MODULATORS OF MAS-RELATED G-PROTEIN RECEPTOR X4 AND RELATED PRODUCTS AND METHODS

Information

  • Patent Application
  • 20240254089
  • Publication Number
    20240254089
  • Date Filed
    September 16, 2021
    3 years ago
  • Date Published
    August 01, 2024
    5 months ago
Abstract
Methods are provided for modulating MRGPR X4 generally, or for treating a MRGPR X4-dependent condition more specifically, by contacting MRGPR X4 or administering to a subject in need thereof, respectively, an effective amount of a compound having structure (I): or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein E, Q, W, Z, R1, R2, R3, and R4 are as defined herein. Pharmaceutical compositions containing such compounds, as well as compounds themselves, are also provided.
Description
BACKGROUND
Technical Field

The invention relates to modulators of the Mas-related G-protein coupled receptor X4, to products containing the same, as well as to methods of their use and preparation.


Description of the Related Art

Mas-related G protein receptors (MRGPRs) are a group of orphan receptors with limited expression in very specialized tissues. Very little is known about the function of most of these receptors. There are eight related receptors in this class expressed in humans, only four of which have readily identifiable orthologs in other species (i.e., MRGPR D, E, F and G). The other four receptors (MRGPR X1, X2, X3 and X4) have no counterpart, based on homology, in species other than human. Considerable differences exist in the MRGPR family of receptors between humans and non-clinical species (18 genes and pseudogenes exist in humans and ˜50 in mice), resulting in considerable challenges to both pharmacodynamic and on-target safety characterizations. Mouse MRGPRa1 and Monkey MRGPRX3-like or MRGPRX8 receptors are proposed to be putative orthologs of the human MRGPRX4 given that they are activated by bilirubin. However, these primate receptors only are likely partial functional orthologs of the human MRGPRX4 receptor as they do not respond to all the human MRGPRX4 agonists.


BRIEF SUMMARY

This invention is based, in part, on the identification that functionally in mice MRGPR A1 corresponds, at least in part, to the human MRGPR X4. These receptors mediate disorders including chronic itch (e.g., pruritus), inflammation disorders, autoimmunity, skin disorders, cardiovascular disease, lung inflammation/COPD, and adverse skin reactions to drugs. More specifically, both MRGPR A1 and MRGPR X4 are expressed in sensory neurons, skin melanocytes, dendritic cells, polymorphonuclear cells, macrophages, bronchial epithelial cells, lung smooth muscle and dorsal root ganglia. It has now been identified that both MRGPR A1 and MRGPR X4 are receptors for (or sensitive to activation by) circulating bilirubin and its metabolites, and thus are important for itch sensation in conditions of elevated bilirubin such as cholestatic pruritus. In addition, MRGPR X4 is activated by multiple additional components of bile including bile acids and metabolites thereof and heme metabolites including bilirubin and urobilin. Bile acids and bilirubin are highly elevated in cholestatic pruritus while urobilin, which is a potent mediator of itch induction in a mouse model, and thus may be important for itch sensation in conditions of elevated urobilin such as uremic pruritus. Furthermore, MRGPR X4 is a receptor for urobilin, which is a potent mediator of itch induction in a mouse model, and thus may be important for itch sensation in conditions of elevated urobilin such as uremic pruritus. Thus, modulating MRGPR X4 allows for treatment of autoimmune diseases such as psoriasis, multiple sclerosis, Steven Johnson's Syndrome, and other chronic itch conditions as explained in more detail below.


Accordingly, in an embodiment, methods are provided for modulating a MRGPR X4 by contacting the MRGPR X4 with an effective amount of a compound having structure (I):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein E, Q, W, Z, R1, R2, R3, and R4 are as defined herein.


In another embodiment, methods are provided for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having structure (I), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.


In more specific embodiments, the MRGPR X4-dependent condition is one or more of an itch associated condition, a pain associated condition, an inflammation-associated condition, or an autoimmune disorder.


In one embodiment, the methods of treating the MRGPR X4-dependent condition are provided which comprise administering an effective amount of a compound of structure (I) with formula (IA), (IB), (IC), (ID), (IE), (IF), (IG) or (IH) as defined herein or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.


In yet another embodiment, pharmaceutical compositions are provided comprising a carrier or excipient and a compound having structure (I), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.


In specific embodiments, pharmaceutical compositions are provided comprising substructures of structure (I) with formula (IA), (IB), (IC), (ID), (IE), (IF), (IG) or (IH) as defined herein or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.


In yet other embodiments, compounds are provided having formula (IA), (IB), (IC), (ID), (IE), (IF), (IG) or (IH) as defined herein, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.


In another embodiment, compounds are provided having one or more of the structures disclosed herein, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.







DETAILED DESCRIPTION

As mentioned above, the invention relates to modulators of the MRGPR X4, to products containing the same, as well as to methods of their use and preparation. This invention is based, in part, on the identification that in mice MRGPR A1 functionally corresponds to the human MRGPR X4. These receptors mediate disorders including chronic and intermittent itch (e.g., pruritus), inflammation disorders, autoimmunity, skin disorders, and adverse skin reactions to drugs and infectious diseases. More specifically, both MRGPR A1 and MRGPR X4 are expressed in sensory neurons and dorsal root ganglia. It has now been identified that both MRGPR A1 and MRGPR X4 are receptors for (or sensitive to activation by) circulating bilirubin and its metabolites, and thus are important for itch sensation in conditions of elevated bilirubin such as cholestatic pruritus and end-stage renal failure. In addition, MRGPR X4 is also activated by bile acids, which are also elevated in cholestatic pruritus. Furthermore, urobilin, an oxidative product of the heme metabolite urobilinogen solely excreted by the kidney, is a potent agonist of MRGPR X4 and pruritogen, and thus may be important for itch sensation in conditions of elevated urobilin such as uremic pruritus, kidney disease and end-stage renal failure. Thus, modulating MRGPR X4 allows for treatment of autoimmune diseases such as psoriasis, multiple sclerosis, Steven Johnson's Syndrome, atopic disorders such as atopic dermatitis and other chronic itch conditions as explained in more detail below.


MRGPRs appear to be sensory receptors that recognize their external environment to exogenous or endogenous signals/chemicals. These receptors likely respond to multiple chemical ligands/agonists. For example, MRGPR X4 recognizes bilirubin, bile acids, and urobilin as agonist signals. In certain embodiments, molecules of this invention modulate MRGPR X4 by functioning as inverse agonists that are capable of blocking multiple chemical entities, and/or as competitive antagonists that can specifically block individual ligands. In one embodiment, such modulations are selective against other MRGPRs, such as MRGPR X1, X2 and/or X3.


