.48



 8
910.34
911.44



 9
874.36
875.5



 10
878.35
879.48



 11
872.34
873.37



 12
922.34
923.38



 13
860.34
861.36



 14
936.35
937.43



 15
940.4
941.43



 16-P1
926.39
927.45



 16-P2
926.39
927.45



 17
970.42
971.42



 18
1011.45
1012.45



 19
998.37
999.34



 20
1013.46
1014.51



 21
972.44
973.49



 22
1004.42
1005.41



 23
1047.41
1048.43



 24
984.44
985.42



 25
938.37
939.5



 26
968.42
969.45



 27
1010.43
1011.48



 28
998.43
999.48



 29
982.43
983.49



 30
980.42
981.49



 31
1025.46
1026.52



 32
974.44
975.51



 33
962.44
963.5



 34
946.45
947.5



 35
944.43
945.49



 36
989.48
990.53



 37
966.46
967.49



 38
964.44
965.51



 39
1009.49
1010.54



 40
930.48
931.53



 41
928.46
929.51



 42
973.51
974.54



 43
1012.44
1013.55



 44
1026.46
1027.61



 45
1013.52
1014.56



 46
952.47
953.52



 47
982.48
983.51



 48
900.37
901.46



 49
984.45
985.5



 50
1027.48
1028.55



 51
986.45
987.49



 52
1029.49
1030.56



 53
1041.46
1042.57



 54
896.32
897.42



 55
898.34
897.42



 56-P1
983.45
984.51



 56-P2
983.45
984.51



 57-P1
1053.49
1054.58



 57-P2
1053.49
1054.58



 58
1037.52
1038.59



 59-P1
1039.54
1040.62



 59-P2
1039.54
1040.62



 60
1041.49
1042.6



 61
930.42
931.48



 62
944.44
945.52



 63
932.44
933.48



 64
946.46
947.5



 65
968.48
969.51



 66
970.43
971.46



 67
996.45
997.49



 68
994.49
995.53



 69
932.49
933.54



 70
934.45
935.51



 71
972.44
973.49



 72
970.43
971.47



 73
996.45
997.53



 74
1013.46
1014.55



 75
1001.41
1002.47



 76
930.47
931.53



 77
966.46
967.51



 78
1008.45
1009.51



 79
1012.44
1013.52



 80
976.46
977.55



 81
972.47
973.51



 82
905.47
906.55



 83
980.48
981.69



 84
1036.5
1037.7



 85
1022.49
1023.64



 86
992.48
993.68



 87
1030.42
1031.58



 88
1006.49
1007.67



 89
1037.46
1038.64



 90
1039.48
1040.68



 91
1053.49
1054.71



 92
982.49
983.67



 93
1038.52
1039.71



 94
1024.5
1025.71



 95
1032.44
1033.62



 96
1008.51
1009.68



 97
1039.48
1040.69



 98
1041.49
1042.71



 99
1055.51
1056.73



100
1047.47
1048.67



101
1039.54
1038.7



102-P1
945.4
946.3



102-P2
945.4
946.3



103
947.41
948.31



104
959.41
960.33



105
961.43
962.36



106
923.43
924.36



107-P1
909.42
910.34



107-P2
909.42
910.34



108
911.43
912.34



109
925.45
926.37



110
1010.46
1011.38



111
1012.48
1013.4



112
1024.48
1025.39



113
1040.47
1041.4



114
1054.49
1055.43



115
948.43
949.36



116
984.41
985.34



117-P1
934.41
935.32



117-P2
934.41
935.32



118-P1
970.39
971.3



118-P2
970.39
971.3



119
927.46
928.44



120
963.45
964.44



121
928.47
929.45



122
915.52
916.48



123
902.45
903.41



124
1053.52
1054.52



125
929.43
930.4



126
943.44
944.45



127
931.44
932.43



128
945.46
946.46



129
947.41
948.39



130
961.43
962.44



131
949.43
950.41



132
893.45
894.43



133
907.46
908.45



134
895.46
895.45



135
909.48
910.48



136-P1
911.43
912.39



136-P2
911.43
912.39



137
925.45
926.44



138
913.45
914.41



139
939.46
940.43



140
975.45
976.44



141
925.45
926.42



142
961.43
962.4



143
1055.51
1056.51



144
1069.52
1070.54



145
1057.49
1058.49



146
988.52
989.52



147
988.52
989.52



148
994.49
995.49



149
1037.52
1038.54



150
1065.55
1066.59



151
1081.55
1082.58



152
982.39
983.32



153
996.41
997.36



154
984.41
985.34



155
998.43
999.39



156-P1
946.41
947.34



156-P2
946.41
947.34



157
960.43
961.36



158
948.43
949.36



159
962.44
963.37



160
972.41
973.35



161
936.43
937.36



162
986.43
987.36



163
950.44
951.38



164
968.48
969.41



165
995.51
996.47



166
1011.51
1012.46



167
970.43
971.46



168
982.43
983.43



169
1037.41
1038.43



170
1022.43
1023.45



171
1023.42
1024.45



172
1021.44
1022.45



173
946.45
947.45



174
976.46
977.47



175
1008.45
1009.47



176
972.47
973.48



177
1035.46
1036.5



178
1021.44
1022.46



179
1021.44
1022.47



180
997.43
998.45



181
1041.44
1042.46



182
1025.46
1026.49



183
978.48
979.49



184
978.48
979.5



185
948.47
949.94



186
958.51
959.67



187
1026.49
1027.66



188
960.47
960.82



189
994.43
995.56



190
1010.46
1010.9



191
1026.46
1027.3



192
981.44
982.16



193
1067.51
1068.73



194
1083.5
1084.68



195
952.44
953.22



196
981.44
982.42



197
1011.45
1012.45



198
999.43
1000.43



199
1011.45
1012.46



200
1020.43
1021.43



201
1059.41
1060.43



202
1027.42
1028.45



203
988.5
989.5



204
936.47
937.45



205
984.45
985.48



206
985.43
986.46



207
1015.42
1016.46



208
997.43
998.46



209
972.44
973.42



210
999.45
1000.47



211
984.45
985.49



212
985.43
986.42



213
980.48
981.51



214
980.48
981.46



215
954.46
955.45



216
1009.44
1010.45



217
997.49
998.5



218
970.43
971.45



219
975.45
976.46



220
939.46
940.48



221
969.48
970.49



222
935.41
936.46



223
929.44
930.49



224
899.43
900.48



225
949.43
950.47



226
943.46
944.49



227
913.45
914.48



228
977.46
978.51



229
971.49
972.52



230
941.48
942.52



231
1094.5
1095.53



232
1021.44
1022.47



233
1026.46
1027.45



234
998.46
999.5



235
998.46
999.51



236
1020.45
1021.47



237
1013.46
1014.47



238
1048.41
1049.41



239
987.47
988.48



240
1012.44
1013.41



241
1067.53
1068.56



242
984.45
985.47



243
1045.49
1046.53



244
969.47
970.49



245
933.49
934.52



246
985.44
986.46



247
949.46
950.49



248
979.47
980.49



249
1035.46
1036.48



250
1035.46
1036.49



251
1034.46
1035.3



252
1058.45
1059.44



253
1088.44
1089.44



254
1074.42
1075.44



255
1074.42
1075.44



256
1083.51
1084.52



257
1076.44
1077.46



258
959.41
960.41



259
974.45
975.46



260
968.48
969.48



261
1065.49
1066.4



262
1058.42
1059.31



263
918.42
919.136



264
888.44
889.37



265
1038.46
1039.37



266
1113.46
1114.36



267
1011.45
1012.36



268
887.43
888.33



269
901.45
902.4



270
904.41
905.52



271
965.46
966.5



272
1036.44
1037.31



273
1032.38
1033.3



274
915.43
916.35



275
921.4
922.31



276
967.42
968.34



277
1041.49
1042.43



278
1030.42
1031.28



279
966.46
967.32



280
951.45
952.33



281
1044.44
1045.27



282
1052.48
1053.32



283
1068.45
1069.28



284
1049.43
1050.25



285
968.42
969.28



286
1011.45
1012.29



287
994.43
995.29



288
1027.48
1028.2



289
1041.49
1042.2



290
952.44
953.32



291
968.42
969.27



292
982.43
983.32



293
980.48
981.35



294
966.46
967.38



295
982.43
983.31



296
996.45
997.31



297
1021.44
1022.28



298
1046.39
1047.25



299
1042.44
1043.25



300
1060.41
1061.21



301
1000.43
1001.2



302
1014.46
1015.12



303
1026.47
1027.23



304
984.46
985.22



305
1021.44
1022.17



306
1031.44
1032.23



307
1047.41
1048.15



308
1013.46
1014.2



309
1049.43
1050.16



310
1005.47
1006.21



311
946.42
947.27



312
1067.51
1068.31



313
1067.51
1068.3



314
960.44
961.29



315
1051.54
1052.35



316
1051.54
1052.33



317
1065.55
1066.34



318
1065.55
1066.37



319
989.46
990.28



320
990.46
991.27



321
962.41
963.25



322
919.42
920.26



323
1001.44
1002.29



324
1018.45
1019.27



325
1067.53
1068.37



326
1067.53
1068.36



327
1083.5
1084.35



328
1081.55
1082.39



329
1081.55
1082.38



330
948.4
949.22



331
905.4
906.26



332
995.47
996.3



333
1051.48
1052.32



334
1035.51
1036.36



335
1017.51
1018.20



336
1078.52
1079.30



337
1049.52
1050.14



338
1060.48
1062.12



339
1109.52
1110.17



340
1080.49
1081.14



341
1064.51
1065.18



342
1051.48
1052.17



343
1002.44
1003.14



344
1079.51
1080.13



345
1099.45
1100.08



346
1120.54
1121.16



347
1106.56
1107.18



348
1083.48
1084.13



349
1084.48
1085.14



350
1047.5
1048.12



351
1075.51
1076.21



352
1128.52
1129.12



353
1062.55
1063.26



354
1129.45
1130.17



355
1077.49
1079.03



356
1166.58
1167.33



357
1090.52
1091.26



358
1077.53
1078.23



359
1079.51
1080.26



360
1092.54
1093.70



361
1095.5
1096.21



362
1104.54
1105.27



363
1091.51
1093.21



364
1120.54
1121.90



365
1011.52
1012.93



366
1093.52
1095.00



367
1095.5
1096.98



368
1155.55
1157.08



369
1077.49
1078.99



370
1095.52
1097.03



371
1093.52
1095.01



372
1158.56
1160.05



373
1109.52
1110.93



374
1092.54
1093.94



375
1057.47
1059.09



376
1082.56
1084.03



377
1021.49
1022.82



378
1102.49
1103.88



379
1090.49
1092.09



380
1143.55
1145.14



381
1118.56
1120.01



382
1066.49
1068.17



383
1133.57
1135.25



384
1106.56
1108.21



385
1073.46
1074.99



386
1101.47
1103.21



387
1092.54
1094.09



388
1092.54
1094.08



389
1093.52
1095.13



390
1106.52
1107.98



391
1063.55
1064.89



392
957.44
958.48



393
957.44
958.38



394
944.4
945.46



395
1063.48
1064.97



396
1094.52
1096



397
1076.51
1077.72



398
981.47
982.73



399
997.51
998.91



400
997.51
998.92



401
1080.54
1081.93



402
1068.48
1069.98



403
1093.52
1094.98



404
1079.51
1080.7



405
1092.54
1093.73



406
1049.5
1050.95



407
1049.5
1050.95



408
1079.47
1080.68



409
1092.54
1093.52



410
1122.55
1123.67



411
1077.53
1078.7



412
1077.53
1078.67



413
1124.55
1125.74



414
1079.51
1080.56



415
1111.5
1112.66



416
1155.51
1156.35



417
1077.53
1078.7



418
1097.48
1098.73



419
1124.53
1125.69



420
1179.57
1180.49



421
1083.5
1084.68



422
1081.54
1082.82



423
939.45
940.41



424
958.43
959.47



425
954.42
955.42



426
910.37
911.44










Table 4, below, provides the full chemical structure for each exemplified compound in Table 2.









TABLE 4







Chemical Structure for Exemplary


Compounds Described in Table 2








Ex.



No.
Chemical Structure











1


embedded image







2


embedded image







3


embedded image







4


embedded image







5- P1/ 5- P2


embedded image







6- P1/ 6- P2


embedded image







7


embedded image







8


embedded image







9


embedded image







10


embedded image







11


embedded image







12


embedded image







13


embedded image







14


embedded image







15


embedded image







16- P1/ 16- P2


embedded image







17


embedded image







18


embedded image







19


embedded image







20


embedded image







21


embedded image







22


embedded image







23


embedded image







24


embedded image







25


embedded image







26


embedded image







27


embedded image







28


embedded image







29


embedded image







30


embedded image







31


embedded image







32


embedded image







33


embedded image







34


embedded image







35


embedded image







36


embedded image







37


embedded image







38


embedded image







39


embedded image







40


embedded image







41


embedded image







42


embedded image







43


embedded image







44


embedded image







45


embedded image







46


embedded image







47


embedded image







48


embedded image







49


embedded image







50


embedded image







51


embedded image







52


embedded image







53


embedded image







54


embedded image







55


embedded image







56- P1/ 56- P2


embedded image







57- P1/ 57- P2


embedded image







58


embedded image







59- P1/ 59- P2


embedded image







60


embedded image







61


embedded image







62


embedded image







63


embedded image







64


embedded image







65


embedded image







66


embedded image







67


embedded image







68


embedded image







69


embedded image







70


embedded image







71


embedded image







72


embedded image







73


embedded image







74


embedded image







75


embedded image







76


embedded image







77


embedded image







78


embedded image







79


embedded image







80


embedded image







81


embedded image







82


embedded image







83


embedded image







84


embedded image







85


embedded image







86


embedded image







87


embedded image







88


embedded image







89


embedded image







90


embedded image







91


embedded image







92


embedded image







93


embedded image







94


embedded image







95


embedded image







96


embedded image







97


embedded image







98


embedded image







99


embedded image







100


embedded image







101


embedded image







102- P1/ 102- P2


embedded image







103


embedded image







104


embedded image







105


embedded image







106


embedded image







107- P1/ 107- P2


embedded image







108


embedded image







109


embedded image







110


embedded image







111


embedded image







112


embedded image







113


embedded image







114


embedded image







115


embedded image







116


embedded image







117- P1/ 117- P2


embedded image







118- P1/ 118- P2


embedded image







119


embedded image







120


embedded image







121


embedded image







122


embedded image







123


embedded image







124


embedded image







125


embedded image







126


embedded image







127


embedded image







128


embedded image







129


embedded image







130


embedded image







131


embedded image







132


embedded image







133


embedded image







134


embedded image







135


embedded image







136- P1/ 136- P2


embedded image







137


embedded image







138


embedded image







139


embedded image







140


embedded image







141


embedded image







142


embedded image







143


embedded image







144


embedded image







145


embedded image







146


embedded image







147


embedded image







148


embedded image







149


embedded image







150


embedded image







151


embedded image







152


embedded image







153


embedded image







154


embedded image







155


embedded image







156- P1/ 156- P2


embedded image







157


embedded image







158


embedded image







159


embedded image







160


embedded image







161


embedded image







162


embedded image







163


embedded image







164


embedded image







165


embedded image







166


embedded image







167


embedded image







168


embedded image







169


embedded image







170


embedded image







171


embedded image







172


embedded image







173


embedded image







174


embedded image







175


embedded image







176


embedded image







177


embedded image







178


embedded image







179


embedded image







180


embedded image







181


embedded image







182


embedded image







183


embedded image







184


embedded image







185


embedded image







186


embedded image







187


embedded image







188


embedded image







189


embedded image







190


embedded image







191


embedded image







192


embedded image







193


embedded image







194


embedded image







195


embedded image







196


embedded image







197


embedded image







198


embedded image







199


embedded image







200


embedded image







201


embedded image







202


embedded image







203


embedded image







204


embedded image







205


embedded image







206


embedded image







207


embedded image







208


embedded image







209


embedded image







210


embedded image







211


embedded image







212


embedded image







213


embedded image







214


embedded image







215


embedded image







216


embedded image







217


embedded image







218


embedded image







219


embedded image







220


embedded image







221


embedded image







222


embedded image







223


embedded image







224


embedded image







225


embedded image







226


embedded image







227


embedded image







228


embedded image







229


embedded image







230


embedded image







231


embedded image







232


embedded image







233


embedded image







234


embedded image







235


embedded image







236


embedded image







237


embedded image







238


embedded image







239


embedded image







240


embedded image







241


embedded image







242


embedded image







243


embedded image







244


embedded image







245


embedded image







246


embedded image







247


embedded image







248


embedded image







249


embedded image







250


embedded image







251


embedded image







252


embedded image







253


embedded image







254


embedded image







255


embedded image







256


embedded image







257


embedded image







258


embedded image







259


embedded image







260


embedded image







261


embedded image







262


embedded image







263


embedded image







264


embedded image







265


embedded image







266


embedded image







267


embedded image







268


embedded image







269


embedded image







270


embedded image







271


embedded image







272


embedded image







273


embedded image







274


embedded image







275


embedded image







276


embedded image







277


embedded image







278


embedded image







279


embedded image







280


embedded image







281


embedded image







282


embedded image







283


embedded image







284


embedded image







285


embedded image







286


embedded image







287


embedded image







288


embedded image







289


embedded image







290


embedded image







291


embedded image







292


embedded image







293


embedded image







294


embedded image







295


embedded image







296


embedded image







297


embedded image







298


embedded image







299


embedded image







300


embedded image







301


embedded image







302


embedded image







303


embedded image







304


embedded image







305


embedded image







306


embedded image







307


embedded image







308


embedded image







309


embedded image







310


embedded image







311


embedded image







312


embedded image







313


embedded image







314


embedded image







315


embedded image







316


embedded image







317


embedded image







318


embedded image







319


embedded image







320


embedded image







321


embedded image







322


embedded image







323


embedded image







324


embedded image







325


embedded image







326


embedded image







327


embedded image







328


embedded image







329


embedded image







330


embedded image







331


embedded image







332


embedded image







333


embedded image







334


embedded image







335


embedded image







336


embedded image







337


embedded image







338


embedded image







339


embedded image







340


embedded image







341


embedded image







342


embedded image







343


embedded image







344


embedded image







345


embedded image







346


embedded image







347


embedded image







348


embedded image







349


embedded image







350


embedded image







351


embedded image







352


embedded image







353


embedded image







354


embedded image







355


embedded image







356


embedded image







357


embedded image







358


embedded image







359


embedded image







360


embedded image







361


embedded image







362


embedded image







363


embedded image







364


embedded image







365


embedded image







366


embedded image







367


embedded image







368


embedded image







369


embedded image







370


embedded image







371


embedded image







372


embedded image







373


embedded image







374


embedded image







375


embedded image







376


embedded image







377


embedded image







378


embedded image







379


embedded image







380


embedded image







381


embedded image







382


embedded image







383


embedded image







384


embedded image







385


embedded image







386


embedded image







387


embedded image







388


embedded image







389


embedded image







390


embedded image







391


embedded image







392


embedded image







393


embedded image







394


embedded image







395


embedded image







396


embedded image







397


embedded image







398


embedded image







399


embedded image







400


embedded image







401


embedded image







402


embedded image







403


embedded image







404


embedded image







405


embedded image







406


embedded image







407


embedded image







408


embedded image







409


embedded image







410


embedded image







411


embedded image







412


embedded image







413


embedded image







414


embedded image







415


embedded image







416


embedded image







417


embedded image







418


embedded image







419


embedded image







420


embedded image







421


embedded image







422


embedded image







423


embedded image







424


embedded image







425


embedded image







426


embedded image











B. Biological Examples
1. Fluorescence Polarization Assay

Binding affinity for the compounds of Formula (I) were determined by Fluorescence Polarization (FP) competitive assay based on previously established protocols (Andrews et. al., Org Biomol Chem., 2004. 2(19):2735-41; Premnath et. al., J Med Chem., 2015. 58(1):433-42.) with modifications as described below. Cyclin/CDK protein complexes were sourced as follows: CyclinA2/CDK2 (CRELUX Protein Services), CyclinB1/CDK1 (Eurofins, discovery. Cat. No. 14-450) and CyclinE1/CDK2 (Eurofins, discovery. Cat. No. 14-475).


FP binding assays were performed in 25 mM HEPES pH 7.5, 100 mM NaCl, 1 mM DTT, 0.01% NP-40 and 1 mg/mL BSA for all 3 protein complexes in black 96-well plates. After experimental plates are set, they were equilibrated by gentle mixing by placing them on an orbital shaker at 100 rpm for 2 hours at rt and then read on a SpectraMax i3X Multi-Mode Microplate Detection platform.


Affinity of the Cyclin/Cdk complexed for the fluorescent labeled probe was determined by adding increasing concentration of each protein complex in buffer containing a carboxyfluorescein labeled probe (FAM probe) at 2 nM (preparation of FAM probe is described below). The half maximal concentration of protein needed for the maximal FP signal were 2 nM for Cyclin A2/Cdk2, 9 nM for Cyclin B1/Cdk1 and 3 nM for Cyclin E1/Cdk2. Methods to prepare the FAM probe are described in the heading below.


The protein concentration used for the competitive FP assays were 8 nM for Cyclin A2/Cdk2 and 10 nM for Cyclin B1/Cdk1 and Cyclin E1/Cdk2 with 2 nM of FAM probe FAM probe. Under these conditions, the dynamic range was about 120 mP 100% binding of FAM probe and complete inhibition of binding by excess of an unlabeled competitor compound, with all experiment showing a Z′ factor >0.80. IC50 for test compounds were determined in eight-point serial dilution dose response curves. Reported IC50 are the average of 2-3 independent experiments. Data from these assays are reported in Table 5.









TABLE 5







Cyclin A, B, and E Activity Data of Exemplary Compounds













Cyclin A
Cyclin B
Cyclin E



Example
FP IC50
FP IC50
FP IC50



Number
(μM)
(μM)
(μM)
