Accordingly, in one embodiment a method is provided for modulating a Mas-Related G-Protein Receptor (MRGPR) X4 by contacting the MRGPR X4 with an effective amount of a compound having structure (I):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • W is




embedded image




    • P is C1-C4 alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rp;

    • Q is cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rq;

    • T1 is —C(O)NH—, —N(H)C(O)—, —S(O)2CH2C(O)N(H)—, —C(O)N(H)S(O)2—, —S(O)2N(H)—, or —SCH2C(O)—;

    • T2 is —(C(Rt)(Rt′))—;

    • E is —(C(H)Re)n— or (C(H)Re)n—X2—(C(Rt)(Rt′)p;

    • Z is C or N;

    • X is —X1—(C(Rt)(Rt′)—;

    • X1 is —O—, —NH—, —N(H)C(O)—, —C(O)NH— or —S(═O)2—;

    • X2 is —O—, —C(═O)—, —N(H)C(O)— or —C(O)NH—;

    • Rp and Rq are at each occurrence independently H, —OH, —NH2, —N(Me)2, —NHC(═NH)NH2, —S(═O)2Me, —NHS(═O)2Me, halo, oxo, —CO2H, —C(O)Me, —C(O)NHMe, alkoxy, haloalkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • Re is at each occurrence independently H, OH, or C1-C4 alkyl;

    • Rt is at each occurrence independently H, C1-C4 alkyl, or cycloalkyl;

    • Rt′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or Rt and Rt′ together with the atom to which they are bonded form a ring;

    • R1, R2, R3, and R4 are at each occurrence independently H, —OH, —NH2, halo, —C(O)Me, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • m is 0, 1, 2 or 3;

    • n is 0, 1, 2 or 3; and

    • p is 0 or 1.





As used herein, the following terms have the meaning defined below, unless the context indicates otherwise.


“Modulating” MRGPR X4 means that the compound interacts with the MRGPR X4 in a manner such that it functions as an inverse agonist to the receptor, and/or as a competitive antagonist to the receptor. In one embodiment, such modulation is partially or fully selective against other MRGPRs, such as MRGPR X1, X2 and/or X3.


“MRGPR” refers to one or more of the Mas-related G protein coupled receptors, which are a group of orphan receptors with limited expression in very specialized tissues (e.g., in sensory neurons and dorsal root ganglia) and barrier tissues. There are eight related receptors in this class expressed in humans, only 4 of which have readily identifiable orthologs in other species (i.e., MRGPR D, E, F and G). The other four receptors (MRGPR X1, X2, X3 and X4) have no counterpart, based on homology, in non-human species.


“Effective amount” refers to a quantity of a specified agent sufficient to achieve a desired effect in a subject being treated with that agent. Ideally, an effective amount of an agent is an amount sufficient to inhibit or treat the disease without causing substantial toxicity in the subject. The effective amount of an agent will be dependent on the subject being treated, the severity of the affliction, and the manner of administration of the pharmaceutical composition. Methods of determining an effective amount of the disclosed compound sufficient to achieve a desired effect in a subject will be understood by those of skill in the art in light of this disclosure.


“Alkyl” means a saturated or unsaturated straight chain or branched alkyl group having from 1 to 8 carbon atoms, in some embodiments from 1 to 6 carbon atoms, in some embodiments from 1 to 4 carbon atoms, and in some embodiments from 1 to 3 carbon atoms. Examples of saturated straight chain alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl-, n-hexyl, n-heptyl, and n-octyl groups. Examples of branched alkyl groups include, but are not limited to, isopropyl, iso-butyl, see-butyl, t-butyl, neopentyl, isopentyl, and 2,2-dimethylpropyl groups. An unsaturated alkyl includes alkenyl and alkynyl as defined below.


“Alkenyl” means a straight chain or branched alkenyl group having from 2 to 8 carbon atoms, in some embodiments from 2 to 6 carbon atoms, in some embodiments from 2 to 4 carbon atoms, and in some embodiments from 2 to 3 carbon atoms. Alkenyl groups are unsaturated hydrocarbons that contain at least one carbon-carbon double bond. Examples of lower alkenyl groups include, but are not limited to, vinyl, propenyl, butenyl, pentenyl, and hexenyl.


“Alkynyl” means a straight chain or branched alkynyl group having from 2 to 8 carbon atoms, in some embodiments from 2 to 6 carbon atoms, in some embodiments from 2 to 4 carbon atoms, and in some embodiments from 2 to 3 carbon atoms. Alkynyl groups are unsaturated hydrocarbons that contain at least one carbon-carbon triple bond. Examples of alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl.


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


“Hydroxy” refers to —OH.


“Oxo” refers to ═O.


“Cyano” refers to —CN.


Amino refers to —NH2, -NHalkyl or N(alkyl)2, wherein alkyl is as defined above. Examples of amino include, but are not limited to —NH2, —NHCH3, —N(CH3)2, and the like.


“Haloalkyl” refers to alkyl as defined above with one or more hydrogen atoms replaced with halogen. Examples of lower haloalkyl groups include, but are not limited to, —CF3, —CHF2, and the like.


“Alkoxy” refers to alkyl as defined above joined by way of an oxygen atom (i.e., —O-alkyl). Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, n-butoxy, isopropoxy, see-butoxy, tert-butoxy, and the like.


“Haloalkoxy” refers to haloalkyl as defined above joined by way of an oxygen atom (i.e., —O-haloalkyl). Examples of lower haloalkoxy groups include, but are not limited to, —OCF3, and the like.


“Cycloalkyl” refers to alkyl groups forming a ring structure, which can be substituted or unsubstituted, wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring do not give rise to aromaticity. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 5, 3 to 6, or 3 to 7. Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbornyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like.


“Aryl” groups are cyclic aromatic hydrocarbons that do not contain heteroatoms. Representative aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups. In some embodiments, aryl groups contain 6-14 carbons in the ring portions of the groups. The terms “aryl” and “aryl groups” include fused rings wherein at least one ring, but not necessarily all rings, are aromatic, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like). In one embodiment, aryl is phenyl or naphthyl, and in another embodiment aryl is phenyl.


“Carbocycle” refers to alkyl groups forming a ring structure, which can be substituted or unsubstituted, wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring may give rise to aromaticity. In one embodiment, carbocycle includes cycloalkyl as defined above. In another embodiment, carbocycle includes aryl as defined above.


“Heterocycle” refers to aromatic and non-aromatic ring moieties containing 3 or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, S, or P. In some embodiments, heterocyclyl include 3 to 20 ring members, whereas other such groups have 3 to 15 ring members. At least one ring contains a heteroatom, but every ring in a polycyclic system need not contain a heteroatom. For example, a dioxolanyl ring and a benzdioxolanyl ring system (methylenedioxyphenyl ring system) are both heterocyclyl groups within the meaning herein.


Heterocyclyl groups also include fused ring species including those having fused aromatic and non-aromatic groups. A heterocyclyl group also includes polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl, and also includes heterocyclyl groups that have substituents, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring members. A heterocyclyl group as defined herein can be a heteroaryl group or a partially or completely saturated cyclic group including at least one ring heteroatom. Heterocyclyl groups include, but are not limited to, pyrrolidinyl, furanyl, tetrahydrofuranyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups.