 1
0.21
0.02
0.44



 2
0.11
0.02
0.10



 3
0.62
0.02
0.68



 4
0.09
0.02
0.10



 5-P1
0.79
0.02
0.70



 5-P2
0.56
0.02
0.46



 6-P1
0.16
0.02
0.50



 6-P2
0.10
0.02
0.24



 7
0.06
0.02
0.29



 8
0.03
0.02
0.10



 9
0.25
0.02
0.40



 10
0.10
0.02
0.12



 11
0.17
0.02
0.10



 12
0.06
0.02
0.10



 13
0.10
0.02
0.10



 14
0.10
0.02
0.34



 15
0.28
0.02
1.04



 16-P1
0.74
0.02
3.15



 16-P2
0.60
0.02
2.71



 17
0.09
0.02
4.11



 18
0.09
0.02
6.19



 19
0.43
0.02
20.00



 20
0.02
0.02
1.52



 21
0.02
0.02
1.13



 22
0.21
0.02
20.00



 23
0.12
0.02
5.47



 24
0.06
0.02
1.60



 25
0.04
0.02
0.23



 26
0.06
0.02
1.17



 27
0.09
0.02
1.90



 28
0.04
0.02
0.74



 29
0.03
0.02
0.69



 30
0.24
0.02
5.05



 31
0.03
0.02
2.20



 32
0.05
0.02
0.59



 33
0.02
0.02
0.27



 34
0.02
0.02
0.21



 35
0.12
0.02
1.69



 36
0.03
0.02
0.82



 37
0.05
0.02
0.37



 38
0.18
0.02
1.76



 39
0.06
0.02
0.94



 40
0.05
0.02
0.24



 41
0.16
0.02
0.81



 42
0.04
0.02
0.44



 43
0.04
0.02
0.37



 44
0.03
0.02
0.27



 45
0.10
0.02
0.33



 46
0.02
0.02
0.35



 47
0.04
0.02
0.80



 48
0.08
0.02
0.13



 49
0.02
0.02
0.35



 50
0.02
0.02
1.22



 51
0.02
0.02
0.89



 52
0.04
0.02
1.13



 53
0.09
0.02
12.40



 54
0.39
0.03
3.39



 55
0.70
0.08
3.93



 56-P1
0.86
0.04
20.00



 56-P2
0.59
0.03
20.00



 57-P1
0.02
0.02
1.41



 57-P2
0.02
0.02
1.31



 58
0.02
0.02
0.62



 59-P1
0.12
0.02
0.85



 59-P2
0.18
0.02
0.87



 60
0.02
0.02
0.74



 61
0.02
0.02
1.01



 62
0.03
0.02
0.39



 63
0.12
0.02
3.51



 64
0.38
0.02
20.00



 65
0.02
0.02
0.27



 66
0.02
0.02
0.45



 67
0.02
0.02
0.50



 68
0.02
0.02
0.12



 69
0.02
0.02
0.11



 70
0.02
0.02
0.29



 71
0.02
0.02
1.59



 72
0.03
0.02
1.05



 73
0.03
0.02
0.93



 74
0.05
0.02
3.62



 75
0.02
0.02
0.44



 76
0.02
0.02
0.88



 77
0.02
0.02
1.71



 78
0.04
0.02
0.64



 79
0.05
0.02
0.63



 80
0.04
0.02
0.30



 81
0.04
0.02
0.30



 82
0.02
0.02
0.22



 83
0.04
0.02
0.10



 84
0.05
0.02
0.29



 85
0.06
0.02
0.28



 86
0.06
0.02
0.25



 87
0.18
0.02
0.89



 88
0.05
0.02
0.20



 89
0.12
0.02
5.41



 90
0.09
0.02
3.70



 91
0.10
0.02
5.73



 92
0.02
0.02
0.10



 93
0.02
0.02
0.14



 94
0.02
0.02
0.18



 95
0.05
0.02
0.53



 96
0.02
0.02
0.12



 97
0.02
0.02
1.45



 98
0.02
0.02
1.10



 99
0.02
0.02
1.54



100
0.02
0.02
0.45



101
0.03
0.02
0.47



102-P1
0.31
0.02
1.11



102-P2
0.12
0.02
0.31



103
0.06
0.02
0.35



104
0.16
0.02
0.49



105
0.05
0.02
0.29



106
0.12
0.02
0.24



107-P1
0.25
0.02
0.54



107-P2
0.11
0.02
0.18



108
0.05
0.02
0.18



109
0.04
0.02
0.17



110
0.02
0.02
0.33



111
0.02
0.02
0.28



112
0.02
0.02
0.34



113
0.02
0.02
0.31



114
0.02
0.02
0.40



115
0.04
0.02
0.17



116
0.04
0.02
0.33



117-P1
0.08
0.02
0.34



117-P2
0.06
0.02
0.16



118-P1
0.11
0.02
0.74



118-P2
0.06
0.02
0.30



119
0.03
0.02
0.10



120
0.03
0.02
0.21



121
0.05
0.02
0.13



122
0.02
0.02
0.10



123
0.03
0.02
0.12



124
0.02
0.02
0.54



125
1.01
0.02
1.14



126
0.50
0.02
0.62



127
0.08
0.02
0.23



128
0.08
0.02
0.19



129
0.21
0.02
0.82



130
0.07
0.02
0.31



131
0.03
0.02
0.24



132
0.71
0.02
0.45



133
0.47
0.02
0.34



134
0.10
0.02
0.12



135
0.08
0.02
0.12



136-P1
0.14
0.02
0.32



136-P2
0.08
0.02
0.12



137
0.06
0.02
0.13



138
0.03
0.02
0.11



139
0.02
0.02
0.10



140
0.02
0.02
0.12



141
0.04
0.02
0.10



142
0.04
0.02
0.15



143
0.02
0.02
0.94



144
0.02
0.02
0.84



145
0.02
0.02
1.30



146
0.03
0.02
0.10



147
0.02
0.02
0.10



148
0.03
0.02
0.31



149
0.02
0.02
0.56



150
0.02
0.02
0.21



151
0.02
0.02
0.22



152
1.02
0.02
1.90



153
0.47
0.02
1.00



154
0.15
0.02
0.48



155
0.10
0.02
0.41



156-P1
0.89
0.02
0.83



156-P2
0.35
0.02
0.24



157
0.36
0.02
0.41



158
0.10
0.02
0.21



159
0.08
0.02
0.20



160
0.02
0.02
0.27



161
0.02
0.02
0.12



162
0.02
0.02
0.22



163
0.02
0.02
0.10



164
0.07
0.02
0.93



165
0.11
0.02
5.47



166
0.06
0.02
2.30



167
0.02
0.02
0.28



168
0.02
0.02
0.16



169
0.03
0.02
1.09



170
0.05
0.02
2.22



171
0.08
0.02
4.21



172
0.06
0.02
2.43



173
0.02
0.02
0.10



174
0.02
0.02
0.10



175
0.02
0.02
0.13



176
0.02
0.02
0.10



177
0.02
0.02
0.46



178
0.05
0.02
5.46



179
0.03
0.02
1.13



180
0.03
0.02
1.86



181
0.04
0.02
1.58



182
0.02
0.02
1.17



183
0.03
0.02
0.28



184
0.02
0.02
0.10



185
0.02
0.02
0.11



186
0.02
0.02
0.10



187
0.02
0.02
0.34



188
0.02
0.02
0.23



189
0.05
0.02
0.85



190
0.02
0.02
0.66



191
0.02
0.02
0.26



192
0.02
0.02
1.14



193
0.02
0.02
0.71



194
0.02
0.02
0.54



195
0.04
0.02
0.18



196
0.03
0.02
0.74



197
0.02
0.02
0.31



198
0.05
0.02
1.45



199
0.03
0.02
0.91



200
0.03
0.02
0.89



201
0.03
0.02
1.95



202
0.05
0.02
1.49



203
0.04
0.02
1.26



204
0.02
0.02
0.16



205
0.03
0.02
0.48



206
0.02
0.02
0.20



207
0.05
0.02
2.73



208
0.21
0.02
6.40



209
0.02
0.02
0.54



210
0.42
0.02
20.00



211
0.05
0.02
1.11



212
0.07
0.02
4.16



213
0.03
0.02
0.19



214
0.05
0.02
0.50



215
0.02
0.02
0.20



216
0.03
0.02
0.47



217
0.03
0.02
0.76



218
0.10
0.02
2.22



219
0.10
0.02
1.43



220
0.09
0.02
0.68



221
0.06
0.02
0.18



222
0.04
0.02
0.23



223
0.02
0.02
0.10



224
0.03
0.02
0.10



225
0.03
0.02
0.19



226
0.02
0.02
0.10



227
0.03
0.02
0.12



228
0.03
0.02
0.25



229
0.02
0.02
0.10



230
0.03
0.02
0.14



231
0.06
0.02
2.94



232
0.05
0.02
2.87



233
0.05
0.02
1.60



234
0.03
0.02
0.83



235
0.14
0.02
6.08



236
0.03
0.02
0.78



237
0.02
0.02
1.14



238
0.03
0.02
0.99



239
0.02
0.02
0.42



240
0.02
0.02
0.14



241
0.02
0.02
0.20



242
0.03
0.02
0.70



243
0.05
0.02
0.69



244
0.06
0.02
0.58



245
0.04
0.02
0.22



246
0.02
0.02
0.64



247
0.02
0.02
0.31



248
0.02
0.02
0.10



249
0.02
0.02
0.59



250
0.03
0.02
1.62



251
0.04
0.02
0.93



252
0.03
0.02
0.38



253
0.02
0.02
0.33



254
0.03
0.02
0.97



255
0.02
0.02
0.21



256
0.03
0.02
0.48



257
0.02
0.02
0.12



258
0.07
0.02
0.12



259
0.07
0.02
0.28



260
0.05
0.02
0.10



261
0.02
0.02
0.43



262
0.02
0.02
0.91



263
0.03
0.02
0.20



264
0.09
0.02
0.29



265
0.02
0.02
0.64



266
0.02
0.02
2.22



267
0.02
0.02
1.95



268
0.06
0.02
0.10



269
0.06
0.02
0.10



270
0.06
0.02
0.37



271
0.03
0.02
0.24



272
0.05
0.02
1.60



273
0.05
0.02
1.94



274
0.03
0.02
0.10



275
0.05
0.02
0.23



276
0.03
0.02
0.70



277
0.60
0.02
20.00



278
0.07
0.02
0.28



279
0.10
0.02
1.92



280
0.04
0.02
0.32



281
0.04
0.02
0.53



282
0.03
0.02
0.10



283
0.03
0.02
0.18



284
0.02
0.02
1.74



285
0.06
0.02
1.70



286
0.08
0.02
4.02



287
0.06
0.02
1.94



288
0.03
0.02
0.36



289
0.03
0.02
0.32



290
0.12
0.02
1.79



291
0.07
0.02
5.16



292
0.06
0.02
2.88



293
0.10
0.02
1.99



294
0.08
0.02
1.44



295
0.07
0.02
3.69



296
0.07
0.02
2.81



297
0.04
0.02
1.27



298
0.05
0.02
0.44



299
0.02
0.02
0.13



300
0.03
0.02
0.68



301
0.04
0.02
0.32



302
0.05
0.02
3.56



303
0.03
0.02
0.10



304
0.04
0.02
0.19



305
0.05
0.02
1.75



306
0.02
0.02
0.20



307
0.02
0.02
0.44



308
0.02
0.02
0.84



309
0.02
0.02
0.93



310
0.08
0.02
0.99



311
0.05
0.02
0.20



312
0.02
0.02
0.74



313
0.02
0.02
0.64



314
0.03
0.02
0.14



315
0.03
0.02
0.42



316
0.02
0.02
0.29



317
0.03
0.02
0.25



318
0.02
0.02
0.28



319
0.03
0.02
1.35



320
0.03
0.02
1.45



321
0.02
0.02
0.10



322
0.08
0.02
0.53



323
0.03
0.02
0.24



324
0.06
0.02
0.52



325
0.02
0.02
0.30



326
0.02
0.02
0.15



327
0.02
0.02
0.40



328
0.02
0.02
0.21



329
0.02
0.02
0.25



330
0.03
0.02
0.10



331
0.17
0.02
1.14



332
0.02
0.02
0.10



333
0.02
0.02
1.15



334
0.02
0.02
0.19



335
0.02
0.02
0.16



336
0.02
0.02
0.42



337
0.02
0.02
0.19



338
0.03
0.02
1.70



339
0.02
0.02
0.40



340
0.02
0.02
1.65



341
0.02
0.02
0.51



342
0.02
0.02
0.59



343
0.02
0.02
0.67



344
0.02
0.02
1.27



345
0.03
0.02
3.05



346
0.02
0.02
0.36



347
0.02
0.02
0.65



348
0.02
0.02
0.88



349
0.02
0.02
0.23



350
0.02
0.02
0.88



351
0.02
0.02
0.97



352
0.02
0.02
0.51



353
0.02
0.02
0.22



354
0.03
0.02
1.34



355
0.02
0.02
1.76



356
0.02
0.02
0.34



357
0.03
0.02
1.97



358
0.02
0.02
0.55



359
0.02
0.02
0.68



360
0.02
0.02
0.86



361
0.05
0.02
1.84



362
0.02
0.02
0.60



363
0.02
0.02
0.47



364
0.02
0.02
0.43



365
0.02
0.02
1.17



366
0.02
0.02
1.05



367
0.02
0.02
1.10



368
0.02
0.02
0.48



369
0.02
0.02
1.49



370
0.02
0.02
0.78



371
0.02
0.02
0.92



372
0.02
0.02
0.43



373
0.02
0.02
0.48



374
0.02
0.02
0.87



375
0.02
0.02
1.73



376
0.02
0.02
1.33



377
0.02
0.02
0.34



378
0.02
0.02
1.12



379
0.03
0.02
2.73



380
0.03
0.02
2.17



381
0.02
0.02
1.31



382
0.04
0.02
2.49



383
0.02
0.02
0.53



384
0.02
0.02
0.59



385
0.02
0.02
1.63



386
0.02
0.02
0.90



387
0.03
0.02
1.89



388
0.02
0.02
3.93



389
0.02
0.02
0.79



390
0.02
0.02
0.87



391
0.03
0.02
0.61



392
0.23
0.02
0.83



393
6.67
0.047
1.69



394
0.15
0.02
0.50



395
0.06
0.02
2.81



396
0.03
0.02
2.53



397
0.02
0.02
0.26



398
0.02
0.02
0.59



399
0.07
0.02
3.00



400
0.02
0.02
0.49



401
0.02
0.02
0.38



402
0.02
0.02
0.96



403
0.02
0.02
0.67



404
0.02
0.02
1.53



405
0.05
0.02
2.56



406
0.02
0.02
1.78



407
0.02
0.02
1.46



408
0.02
0.02
0.51



409
0.02
0.02
0.67



410
0.02
0.02
0.65



411
0.02
0.02
0.76



412
0.02
0.02
1.39



413
0.02
0.02
1.31



414
0.03
0.02
0.94



415
0.02
0.02
0.30



416
0.04
0.02
3.98



417
0.03
0.02
2.65



418
0.12
0.02
5.26



419
0.02
0.02
0.54



420
0.02
0.02
0.19



421
0.02
0.02
0.51



422
0.02
0.02
0.42



423
0.72
0.02
0.71



424
20.00
0.0455
3.82



425
0.15
0.02
0.21



426
0.11
0.02
2.06










Preparation of Fluorescent Probe (FAM Probe)



embedded image


The fluorescent probe was synthesized via solid phase peptide synthesis followed by cyclization, fluorescent labeling, and deprotection in solution.


To load Fmoc-Glycine onto ˜50 mg of CTC resin, Fmoc-Glycine (G), CAS #29022-11-5, (4 equiv.) was dissolved in 1.0 mL of anhydrous NMP. Neat DIEA (8 equiv.) was added to the Fmoc-amino acid solution. The solution was dispensed in a peptide reactor vessel containing 50 mg of 2-chlorotrityl chloride (CTC) resin and was agitated for 2 hours at rt. The amino acid solution was drained then the resin was washed with 1.0 mL DMF three times. Unreacted CTC resin was capped with 1.0 mL solution of methanol:DMF (50:50), and DIEA (8 equiv.) for 10 min at rt. The methanol solution was drained then the resin was washed with 1.0 mL DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 10 to 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