“Heteroaryl” refers to aromatic ring moieties containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S. Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, pyrazinyl, pyrimidinyl, thienyl, triazolyl, tetrazolyl, triazinyl, thiazolyl, thiophenyl, oxazolyl, isoxazolyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, and quinazolinyl groups. The terms “heteroaryl” and “heteroaryl groups” include fused ring compounds such as wherein at least one ring, but not necessarily all rings, are aromatic, including tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolyl, and 2,3-dihydro indolyl.


“Isomer” is used herein to encompass all chiral, diastereomeric or racemic forms of a structure (also referred to as a stereoisomer, as opposed to a structural or positional isomer), unless a particular stereochemistry or isomeric form is specifically indicated. Such compounds can be enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions, at any degree of enrichment. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these are all within the scope of certain embodiments of the invention. The isomers resulting from the presence of a chiral center comprise a pair of nonsuperimposable-isomers that are called “enantiomers.” Single enantiomers of a pure compound are optically active (i.e., they are capable of rotating the plane of plane polarized light and designated R or S).


“Isolated optical isomer” means a compound which has been substantially purified from the corresponding optical isomer(s) of the same formula. For example, the isolated isomer may be at least about 80%, at least 80% or at least 85% pure by weight. In other embodiments, the isolated isomer is at least 90% pure or at least 98% pure, or at least 99% pure by weight.


“Substantially enantiomerically or diastereomerically” pure means a level of enantiomeric or diastereomeric enrichment of one enantiomer with respect to the other enantiomer or diastereomer of at least about 80%, and more specifically in excess of 80%, 85%, 90%, 95%, 98%, 99%, 99.5% or 99.9%.


The terms “racemate” and “racemic mixture” refer to an equal mixture of two enantiomers. A racemate is labeled “(±)” because it is not optically active (i.e., will not rotate plane-polarized light in either direction since its constituent enantiomers cancel each other out). All compounds with an asterisk (*) adjacent to a tertiary or quaternary carbon are optically active isomers, which may be purified from the respective racemate and/or synthesized by appropriate chiral synthesis.


A “hydrate” is a compound that exists in combination with water molecules. The combination can include water in stoichiometric quantities, such as a monohydrate or a dihydrate, or can include water in random amounts. As the term is used herein a “hydrate” refers to a solid form; that is, a compound in a water solution, while it may be hydrated, is not a hydrate as the term is used herein.


A “solvate” is similar to a hydrate except that a solvent other that water is present. For example, methanol or ethanol can form an “alcoholate”, which can again be stoichiometric or non-stoichiometric. As the term is used herein a “solvate” refers to a solid form; that is, a compound in a solvent solution, while it may be solvated, is not a solvate as the term is used herein.


“Isotope” refers to atoms with the same number of protons but a different number of neutrons, and an isotope of a compound of structure (I) includes any such compound wherein one or more atoms are replaced by an isotope of that atom. For example, carbon 12, the most common form of carbon, has six protons and six neutrons, whereas carbon 13 has six protons and seven neutrons, and carbon 14 has six protons and eight neutrons. Hydrogen has two stable isotopes, deuterium (one proton and one neutron) and tritium (one proton and two neutrons). While fluorine has a number of isotopes, fluorine-19 is longest-lived. Thus, an isotope of a compound having the structure of structure (I) includes, but not limited to, compounds of structure (I) wherein one or more carbon 12 atoms are replaced by carbon-13 and/or carbon-14 atoms, wherein one or more hydrogen atoms are replaced with deuterium and/or tritium, and/or wherein one or more fluorine atoms are replaced by fluorine-19.


“Salt” generally refers to an organic compound, such as a carboxylic acid or an amine, in ionic form, in combination with a counter ion. For example, salts formed between acids in their anionic form and cations are referred to as “acid addition salts”. Conversely, salts formed between bases in the cationic form and anions are referred to as “base addition salts.”


The term “pharmaceutically acceptable” refers an agent that has been approved for human consumption and is generally non-toxic. For example, the term “pharmaceutically acceptable salt” refers to nontoxic inorganic or organic acid and/or base addition salts (see, e.g., Lit et al., Salt Selection for Basic Drugs, Int. J. Pharm., 33, 201-217, 1986) (incorporated by reference herein).


Pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal, and transition metal salts such as, for example, calcium, magnesium, potassium, sodium, and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N′dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.


Pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, aromatic aliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, hippuric, malonic, oxalic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, panthothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, Phydroxybutyric, salicylic, -galactaric, and galacturonic acid.


Although pharmaceutically unacceptable salts are not generally useful as medicaments, such salts may be useful, for example as intermediates in the synthesis of compounds having the structure of Formula I, for example in their purification by recrystallization.


One embodiment provides a method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having structure (I):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • W is




embedded image




    • P is C1-C4 alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rp;

    • Q is cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rq;

    • T1 is —C(O)NH—, —N(H)C(O)—, —S(O)2CH2C(O)N(H)—, —C(O)N(H)S(O)2—, —S(O)2N(H)—, or —SCH2C(O)—;

    • T2 is —(C(Rt)(Rt′))—;

    • E is —(C(H)Re)n— or —(C(H)Re)n—X2—(C(Rt)(Rt′)p;

    • Z is C or N;

    • X is —X1—(C(Rt)(Rt′)—;

    • X1 is —O—, —NH—, —N(H)C(O)—, —C(O)NH— or —S(═O)2—;

    • X2 is —O—, —C(═O)—, —N(H)C(O)— or —C(O)NH—;

    • Rp and Rq are at each occurrence independently H, —OH, —NH2, —N(Me)2, —NHC(═NH)NH2, —S(═O)2Me, —NHS(═O)2Me, halo, oxo, —CO2H, —C(O)Me, —C(O)NHMe, alkoxy, haloalkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • Re is at each occurrence independently H, OH or C1-C4 alkyl;

    • Rt is at each occurrence independently H, C1-C4 alkyl or cycloalkyl; Rt′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or Rt and Rt′ together with the atom to which they are bonded form a ring;

    • R1, R2, R3, and R4 are at each occurrence independently H, —OH, —NH2, halo, —C(O)Me, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • m is 0, 1, 2 or 3;

    • n is 0, 1, 2 or 3; and

    • p is 0 or 1.





As used herein, the phrase “MRGPR X4-dependent condition” means a condition where the activation, over sensitization, or desensitization of MRGPR X4 by a natural or synthetic ligand initiates, mediates, sustains, or augments a pathological condition. For example, it is known that some itch or pain sensations are caused by elevated bilirubin and its metabolites or bile acids in patients suffering from pruritus, atopic or other autoimmune or inflammatory diseases. It has been found that MRGPR X4 is sensitive to (or activated by) bilirubin and its metabolites, including urobilin, or bile acids. Without limited by theory, it is to be understood that by modulating MRGPR X4, the itch or pain sensations can be eased.


In some embodiments, the MRGPR X4-dependent condition is a condition that is caused by the activation of MRGPR X4 by a bile acid. As used herein, the term “bile acid” includes primary bile acids (e.g., cholic acid, chenodeoxycholic acid), conjugated bile acids, also referred to as bile salts (e.g., taurocholic acid, glycocholic acid, taurochenodeoxycholic acid, glycochenodeoxycholic acid), secondary bile acids (e.g., deoxycholic acid, lithocholic acid), and bile acid analogs. In some embodiments, a bile acid analog is a farnesoid X-receptor (FXR) agonist. Thus, the compounds of the present disclosure may be used for treating an MRGPR X4 dependent condition caused by activation of MRGPR X4 by a bile acid and that would benefit from modulating MRGPR X4.


In some embodiments, the MRGPR X4-dependent condition is an itch associated condition, a pain associated condition, an autoimmune condition, or an autoimmune or inflammatory disorder.


As used herein, the phrase “itch associated condition” means pruritus (including acute and chronic pruritus) associated with any condition. The itch sensation can originate, e.g., from the peripheral nervous system (e.g., dermal or neuropathic itch) or from the central nervous system (e.g., neuropathic, neurogenic or psychogenic itch). Thus, in one embodiment, the method of present invention is provided to treat an itch associated condition, such as chronic itch; cholestatic pruritus; contact dermatitis; Allergic blepharitis; Anemia; Atopic dermatitis; Bullous pemphigoid; Candidiasis; Chicken pox; Cholestasis; end-stage renal failure; hemodialysis; Contact dermatitis, Atopic Dermatitis; Dermatitis herpetiformis; Diabetes; Drug allergy, Dry skin; Dyshidrotic dermatitis; Ectopic eczema; Erythrasma; Folliculitis; Fungal skin infection; Hemorrhoids; Herpes; HIV infection; Hodgkin's disease; Hyperthyroidism; Iron deficiency anemia; Kidney disease; Leukemia, porphyrias; Liver disease, including primary biliary cholangitis, primary sclerosing cholangitis, Alagille syndrome, Progressive familial intrahepatic cholestasis, Intrahepatic cholestasis of pregnancy, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), biliary atresia, chronic B hepatitis, drug-chronic viral hepatitis, induced liver injury (DILI), liver fibrosis, cholestatic liver disease, and alcoholic liver disease; Lymphoma; Malignancy; Multiple myeloma; Neurodermatitis; Onchocerciasis; Paget's disease; Pediculosis; Polycythemia rubra vera; Lichen Planus; Lichen Sclerosis; Pruritus ani; Pseudorabies; Psoriasis; Rectal prolapse; Scabies; Schistosomiasis; Scleroderma, Severe stress, Stasia dermatitis; Swimmer's itch; Thyroid disease; Tinea cruris; Uremic Pruritus; Rosacea; Cutaneous amyloidosis; Scleroderma; Acne; wound healing; ocular itch; and Urticaria.


As used herein, the phrase “pain associated condition” means any pain due to a medical condition. Thus, in one embodiment, the method of present invention is provided to treat a pain associated condition, such as Acute Pain, Advanced Prostate Cancer, AIDS-Related Pain, Ankylosing Spondylitis, Arachnoiditis, Arthritis, Arthrofibrosis, Ataxic Cerebral Palsy, Autoimmune Atrophic Gastritis, Avascular Necrosis, Back Pain, Behcet's Disease (Syndrome), Burning Mouth Syndrome, Bursitis, Cancer Pain, Carpal Tunnel, Cauda Equina Syndrome, Central Pain Syndrome, Cerebral Palsy, Cervical Stenosis, Charcot-Marie-Tooth (CMT) Disease, Chronic Fatigue Syndrome (CFS), Chronic Functional Abdominal Pain (CFAP), Chronic Pain, Chronic Pancreatitis, Collapsed Lung (Pneumothorax), Complex Regional Pain Syndrome (RSD), Corneal Neuropathic Pain, Crohn's Disease, Degenerative Disc Disease, Dercum's Disease, Dermatomyositis, Diabetic Peripheral Neuropathy (DPN), Dystonia, Ehlers-Danlos Syndrome (EDS), Endometriosis, Eosinophilia-Myalgia Syndrome (EMS), Erythromelalgia, Fibromyalgia, Gout, Headaches, Herniated disc, Hydrocephalus, Intercostal Neuraligia, Interstitial Cystitis, Irritable Bowel syndrome (IBS), Juvenile Dermatositis (Dermatomyositis), Knee Injury, Leg Pain, Loin Pain-Haematuria Syndrome, Lupus, Lyme Disease, Medullary Sponge Kidney (MSK), Meralgia Paresthetica, Mesothelioma, Migraine, Musculoskeletal pain, Myofascial Pain, Myositis, Neck Pain, Neuropathic Pain, Occipital Neuralgia, Osteoarthritis, Paget's Disease, Parsonage Turner Syndrome, Pelvic Pain, Peripheral Neuropathy, Phantom Limb Pain, Pinched Nerve, Polycystic Kidney Disease, Polymyalgia Rhuematica, Polymyositis, Porphyria, Post Herniorraphy Pain Syndrome, Post Mastectomy, Pain Syndrome, Post Stroke Pain, Post Thorocotomy Pain Syndrome, Postherpetic Neuralgia (Shingles), Post-Polio Syndrome, Primary Lateral Sclerosis, Psoriatic Arthritis, Pudendal Neuralgia, Radiculopathy, Raynaud's Disease, Rheumatoid Arthritis (RA), Sacroiliac Joint Dysfunction, Sarcoidosi, Scheuemann's Kyphosis Disease, Sciatica, Scoliosis, Shingles (Herpes Zoster), Sjogren's Syndrome, Spasmodic Torticollis, Sphincter of Oddi Dysfunction, Spinal Cerebellum Ataxia (SCA Ataxia), Spinal Cord Injury, Spinal Stenosis, Syringomyelia, Tarlov Cysts, Transverse Myelitis, Trigeminal Neuralgia, Neuropathic Pain, Ulcerative Colitis, Vascular Pain and Vulvodynia.