A solution of Fmoc-L-2,5-dichlorophenylalanine-OH (25C1F), CAS #1260614-80-9, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 10 to 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


To N-Methylate the amine of 25ClF, 2,6-lutidine (6 equiv.) dissolved in 0.5 mL of anhydrous DCE was added to the resin. 2-nitrobenzenesulfonyl chloride (5 equiv.) dissolved in 0.5 mL anhydrous toluene was added to the resin and then was agitated at 40 to 45° C. for 10 to 15 min. The mixture was drained, then the resin was washed with 1.0 mL of anhydrous toluene three times. The method was repeated twice. Triphenylphosphine (10 equiv.) dissolved in 0.7 mL anhydrous toluene was added to the resin. Dry methanol (MeOH), (20 equiv.) was added to the resin. Azodicarboxylate (10 equiv.) was added to the resin and the mixture was agitated at 45° C. for 30 min. The mixture was drained and the resin was washed with 1.0 mL of anhydrous DMF three times. Alkylation was repeated twice. The nosyl group was then deprotected. A solution of 2-mercaptoethanol (5 equiv.) and 1,8-Diazabicyclo[5.4.0]undec-7-ene (5 equiv.) in 1.0 mL NMP was added to the resin and the mixture was agitated at 45° C. for 10 min. The mixture was drained and then the resin was washed with 1.0 mL of anhydrous DMF three times. Deprotection was repeated twice.


Fmoc-L-Leucine-OH (L), CAS #35661-60-0 (12 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


Fmoc-L-Lysine(Mtt)-OH (KMtt), CAS #167393-62-6, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


Fmoc-L-Arginine(Pbf)-OH (RPbf), CAS #154445-77-9, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


Fmoc-L-Lysine(Boc)-OH (KBoc), CAS #71989-26-9, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


Fmoc-L-Alanine-OH (A), CAS #35661-39-3, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


Fmoc-L-Histidine(Trt)-OH (HTrt), CAS #109425-51-6, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv.) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained then the resin was washed with 1.0 mL of DMF three times. To remove Fmoc, A mixture of piperidine:DMF (20:80, 1 mL) was added to the resin and agitated for 15 min at rt. The piperidine solution was drained and then the resin was washed with 1.0 mL DMF three times.


Fmoc-6-aminohexanoic acid (Ahx), CAS #88574-06-5, (4 equiv.), HATU (4 equiv.), and DIEA (8 equiv) in 1.0 mL of anhydrous NMP was prepared. The mixture was allowed to pre-activate at rt for 5 min, and then was added to the resin and agitated at 35° C. for 30 min. The mixture was drained and then the resin was washed with 1.0 mL of DMF three times.


To cleave peptide from CTC resin and simultaneously deprotect the Mtt protecting group, approximately 2 mL of a solution of 24% HFIP, 2% TIPS, in DCM was added to the polystyrene resin in a solid phase reaction vessel. The contents were shaken for 1 hour. The cleavage solution was filtered into a 50 mL conical vial. The cleaved resin was washed with an additional 2 mL of DCM and the wash was collected in the conical vial. The solution was evaporated in a Genevac. The linear peptide was purified via reverse-phase HPLC using an Acetonitrile/Water gradient with 0.05% formic acid and the purified fractions were pooled and lyophilized to yield white powder of intermediate X (M/z observed=1968.65 [M+H]+).




embedded image


The linear intermediate X (˜15 mg) was cyclized using a medium volume, T3P solution cyclization method. The deprotected and purified linear product was transferred to a 50 mL conical vial and dissolved in 1 mL NMP followed by the addition of DIEA (0.5 mL) and DCM (35 mL). T3P (3 eqv) was added to the solution and the reaction pH was adjusted to pH 9 via dropwise addition of DIEA. The closed conical vial was agitated at rt for 2 hours at 150 rotations per minute. The solution was concentrated at 45° C. under reduced pressure in a Genevac system. The Fmoc group was then removed with the addition of a 10% of KOH/Water solution (5 mL) heated at 70° C. for 30 min. The resulting LCMS trace revealed that the trityl group had been unexpectedly removed during the cyclization and Fmoc-deprotection steps. The cyclic peptide was then purified via reverse phase HPLC using an Acetonitrile/Water gradient with 0.05% formic acid. The purified fractions were pooled and lyophilized to yield intermediate Y (M/z observed=1485.94 [M+Z]+).




embedded image


The probe was fluorescently labeled via a peptide coupling in solution. A solution of 5-carboxyfluorescein (CAS #76823-03-5, FAM) (4 equiv.), EDC (4 equiv.), HOAt (3.9 equiv.) and DIEA (8 equiv.) in 1.0 mL of anhydrous DCM was prepared. The mixture was allowed to pre-activate at rt for 5 min. Intermediate Y was added to the coupling solution, and the reaction was agitated at rt until starting material was not observed by LCMS, resulting in the formation of Intermediate Z (M/z observed=1844.29 [M+Z]+).




embedded image


The Boc and Pbf protecting groups were removed from the cyclic intermediate Z by dissolving the cyclic peptide in a 1 mL solution of 90% TFA, 5% TIPS, 5% DCM and agitating for 1 hour. The reaction was monitored by LCMS for the disappearance of starting material. Upon completion, the reaction was concentrated. The crude material was co-evaporated with DCE (5 mL×2), and then purified via reverse phase-HPLC to yield fluorescent probe (FAM probe) (M/z observed=1492.14 [M+Z]+, 0.7 mg, 99% purity by HPLC).


2. MIT Proliferation Assay

MTT proliferation assay was used to determine the 50% growth inhibition (GI50) of disclosed compounds. 5×103 cells were seeded into 96 well plates. 24 hours later, cells were dosed with compound in an 8- or 10-point 1:3 serial dilution starting at 10 μM. Cells were exposed to compound for a sufficient time to allow 3-4 cell doublings (3 days (WI-38); 5 days (NCI-H1048 and OVCAR3)). Roscovitine and staurosporine were used as plate controls. At the end of the compound incubation, MTT reagent (TACS MTT Cell Proliferation Assay R&D Systems Catalog #4890-025-K) was added and assay carried out. Results are summarized in Table 6









TABLE 6







Cellular Activity Data of Exemplary Compounds












Example
H1048 GI50
OVCAR3 GI50
WI-38 GI50



Number
(μM)
(μM)
(μM)
















 1
4.00
6.61
14.59



 2
0.38
0.41
20.00



 3
0.46
0.72
20.00



 4
0.12
0.06
20.00



 5-P1
0.39
0.69
20.00



 5-P2
0.29
0.56
20.00



 6-P1
0.05
0.09
16.37



 6-P2
0.04
0.07
15.60



 7
0.02
0.05
20.00



 8
0.02
0.03
20.00



 9
0.08
0.11
20.00



 10
0.02
0.03
20.00



 11
0.08
0.21
20.00



 12
0.03
0.04
20.00



 13
0.07
0.24
20.00



 14
0.10
0.20
20.00



 15
0.18
0.55
7.85



 16-P1
0.34
0.88
20.00



 16-P2
0.35
0.99
20.00



 17
0.24
0.56
17.94



 18
0.17
0.34
9.03



 19
0.55
1.76
20.00



 20
0.02
0.02
11.06



 21
0.01
0.02
6.85



 22
1.10
1.80
20.00



 23
0.18
0.21
20.00



 24
0.09
0.17
20.00



 25
0.03
0.04
8.07



 26
0.11
0.17
8.74



 27
0.09
0.37
11.41



 28
0.08
0.17
20.00



 29
0.04
0.07
20.00



 30
0.13
0.14
20.00



 31
0.08
0.15
20.00



 32
0.14
0.25
11.77



 33
0.09
0.17
15.55



 34
0.06
0.11
13.07



 35
0.17
0.35
12.41



 36
0.11
0.35
15.97



 37
0.04
0.05
20.00



 38
0.06
0.16
20.00



 39
0.09
0.21
20.00



 40
0.06
0.05
15.39



 41
0.10
0.13
20.00



 42
0.07
0.21
20.00



 43
0.03
0.04
8.57



 44
0.02
0.04
6.60



 45
0.02
0.04
20.00



 46
0.72
0.79
20.00



 47
0.03
0.13
12.93



 48
0.04
0.05
15.80



 49
0.01
0.01
11.54



 50
0.01
0.01
20.00



 51
0.01
0.01
18.81



 52
0.01
0.01
13.14



 53
0.18
0.18
4.50



 54
0.47
0.70
20.00



 55
0.56
0.71
20.00



 56-P1
0.47
1.02
5.76



 56-P2
0.19
0.43
3.76



 57-P1
0.01
0.01
13.77



 57-P2
0.01
0.01
16.87



 58
0.01
0.01
20.00



 59-P1
0.02
0.02
20.00



 59-P2
0.06
0.05
20.00



 60
0.01
0.01
5.72



 61
0.02
0.04
20.00



 62
0.06
0.07
20.00



 63
0.11
0.24
3.17



 64
0.51
1.24
20.00



 65
0.01
0.01
20.00



 66
0.01
0.01
12.89



 67
0.01
0.01
20.00



 68
0.01
0.01
12.81



 69
0.01
0.01
17.57



 70
0.01
0.01
10.11



 71
0.07
0.09
13.82



 72
0.04
0.06
14.87



 73
0.04
0.06
3.65



 74
0.11
0.17
20.00



 75
0.01
0.02
5.89



 76
0.08
0.12
20.00



 77
0.06
0.07
20.00



 78
0.03
0.04
4.52



 79
0.03
0.04
20.00



 80
0.03
0.03
11.63



 81
0.04
0.05
4.30



 82
0.03
0.07
20.00



 83
0.02
0.01
20.00



 84
0.02
0.01
20.00



 85
0.02
0.02
20.00



 86
0.02
0.02
20.00



 87
0.05
0.06
20.00



 88
0.03
0.03
7.64



 89
0.31
0.38
4.34



 90
0.26
0.23
4.13



 91
0.23
0.26
20.00



 92
0.01
0.01
20.00



 93
0.01
0.01
20.00



 94
0.01
0.01
20.00



 95
0.01
0.02
20.00



 96
0.01
0.01
8.92



 97
0.02
0.03
20.00



 98
0.02
0.02
16.45



 99
0.02
0.03
20.00



100
0.02
0.02
20.00



101
0.02
0.03
20.00



102-P1
0.35
0.81
20.00



102-P2
0.08
0.13
20.00



103
0.03
0.05
20.00



104
0.16
0.26
20.00



105
0.02
0.03
18.78



106
0.41
0.54
6.39



107-P1
0.70
1.01
5.29



107-P2
0.19
0.32
20.00



108
0.02
0.03
20.00



109
0.02
0.02
9.52



110
0.01
0.01
4.80



111
0.01
0.01
8.13



112
0.01
0.01
20.00



113
0.01
0.01
7.24



114
0.01
0.01
7.19



115
0.07
0.06
11.00



116
0.04
0.05
20.00



117-P1
0.23
0.18
20.00



117-P2
0.09
0.06
20.00



118-P1
0.18
0.17
20.00



118-P2
0.05
0.05
20.00



119
0.25
0.30
8.71



120
0.10
0.13
9.28



121
0.04
0.07
7.61



122
0.05
0.05
20.00



123
0.02
0.02
20.00



124
0.02
0.02
20.00



125
0.49
0.55
20.00



126
0.38
0.31
20.00



127
0.03
0.03
20.00



128
0.03
0.03
20.00



129
1.46
2.58
6.88



130
0.70
1.55
8.99



131
0.29
0.40
20.00



132
0.57
0.38
20.00



133
0.34
0.26
20.00



134
0.03
0.04
20.00



135
0.03
0.02
20.00



136-P1
0.83
1.77
3.96



136-P2
0.45
0.64
6.67



137
0.60
0.76
4.58



138
0.20
0.28
6.67



139
0.07
0.15
6.42



140
0.05
0.13
9.25



141
0.09
0.19
3.39



142
0.11
0.19
10.20



143
0.01
0.02
5.18



144
0.07
0.07
20.00



145
0.07
0.08
10.14



146
0.02
0.02
20.00



147
0.01
0.01
1.24



148
0.01
0.01
20.00



149
0.03
0.04
20.00



150
0.02
0.04
20.00



151
0.01
0.01
20.00



152
0.50
0.45
20.00



153
0.11
0.14
20.00



154
0.02
0.02
20.00



155
0.01
0.02
20.00



156-P1
0.48
0.53
20.00



156-P2
0.09
0.10
20.00



157
0.08
0.14
20.00



158
0.02
0.02
20.00



159
0.01
0.02
9.45



160
0.02
0.02
11.46



161
0.02
0.02
20.00



162
0.01
0.01
20.00



163
0.01
0.01
20.00



164
0.03
0.03
20.00



165
0.34
1.67
20.00



166
0.04
0.10
20.00



167
0.01
0.01
19.74



168
0.01
0.01
13.10



169
0.02
0.03
20.00



170
0.13
0.20
20.00



171
0.15
0.21
20.00



172
0.03
0.06
13.70



173
0.01
0.01
12.74



174
0.01
0.01
6.98



175
0.01
0.01
6.85



176
0.01
0.01
5.48



177
0.01
0.01
8.96



178
1.18
2.57
20.00



179
0.01
0.03
11.61



180
0.73
1.22
20.00



181
0.24
0.42
20.00



182
0.06
0.13
20.00



183
0.01
0.02
5.07



184
0.01
0.01
3.65



185
0.01
0.01
10.35



186
0.01
0.01
8.68



187
0.01
0.01
8.66



188
0.01
0.02
3.08



189
0.13
0.08
3.67



190
0.01
0.01
1.98



191
0.01
0.01
2.05



192
0.02
0.02
3.51



193
0.01
0.02
7.58



194
0.01
0.01
5.61



195
0.01
0.01
20.00



196
0.01
0.01
20.00



197
0.05
0.10