As used herein, the term “autoimmune disorder”, or “inflammatory disorder” means a disease or disorder arising from and/or directed against an individual's own tissues or organs, or a co-segregate or manifestation thereof, or resulting condition therefrom. Typically, various clinical and laboratory markers of autoimmune diseases may exist including, but not limited to, hypergammaglobulinemia, high levels of autoantibodies, antigen-antibody complex deposits in tissues, clinical benefit from corticosteroid or immunosuppressive treatments, and lymphoid cell aggregates in affected tissues. Thus, in one embodiment, the method of present invention is provided to treat an autoimmune disorder, such as chronic inflammation, Multiple Sclerosis, Steven Johnson's Syndrome, appendicitis, bursitis, colitis, cystitis, dermatitis, phlebitis, reflex sympathetic dystrophy/complex regional pain syndrome (rsd/crps), rhinitis, tendonitis, tonsillitis, acne vulgaris, reactive airway disorder, asthma, airway infection, autoinflammatory disease, celiac disease, chronic prostatitis, diverticulitis, glomerulonephritis, hidradenitis suppurativa, hypersensitivities, intestinal disorder, epithelial intestinal disorder, inflammatory bowel disease, irritable bowel syndrome, colitis, interstitial cystitis, otitis, pelvic inflammatory disease, endometrial pain, reperfusion injury, rheumatic fever, rheumatoid arthritis, sarcoidosis, transplant rejection, psoriasis, lung inflammation, chronic obstructive pulmonary disease, cardiovascular disease, and vasculitis.


As used herein, the term “administration” refers to providing a compound, or a pharmaceutical composition comprising the compound as described herein. The compound or composition can be administered by another person to the subject or it can be self-administered by the subject. Non-limiting examples of routes of administration are oral, parenteral (e.g., intravenous), or topical.


As used herein, the term “treatment” refers to an intervention that ameliorates a sign or symptom of a disease or pathological condition. As used herein, the terms “treatment”, “treat” and “treating,” with reference to a disease, pathological condition or symptom, also refers to any observable beneficial effect of the treatment. The beneficial effect can be evidenced, for example, by a delayed onset of clinical symptoms of the disease in a susceptible subject, a reduction in severity of some or all clinical symptoms of the disease, a slower progression of the disease, a reduction in the number of relapses of the disease, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease. A prophylactic treatment is a treatment administered to a subject who does not exhibit signs of a disease or exhibits only early signs, for the purpose of decreasing the risk of developing pathology. A therapeutic treatment is a treatment administered to a subject after signs and symptoms of the disease have developed.


As used herein, the term “subject” refers to an animal (e.g., a mammal), such as a human, as well as to animals typically treated in the veterinary context, such as companion animals, livestock, zoo animals or equines. A subject to be treated according to the methods described herein may be one who has been diagnosed with a MRGPR X4 dependent condition, such as an itch associated condition, a pain associated condition, or an autoimmune disorder. Diagnosis may be performed by any method or technique known in the art. One skilled in the art will understand that a subject to be treated according to the present disclosure may have been subjected to standard tests or may have been identified, without examination, as one at risk due to the presence of one or more risk factors associated with the disease or condition. The term “patient” may be used interchangeably with the term “subject.”


A subject may refer to an adult or pediatric subject. The Federal Food, Drug, and Cosmetic Act defines “pediatric” as a subject aged 21 or younger at the time of their diagnosis or treatment. Pediatric subpopulations are further characterized as: (i) neonates from birth through the first 28 days of life; (ii) infants—from 29 days to less than 2 years; (iii) children—2 years to less than 12 years; and (iv) adolescents—aged 12 through 21. Despite the definition, depending on the susceptible patient population and clinical trial evaluation, an approved regulatory label may include phrasing that specifically modifies the range of a pediatric population, such as, for example, pediatric patients up to 22 years of age.


In some embodiments, the subject is a pediatric subject that has Progressive familial intrahepatic cholestasis, Alagille Syndrome, or Biliary Atresia.


In another embodiment, the method of treating a subject having a MRGPR X4-dependent condition (e.g., an itch associated condition, a pain associated condition, an autoimmune condition, or an autoimmune disorder) described herein further comprises administering to the subject a pharmaceutically effective amount of a second therapeutic agent. In one embodiment, the itch associated condition is a liver disease. In one embodiment, the second therapeutic agent is a liver disease therapeutic agent. In one embodiment, the liver disease therapeutic agent is ursodeoxycholic acid (UDCA), norUrsodeoxycholic acid, cholestyramine, stanozolol, naltrexone, rifampicin, Alisol B 23-acetate (AB23A), curcumin, dihydroartemisinin, fenofibrate, bezafibrate, metronidazole, methotrexate, colchicine, metformin, betaine, glucagon, naltrexone, a farnesoid X-receptor (FXR) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a thyroid hormone receptor beta (TRO) agonist, or any combination thereof.


Examples of FXR agonists that may be used in the methods described herein include obeticholic acid, Turofexorate isopropyl (WAY-362450), 3-(2,6-dichlorophenyl)-4-(3′-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064), PX20606 (PX-102), PX-101, INT-767, INT-787, TERN-101, altenusin, tropifexor (LJN452), nidufexor, turofexorate isopropyl, fexaramine, silymarin, silybin, hedragonic acid, cafestol, Cilofexor (GS-9674 or Px-104), EDP-305, BAR704, BAR502, EYP-001, RDX-023, AGN-242266, HPG-1860, MET-409, AGN-242256, EP-024297, IOT-022, M-480, INV-33, RDX023-02, or any combination thereof. In one embodiment, a FXR agonist is a bile acid or analog thereof (e.g., obeticholic acid, INT-767, INT-787, turofexorate isopropyl (WAY-362450), BAR502, hedragonic acid or BAR704) or a non-bile acid agonist (e.g., EDP-305, tropifexor, nidufexor, cilofexor, GW4064, Turofexorate isopropyl, fexaramine, PX20606 (PX-102), TERN-101, altenusin, silymarin, silybin, hedragonic acid, BAR502, EYP-001, RDX023-2, AGN-242266, HPG-1860, MET-409, EP-024297, M-480, or cafestol). In one embodiment, a PPAR agonist is a PPAR-alpha agonist, a PPAR-gamma agonist, a PPAR-delta agonist, a PPAR-alpha/gamma dual agonist, a PPAR alpha/delta dual agonist, a PPAR gamma/delta dual agonist, or PPAR alpha/gamma/delta pan agonist.


Examples of PPAR alpha agonists that may be used in the methods described herein include fenofibrate, ciprofibrate, pemafibrate, gemfibrozil, clofibrate, binifibrate, clinofibrate, clofibric acid, nicofibrate, pirifibrate, plafibride, ronifibrate, theofibrate, tocofibrate, and SRI 0171.


Examples of PPAR gamma agonists that may be used in the methods described herein include rosiglitazone, pioglitazone, deuterium-stabilized R-pioglitazone, efatutazone, ATx08-001, OMS-405, CHS-131, THR-0921, SER-150-DN, KDT-501, GED-0507-34-Levo, CLC-3001, and ALL-4.


Examples of PPAR delta agonists that may be used in the methods described herein include GW501516 (endurabol or ({4-[({4-methyl-2-[4-(trifluoromethyl)phenyl]-1,3-thiazol-5-yl}methyl)sulfanyl]-2-methylphenoxy} acetic acid)), MBX8025 (seladelpar or {2-methyl-4-[5-methyl-2-(4-trifluoromethyl-phenyl)-2H-[1,2,3]triazol-4-ylmethylsylfanyl]-phenoxy}-acetic acid), GW0742 ([4-[[[2-[3-fluoro-4-(trifluoromethyl)phenyl]-4-methyl-5-thiazolyl]methyl]thio]-2-methyl phenoxy] acetic acid), L165041, HPP-593, and NCP-1046.


Examples of PPAR alpha/gamma agonists that may be used in the methods described herein include saroglitazar, aleglitazar, muraglitazar, tesaglitazar, and DSP-8658.


Examples of PPAR alpha/delta agonists that may be used in the methods described herein include elafibranor and T913659.


Examples of PPAR gamma/delta agonists that may be used in the methods described herein include a conjugated linoleic acid (CLA) and T3D-959.


Examples of PPAR alpha/gamma/delta agonists that may be used in the methods described herein include IVA337 (lanifibranor), TTA (tetradecylthioacetic acid), bavachinin, GW4148, GW9135, bezafibrate, lobeglitazone, 2-(4-(5,6-methylenedioxybenzo[d]thiazol-2-yl)-2-methylphenoxy)-2-methylpropanoic acid (MHY2013), and CS038.


Examples of thyroid hormone receptor beta agonists that may be used in the methods described herein include sobetirome, eprotirome, GC-24, MGL-3196, MGL-3745, VK-2809, KB141 [3,5-dichloro-4-(4-hydroxy-3-isopropylphenoxy) phenylacetic acid], and MB07811 (2R,4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4′-hydroxy-3′-isopropylbenzyl)phenoxy)methyl]-2-oxido-[1,3,2]-dioxaphosphonane).


The second therapeutic agent may be administered simultaneously, separately, or sequentially with the compounds of the present disclosure. If administered simultaneously, the second therapeutic agent and compound of the present disclosure may be administered in separate dosage forms or in the same dosage form.


In another embodiment, a method of treating a subject having an itch associated condition is provided, the method comprising administering to the subject a pharmaceutically effective amount of a compound having structure (I) or pharmaceutically acceptable salt or stereoisomer thereof, or a pharmaceutical composition thereof. In one embodiment, the itch associated condition is cholestatic pruritus, uremic pruritus, atopic dermatitis, dry skin, psoriasis, contact dermatitis, or eczema.


Another embodiment provides a method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IA):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is C6-C10 aryl, C3-C10 cycloalkyl, 7-10 membered monocyclic or bicyclic heterocyclyl, 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Ra;
    • B is C6-C10 aryl, C3-C10 cycloalkyl, 3-10 membered monocyclic or bicyclic heterocyclyl, or 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Ra′;
    • La is —C(O)NH—, or —N(H)C(O)—;
    • La′ is a bond, —CH2C(O)— or —(C(R2a)(R2a′)2—.
    • La″ is —(C(R3a)(R3a′))n—;
    • n is 1 or 2;
    • Ra is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R3a is at each occurrence independently H, OH or C1-C4 alkyl;
    • R3a′ is at each occurrence independently H, OH or C1-C4 alkyl;
    • R2a is at each occurrence independently H or C1-C4 alkyl;
    • R2a′ is at each occurrence H;
    • Ra″ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or heteroaryl; and
    • R1A, R2A, R3A, and R4A are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or heteroaryl.


Yet another embodiment provides a method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IB):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is C6-C10 aryl, C3-C1 cycloalkyl, 7-10 membered monocyclic or bicyclic heterocyclyl, 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Rb;
    • B is phenyl or heterocyclyl, optionally substituted with one or more Rb′;
    • Lb is —C(O)NH—;
    • Lb′ is —(C(R2b)(R2b′))—, —(C(R2b)(R2b′))2— or —(C(R2b)(R2b′))3
    • Lb″ is —(CH2)n—;
    • n is 1 or 2;
    • Rb is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R2b is at each occurrence independently H, C1-C4 alkyl or cycloalkyl;
    • R2b′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or R2b and R2b′ together with the atom to which they are bonded form a ring;
    • Rb′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; and
    • R1B, R2B, R3B, and R4B are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl.


In some embodiments a method is provided method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IC):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl, pyridyl or C1-C4 alkyl, each of which is optionally substituted with one or more Rc;
    • B is phenyl or pyrrolidine optionally substituted with one or more Rc′;
    • Rc is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano or C1-C6 alkyl;
    • Rc′ is at each occurrence independently H or chloro; R1c, R2c, R3C, and R4C are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; and
    • n is 1 or 2.


Another embodiment provides a method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (ID):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl, or 5-6 membered heteroaryl, each of which is optionally substituted with one or more Rd;
    • B is phenyl optionally substituted with one or more Rd′;
    • D is N or C;
    • Ld is —C(O)NHS(O)2—;
    • Ld′ is —(C(R2d)(R2d′))n—;
    • Ld″ is —(CH2)—;
    • Rd is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R2d is at each occurrence H;
    • R2d′ is at each occurrence independently H or C1-C4 alkyl;
    • Rd′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • R1D, R2D, R3D, and R4D are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; and n is 0 or 1.


Yet another embodiment provides a method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IE):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl optionally substituted with one or more Re;
    • Re is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano or C1-C6 alkyl; and
    • R1E, R2E, R3E, and R4E are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl.


In some embodiment a method is provided for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IF):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is an isolated pyrrolidine bonded through its nitrogen atom to the carbon chain, and which is optionally substituted with one or more Rf;
    • Rf is H, —OH, halo, alkoxy or C1-C6 alkyl;
    • Rf is chloro or bromo;
    • R1F, R2F, R3F, and R4F are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; and
    • n is 0 or 1.


Yet another embodiment a method as disclosed for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IG):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • R1G is H or fluoro;
    • R2G is H, chloro or fluoro;
    • R3G is H or fluoro;
    • R4G is H;
    • R5G is H or chloro;
    • R6G is H, methoxy or trifluoromethoxy; and
    • R7G is H, chloro, methyl, isopropyl, trifluoromethoxy, trifluoromethyl or difluoromethoxy.


Still another embodiment provides a method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having formula (IH):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • W is




embedded image




    • P is C1-C4 alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rp;

    • Q is cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rq;

    • T1 is —C(O)NH—, —N(H)C(O)—, —S(O)2CH2C(O)N(H)—, —C(O)N(H)S(O)2—, —S(O)2N(H)—, or —SCH2C(O)—;

    • T2 is —(C(Rt)(Rt′))—;

    • E is —(C(H)Re)n— or (C(H)Re)n—X2—(C(Rt)(Rt′);

    • Z is C or N;

    • X is —X1—(C(Rt)(Rt′)p;

    • X1 is —O—, —NH—, —N(H)C(O)—, —C(O)NH— or —S(═O)2—;

    • X2 is —O—, —C(═O)—, —N(H)C(O)—, —C(O)NH—;

    • Rp and Rq are at each occurrence, independently H, —OH, —NH2, N(Me)2, —NHC(═NH)NH2, —S(═O)2Me, —NHS(═O)2Me, halo, oxo, —CO2H, —C(O)Me, —C(O)NHMe, alkoxy, haloalkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • Re is at each occurrence independently H, OH or C1-C4 alkyl;

    • Rt is at each occurrence independently H, C1-C4 alkyl or cycloalkyl; Rt′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or Rt and Rt′ together with the atom to which they are bonded form a ring;

    • R1, R2, R3, and R4 are at each occurrence, independently H, —OH, —NH2, halo, —C(O)Me, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • m is 0, 1, 2 or 3;

    • n is 0, 1, 2 or 3; and

    • p is 0 or 1.