20.00



198
0.05
0.05
20.00



199
0.03
0.05
20.00



200
0.04
0.03
20.00



201
0.06
0.06
20.00



202
0.04
0.06
9.11



203
0.06
0.05
3.70



204
0.01
0.01
20.00



205
0.02
0.03
20.00



206
0.01
0.02
20.00



207
0.03
0.04
4.94



208
0.83
1.96
20.00



209
0.01
0.02
20.00



210
0.10
0.11
20.00



211
0.02
0.03
20.00



212
0.08
0.08
20.00



213
0.01
0.01
20.00



214
0.01
0.01
20.00



215
0.01
0.01
20.00



216
0.01
0.01
15.55



217
0.01
0.02
20.00



218
0.23
0.39
20.00



219
0.04
0.08
14.06



220
0.03
0.07
5.20



221
0.02
0.03
16.64



222
0.01
0.01
20.00



223
0.01
0.01
7.76



224
0.01
0.01
12.41



225
0.02
0.03
8.53



226
0.02
0.03
8.39



227
0.02
0.04
14.09



228
0.06
0.08
9.54



229
0.01
0.02
20.00



230
0.04
0.09
5.05



231
0.02
0.04
13.36



232
0.05
0.08
11.61



233
0.03
0.03
15.15



234
0.01
0.01
10.66



235
0.10
0.09
13.15



236
0.01
0.02
7.33



237
0.01
0.04
20.00



238
0.01
0.02
17.27



239
0.02
0.02
14.31



240
0.01
0.01
8.73



241
0.0
0.01
12.45



242
0.03
0.02
20.00



243
0.05
0.04
13.29



244
0.10
0.04
20.00



245
0.08
0.02
20.00



246
0.03
0.01
20.00



247
0.05
0.04
20.00



248
0.04
0.02
20.00



249
0.01
0.01
20.00



250
0.03
0.03
5.55



251
0.01
0.01
13.92



252
0.01
0.02
20.00



253
0.04
0.02
9.93



254
0.02
0.02
20.00



255
0.04
0.03
15.76



256
0.01
0.01
13.19



257
0.01
0.01
13.26



258
0.07
0.07
14.19



259
0.11
0.11
10.43



260
0.04
0.04
6.39



261
0.01
0.01
14.10



262
0.01
0.02
16.21



263
0.03
0.04
20.00



264
0.04
0.03
20.00



265
0.01
0.02
20.00



266
0.03
0.05
20.00



267
0.05
0.04
20.00



268
0.02
0.01
20.00



269
0.03
0.03
20.00



270
0.08
0.07
13.38



271
0.01
0.01
20.00



272
0.09
0.43
20.00



273
0.03
0.04
20.00



274
0.01
0.01
10.48



275
0.01
0.01
20.00



276
0.02
0.02
23.38



277
0.33
0.36
3.82



278
0.01
0.01
20.00



279
0.08
0.09
20.00



280
0.01
0.01
20.00



281
0.01
0.02
20.00



282
0.01
0.01
20.00



283
0.01
0.01
14.18



284
0.01
0.01
20.00



285
0.05
0.03
20.00



286
0.06
0.05
6.50



287
0.04
0.04
20.00



288
0.02
0.02
13.81



289
0.01
0.01
3.78



290
0.08
0.15
20.00



291
0.12
0.15
20.00



292
0.07
0.11
20.00



293
0.05
0.09
20.00



294
0.08
0.13
20.00



295
0.08
0.16
20.00



296
0.07
0.10
20.00



297
0.02
0.03
9.15



298
0.01
0.02
20.00



299
0.01
0.01
14.29



300
0.03
0.04
20.00



301
0.02
0.02
20.00



302
0.14
0.12
20.00



303
0.01
0.01
20.00



304
0.01
0.01
20.00



305
0.01
0.02
19.20



306
0.01
0.01
20.00



307
0.01
0.01
20.00



308
0.01
0.01
20.00



309
0.01
0.01
20.00



310
0.01
0.01
20.00



311
0.02
0.03
20.00



312
0.02
0.03
20.00



313
0.01
0.01
36.59



314
0.01
0.01
20.00



315
0.02
0.02
20.00



316
0.01
0.01
20.00



317
0.01
0.01
20.00



318
0.01
0.01
9.92



319
0.01
0.02
20.00



320
0.01
0.02
15.50



321
0.01
0.01
89.76



322
0.03
0.06
20.00



323
0.01
0.01
20.00



324
0.04
0.08
20.00



325
0.01
0.01
20.00



326
0.01
0.01
20.00



327
0.01
0.01
83.88



328
0.01
0.01
20.00



329
0.01
0.01
20.00



330
0.01
0.01
18.46



331
0.06
0.05
20.00



332
0.01
0.01
20.00



333
0.01
0.01
20.00



334
0.01
0.01
20.00



335
0.01
0.01
4.75



336
0.01
0.01
2.48



337
0.01
0.01
18.7



338
0.02
0.02
18.9



339
0.01
0.01
5.62



340
0.02
0.02
17.3



341
0.01
0.01
2.44



342
0.01
0.01
20



343
0.01
0.01
20



344
0.01
0.02
20



345
0.02
0.04
20



346
0.01
0.01
3.68



347
0.01
0.01
1.81



348
0.01
0.01
18



349
0.01
0.01
20



350
0.01
0.01
20



351
0.01
0.01
1.78



352
0.01
0.01
20



353
0.01
0.01
3.49



354
0.01
0.02
20



355
0.01
0.01
2.77



356
0.01
0.01
3.43



357
0.02
0.05
3.14



358
0.01
0.01
2.13



359
0.01
0.01
4.56



360
0.01
0.01
2.72



361
0.03
0.01
14.3



362
0.01
0.01
1.28



363
0.01
0.01
1.43



364
0.01
0.01
3.25



365
0.01
0.01
3.18



366
0.02
0.02
20



367
0.02
0.01
14.7



368
0.01
0.01
15.2



369
0.02
0.02
4.54



370
0.02
0.02
1.21



371
0.01
0.01
1.44



372
0.01
0.01
2.84



373
0.01
0.01
20



374
0.01
0.01
2.22



375
0.01
0.02
16.8



376
0.01
0.01
3.45



377
0.01
0.01
8.16



378
0.01
0.02
20



379
0.03
0.02
20



380
0.02
0.03
1.17



381
0.02
0.04
2.3



382
0.09
0.04
20



383
0.01
0.01
2.7



384
0.01
0.01
2.21



385
0.03
0.02
20



386
0.03
0.02
20



387
0.03
0.02
3.08



388
0.02
0.02
3.5



389
0.02
0.01
17.6



390
0.03
0.02
9.24



391
0.01
0.01
7.82



392
0.17
0.17
14.70



393
1.79
1.31
13.70



394
0.09
0.05
20.00



395
0.02
0.01



396
0.02
0.01



397
0.01
0.01
3.56



398
0.14
0.19
3.64



399
0.22
0.11
3.65



400
0.01
0.01
4.61



401
0.01
0.01



402
0.04
0.01
20.00



403
0.01
0.01
3.99



404
0.02
0.03
20.00



405
0.04
0.05
2.16



406
0.02
0.04
3.35



407
0.01
0.02
3.01



408
0.01
0.01
5.53



409
0.01
0.01
2.46



410
0.01
0.01
2.00



411
0.01
0.01
1.61



412
0.01
0.01
1.06



413
0.01
0.01
1.60



414
0.01
0.01
20.00



415
0.01
0.01
14.10



416
0.03
0.04
20.00



417
0.01
0.01
1.16



418
1.42
1.15
20.00



419
0.02
0.02
1.99



420
0.01
0.02
3.51



421
0.01
0.01
19.10



422
0.01
0.01
2.36



423
0.20
0.07
20.00



424
2.51
1.78
20.00



425
0.16
0.24
20.00



426
0.09
0.15
20.00










Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, one of skill in the art will appreciate that certain changes and modifications may be practiced within the scope of the appended claims. In addition, each reference provided herein is incorporated by reference in its entirety to the same extent as if each reference was individually incorporated by reference. Where a conflict exists between the instant application and a reference provided herein, the instant application shall dominate.