One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having structure (I):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • W is




embedded image




    • P is C1-C4 alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rp;

    • Q is cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rq;

    • T1 is —C(O)NH—, —N(H)C(O)—, —S(O)2CH2C(O)N(H)—, —C(O)N(H)S(O)2—, —S(O)2N(H)—, or —SCH2C(O)—;

    • T2 is —(C(Rt)(Rt′))—;

    • E is —(C(H)Re)n— or —(C(H)Re)n—X2—(C(Rt)(Rt′)p;

    • Z is C or N;

    • X is —X1—(C(Rt)(Rt′)p;

    • X1 is —O—, —NH—, —N(H)C(O)—, —C(O)NH— or —S(═O)2—;

    • X2 is —O—, —C(═O)—, —N(H)C(O)—, —C(O)NH—;

    • Rp and Rq are at each occurrence independently H, —OH, —NH2, N(Me)2, —NHC(═NH)NH2, —S(═O)2Me, —NHS(═O)2Me, halo, oxo, —CO2H, —C(O)Me, —C(O)NHMe, alkoxy, haloalkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • Re is at each occurrence independently H, OH or C1-C4 alkyl;

    • Rt is at each occurrence independently H, C1-C4 alkyl or cycloalkyl;

    • Rt′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or Rt and Rt′ together with the atom to which they are bonded form a ring;

    • R1, R2, R3, and R4 are at each occurrence independently H, —OH, —NH2, halo, —C(O)Me, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • m is 0, 1, 2 or 3;

    • n is 0, 1, 2 or 3; and

    • p is 0 or 1.





Another embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IA):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is C6-C10 aryl, C3-C10 cycloalkyl, 7-10 membered monocyclic or bicyclic heterocyclyl, 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Ra;
    • B is C6-C10 aryl, C3-C10 cycloalkyl, 3-10 membered monocyclic or bicyclic heterocyclyl, or 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Ra′;
    • La is —C(O)NH—, or —N(H)C(O)—;
    • La′ is a bond, —CH2C(O)—, or —(C(R2a)(R2a′)2—.
    • La″ is —(C(R3a)(R3a′)n—;
    • Ra is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R3a is at each occurrence independently H, OH or C1-C4 alkyl;
    • R3a′ is at each occurrence independently H, OH or C1-C4 alkyl;
    • R2a is at each occurrence independently H or C1-C4 alkyl;
    • R2a′ is at each occurrence H;
    • Ra′ is at each occurrence, independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or heteroaryl;
    • R1A, R2A, R3A, and R4A are at each occurrence, independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or heteroaryl;
    • n is 1 or 2;
    • when n is 1, then B is C6-C10 aryl, C3-C10 cycloalkyl, 3-10 membered heterocyclyl, or 5-10 membered monocyclic or bicyclic heteroaryl; or when n is 2, then B is C6-C10 aryl, C3-C10 cycloalkyl, or 5-10 membered monocyclic or bicyclic heteroaryl;
    • with the proviso that when La′ is a bond, then La″ is CH2, A is pyridine or pyrimidine; and Ra at each occurrence cannot both be chloro; and
    • with the proviso that when La′ is —CH2C(O)—, then La″ is CH2 and A is a pyridine or pyrimidine.


Some embodiments provide a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IB):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is C6-C10 aryl, C3-C10 cycloalkyl, 7-10 membered monocyclic or bicyclic heterocyclyl, 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Rb;
    • B is phenyl or heterocyclyl, optionally substituted with one or more Rb′;
    • Lb is —C(O)NH—;
    • Lb′ is —(C(R2b)(R2b′))—, —(C(R2b)(R2b′))2— or —(C(R2b)(R2b′))3
    • Lb″ is —(CH2)n—;
    • Rb is at each occurrence, independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R2b is at each occurrence independently H, C1-C4 alkyl or cycloalkyl;
    • R2b′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or R2b and R2b′ together with the atom to which they are bonded form a ring;
    • Rb′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • R1B, R2B, R3B, and R4B are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 1, when B is phenyl or n is 2, when B is heterocyclyl;
    • when n is 1 and R2b′ is OH, then Lb′ is —(C(R2b)(R2b′))2—;
    • when n is 1, A is oxazole, Lb′ is —(C(R2b)(R2b′))2—, R2b and R2b′ are H, then Rb is not aryl;
    • when n is 1, Lb, is —(C(R2b)(R2b′))3—, then A is phenyl;
    • when n is 1, Lb′ is —(C(R2b)(R2b′))2, R2b and R2b are H, then both A and B cannot both be unsubstituted phenyl; and
    • when n is 2, B cannot be an oxygen containing monocyclic heterocyclyl or a nitrogen containing bicyclic heterocyclyl.


In yet another embodiment is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IC):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl, pyridyl or C1-C4 alkyl, each of which is optionally substituted with one or more Rc;
    • B is phenyl or pyrrolidine optionally substituted with one or more Rc′;
    • Rc is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, or C1-C6 alkyl; Rc′ is at each occurrence independently H, or chloro; R1C, R2C, R3C, and R4C are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 1 or 2;
    • when A and B are both phenyl, then n is 1, Rc′ is chloro, and Rc is selected from the group consisting of a fluoro at the ortho or meta positions, a chloro at the ortho, meta or para positions, a methoxy at the ortho or meta positions, a methyl at the para position, a dichloro at the meta and para positions, or a dichloro at the ortho and para positions;
    • when A is pyridyl, then n is 1 and B is phenyl with a para substituted chloro group;
    • when A is C1-C4 alkyl, then n is 1 and B is phenyl with a para substituted chloro group; and
    • when B is pyrrolidine, then n is 2 and A is phenyl.


One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (ID):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl, or 5-6 membered heteroaryl, each of which is optionally substituted with one or more Rd;
    • B is phenyl optionally substituted with one or more Rd′;
    • D is N or C;
    • Ld is —C(O)NHS(O)2—;
    • Ld′ is —(C(R2d)(R2d′))n—;
    • Ld″ is —(CH2)—;
    • Rd is at each occurrence, independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R2d is at each occurrence H;
    • R2d′ is at each occurrence independently H or C1-C4 alkyl;
    • Rd′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; R1D, R2D, R3D, and R4D are at each occurrence, independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 0 or 1; and
    • when A and B are both phenyl rings; then both phenyl must be substituted.