Claims
  • 1. A compound of Formula (I)
  • 2.-3. (canceled)
  • 4. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R3 is (a) C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or C1-6 haloalkyl substituted with 0 to 5 R3a.
  • 5.-6. (canceled)
  • 7. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R3 is (b) C3-7 cycloalkyl substituted with 0 to 3 R3b.
  • 8. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R3 is (b) C3-4 cycloalkyl substituted with 0 to 2 R3b.
  • 9. The compound of claim 7, or the pharmaceutically acceptable salt thereof, wherein each R3b is halo or C1-4 haloalkyl.
  • 10. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R3 is (c) heterocycloalkyl having 3 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 5 R3c.
  • 11. The compound of claim 10, or the pharmaceutically acceptable salt thereof, wherein each R3c is halo or C1-4 haloalkyl.
  • 12. (canceled)
  • 13. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R3 is
  • 14. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R4a, R4b, and R4c are each independently H or C1-6 alkyl;alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 4 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein the heterocycloalkyl is substituted with 0 to 2 R4a1; andeach R4a1 is independently halo.
  • 15. (canceled)
  • 16. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R4b is H or C1-6 alkyl; andR4c and R4a together with the carbon and nitrogen to which each is attached combine to form a heterocycloalkyl having 4 to 6 ring members, wherein the heterocycloalkyl is substituted with 0 to 3 R4a1.
  • 17. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R4a and R4b are each H; andR4c is ethyl;alternatively, R4c and R4a together with the carbon and nitrogen to which each is attached combine to form a pyrrolidinyl, substituted with 0 to 2 fluoro.
  • 18. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R5a is H or C1-6 alkyl;R5b and R5c are each independently H, C1-6 alkyl, —C1-6 alkyl-OH, C2-6 alkoxyalkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or C1-4 alkyl-C3-6 cycloalkyl, wherein each cycloalkyl is substituted with 0 to 3 R5b5; andeach R5b5 is independently C1-4 alkyl, halo, or C1-4 haloalkyl.
  • 19. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R5a is H or C1-6 alkyl;R5b is H; andR5c is C3-6 cycloalkyl or C1-4 alkyl-C3-6 cycloalkyl.
  • 20. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R5a and R5b are each H; andR5c is H, methyl, ethyl,
  • 21. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein X6 is C6-7 alkylene.
  • 22. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein X6 is C6-7 alkenylene.
  • 23. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein X6 is
  • 24. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein X6 is
  • 25. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein X6 is
  • 26. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R6a is H, C1-4 alkyl, or —C1-4 alkyl-C3-6 cycloalkyl; andR6b and R6d are each independently H or C1-6 alkyl.
  • 27. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R6a is H, methyl, ethyl, or
  • 28. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R7a and R7b are each independently H or C1-6 alkyl; andR7c is C1-6 alkyl, C2-6 alkenyl, C1-6 haloalkyl, C1-6 alkyl-OH, or —C1-6 alkyl-C3-6 cycloalkyl.
  • 29. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R7a and R7b are each H; andR7c is C1-6 alkyl.
  • 30. (canceled)
  • 31. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R7a and R7b are each H; andR7c is isobutyl.
  • 32. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R8a, R8b, R8d, and R8e are each independently H or C1-6 alkyl.
  • 33. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R8a is H or methyl; andR8b, R8d and R8e are each H.
  • 34. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein ring B is phenyl.
  • 35. (canceled)
  • 36. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein ring B is a heteroaryl having 5 to 6 ring members and 1 to 3 heteroatoms, wherein each heteroatom is N.
  • 37. (canceled)
  • 38. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein ring B is pyrid-3-yl.
  • 39. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein the subscript m8 is 1, 2, or 3.
  • 40. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein at least one R8f is halo.
  • 41. (canceled)
  • 42. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f is independently halo, C3-6 cycloalkyl, —X8f—C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl, —X8f-heterocycloalkyl, phenyl, —X8f-phenyl, heteroaryl, or —X8f-heteroaryl, wherein each heterocycloalkyl has 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3;each X8f is independently C1-6 alkylene, C2-6 alkenylene, O, or S; andeach R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3.
  • 43. The compound of claim 42, or the pharmaceutically acceptable salt thereof, wherein each X8f is.
  • 44. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f is independently halo, C3-6 cycloalkyl, —CH═CR8f1R8f2, heterocycloalkyl, phenyl, or heteroaryl, wherein each heterocycloalkyl has 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, and each heteroaryl has 5 to 10 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, heterocycloalkyl, phenyl, and heteroaryl is substituted with 0 to 3 R8f3;each R8f1 and R8f2 are combined with the carbon to which they are attached to form a heterocycloalkyl having 3 to 10 ring members and 1 to 3 heteroatoms, each independently N, O or S, wherein the heterocycloalkyl is substituted with 0 to 3 R8f3.
  • 45. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f is independently C1-4 alkyl, C2-4 alkenyl, C1-4 alkoxy, C1-4 deuteroalkoxy, halo, C1-4 haloalkyl, cyano, or —C1-2 alkyl-cyano.
  • 46. (canceled)
  • 47. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f is independently halo, C3-6 cycloalkyl, —O—C3-6 cycloalkyl, heteroaryl, or —O-heteroaryl, wherein each heteroaryl has 5 to 6 ring members and 1 to 3 heteroatoms, each independently N, O, or S, wherein each cycloalkyl, and heteroaryl is substituted with 0 to 3 R8f3.
  • 48. (canceled)
  • 49. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f3 is independently C1-6 alkyl, —Y8—C1-6 alkyl, C1-6 deuteroalky, —Y8—C1-6 deuteroalkyl, —OH, —C1-6 alkyl-OH, —Y8—C1-6 alkyl-OH,—C1-6 alkyl-Y8—C1-6 alkyl, halo, C1-6 haloalkyl, —Y8—C1-6 haloalkyl, or oxo;each Y8 is independently C(O), C(O)O, N(R8f4)C(O), O, S, or S(O)2; andeach R8f4 is independently H or C1-6 alkyl.
  • 50. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f3 is independently C1-6 alkyl, C1-6 deuteroalky, —OH, —C1-6 alkyl-OH, halo, C1-6 haloalkyl, or oxo.
  • 51. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f3 is independently C1-6 alkyl or —Y8—C1-6 alkyl.
  • 52. (canceled)
  • 53. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein each R8f3 is independently C3-6 cycloalkyl, —X8f3—C3-6 cycloalkyl, heterocycloalkyl, or —X8f3-heterocycloalkyl, wherein each heterocycloalkyl has 3 to 6 members and 1 to 2 heteroatoms, each independently N, O, S, or S(O)2; andeach X8f3 is independently C1-6 alkylene, C(O), or S(O)2.
  • 54.-57. (canceled)
  • 58. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein m8 is 1, 2, or 3; andeach R8f is independently methyl, isopropyl, isobutyl, methoxy, ethoxy, fluoro, chloro, cyano,
  • 59. The compound of claim 1, or the pharmaceutically acceptable salt thereof, wherein R3 is
  • 60. (canceled)
  • 61. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having Formula (Ic):
  • 62. (canceled)
  • 63. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure of any one of Examples 1-426.
  • 64. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 65. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 66. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 67. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 68. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 69. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 70. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 71. The compound of claim 1, or the pharmaceutically acceptable salt thereof, having the structure
  • 72. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable excipient.
  • 73. A method of treating a cancer mediated at least in part by cyclin A comprising administering a therapeutically effective amount of a compound of claim 1, thereby treating the disorder or condition.
  • 74.-75. (canceled)
CROSS-REFERENCES TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application No. 63/467,651, filed May 19, 2023; U.S. Provisional Application No. 63/612,533, filed Dec. 20, 2023; and U.S. Provisional Application No. 63/637,984, filed Apr. 24, 2024, the disclosure of each is hereby incorporated by reference in their entirety for all purposes.

Provisional Applications (3)
Number Date Country
63467651 May 2023 US
63612533 Dec 2023 US
63637984 Apr 2024 US