Another embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IE):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl optionally substituted with one or more Re.
    • Re is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano or C1-C6 alkyl; and
    • R1E, R2E, R3E, and R4E are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl.


Some embodiments provide a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IF):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is an isolated pyrrolidine bonded through its nitrogen atom to the carbon chain, and which is optionally substituted with one or more Rf;
    • Rf is H, —OH, halo, alkoxy or C1-C6 alkyl;
    • Rf′ is chloro or bromo;
    • R1F, R2F, R3F, and R4F are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 0 or 1; and
    • when n is 0, then Rr cannot be chloro.


One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IG):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • R1G is H or fluoro;
    • R2G is H, chloro or fluoro;
    • R3G is H or fluoro;
    • R4G is H;
    • R5G is H or chloro;
    • R6G is H, methoxy or trifluoromethoxy;
    • R7G is H, chloro, methyl, isopropyl, trifluoromethoxy, trifluoromethyl or difluoromethoxy;
    • with the proviso that when R5G is H and R6G is H, then R7G is isopropyl, trifluoromethoxy or difluoromethoxy;
    • when R5G is chloro, then R6G is H and R7G is trifluoromethyl;
    • when R6G is methoxy, then R7G is chloro; or when R6G is trifluoromethoxy then R5G and R7G are both H;
    • when R7G is trifluoromethyl, then R5G is chloro;
    • when R7G is chloro, then R5G is not chloro, and one of R1G or R2G or R3G is fluoro;
    • when R7G is methyl, then R2G is chloro; and
    • when R7G is difluoromethoxy, then R2G is not fluoro.


One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and where the compound of structure (I) has formula (IH):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • W is




embedded image




    • P is C1-C4 alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rp;

    • Q is cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rq;

    • T1 is —C(O)NH—, —N(H)C(O)—, —S(O)2CH2C(O)N(H)—, —C(O)N(H)S(O)2—, —S(O)2N(H)—, or —SCH2C(O)—;

    • T2 is —(C(Rt)(Rt′))—;

    • E is —(C(H)Re)n— or (C(H)Re)n—X2—(C(Rt)(Rt′)p;

    • Z is C or N;

    • X is —X1—(C(Rt)(Rt′)p;

    • X1 is —O—, —NH—, —N(H)C(O)—, —C(O)NH— or —S(═O)2—;

    • X2 is —O—, —C(═O)—, —N(H)C(O)—, —C(O)NH—;

    • Rp and Rq are at each occurrence, independently H, —OH, —NH2, N(Me)2, —NHC(═NH)NH2, —S(═O)2Me, —NHS(═O)2Me, halo, oxo, —CO2H, —C(O)Me, —C(O)NHMe, alkoxy, haloalkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • Re is at each occurrence independently H, OH or C1-C4 alkyl;

    • Rt is at each occurrence independently H, C1-C4 alkyl or cycloalkyl; Rt′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or Rt and Rt′ together with the atom to which they are bonded form a ring; R1, R2, R3, and R4 are at each occurrence, independently H, —OH, —NH2, halo, —C(O)Me, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • m is 0, 1, 2 or 3;

    • n is 0, 1, 2 or 3; and

    • p is 0 or 1.





One embodiment provides a compound having formula (IA):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is C6-C10 aryl, C3-C10 cycloalkyl, 7-10 membered monocyclic or bicyclic heterocyclyl, 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Ra;
    • B is C6-C10 aryl, C3-C10 cycloalkyl, 3-10 membered monocyclic or bicyclic heterocyclyl, or 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Ra′;
    • La is —C(O)NH—, or —N(H)C(O)—;
    • La′ is a bond, —CH2C(O)—, or —(C(R2a)(R2a′)2—.
    • La″ is —(C(R3a)(R3a′))n—;
    • Ra is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R3a is at each occurrence independently H, OH or C1-C4 alkyl;
    • R3a′ is at each occurrence independently H, OH or C1-C4 alkyl;
    • R2a is at each occurrence independently H or C1-C4 alkyl;
    • R2a′ is at each occurrence H;
    • Ra′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or heteroaryl;
    • R1A, R2A, R3A, and R4A are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or heteroaryl; and
    • n is 1 or 2;
    • when n is 1, then B is C6-C10 aryl, C3-C10 cycloalkyl, 3-10 membered heterocyclyl, or 5-10 membered monocyclic or bicyclic heteroaryl; or when n is 2, then B is C6-C10 aryl, C3-C10 cycloalkyl, or 5-10 membered monocyclic or bicyclic heteroaryl;
    • with the proviso that when La′ is a bond, then La″ is CH2, A is pyridine or pyrimidine; and Ra at each occurrence cannot both be chloro; and
    • with the proviso that when La′ is —CH2C(O)—, then La″ is CH2 and A is a pyridine or pyrimidine.


Another embodiment provides a compound having formula (IB).




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is C6-C10 aryl, C3-C10 cycloalkyl, 7-10 membered monocyclic or bicyclic heterocyclyl, 5-10 membered monocyclic or bicyclic heteroaryl, each of which is optionally substituted with one or more Rb;
    • B is phenyl or heterocyclyl, optionally substituted with one or more Rb′;
    • Lb is —C(O)NH—;
    • Lb′ is —(C(R2b)(R2b′))—, —(C(R2b)(R2b′))2— or —(C(R2b)(R2b′))3
    • Lb″ is —(CH2)n—;
    • Rb is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R2b is at each occurrence independently H, C1-C4 alkyl or cycloalkyl;
    • R2b′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or R2b and R2b′ together with the atom to which they are bonded form a ring;
    • Rb′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • R1B, R2B, R3B, and R4B are at each occurrence, independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 1, when B is phenyl or n is 2, when B is heterocyclyl;
    • when n is 1 and R2b′ is OH, then Lb′ is —(C(R2b)(R2b′))2—;
    • when n is 1, A is oxazole, Lb′ is —(C(R2b)(R2b′))2—, R2b and R2b′ are H, then Rb is not aryl;
    • when n is 1, Lb, is —(C(R2b)(R2b′))3—, then A is phenyl;
    • when n is 1, Lb′ is —(C(R2b)(R2b′))2, R2b and R2b′ are H, then both A and B cannot both be unsubstituted phenyl;
    • when n is 1, Lb, is CH2, then A is an unsubstituted pyridyl wherein Rb′ is methyl; and
    • when n is 2, B cannot be an oxygen containing monocyclic heterocyclyl or a nitrogen containing bicyclic heterocyclyl.


Some embodiments provide a compound having formula (IC):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl, pyridyl or C1-C4 alkyl, each of which is optionally substituted with one or more Rc;
    • B is phenyl or pyrrolidine optionally substituted with one or more Rc′;
    • Rc is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano or C1-C6 alkyl;
    • Rc′ is at each occurrence, independently H or chloro;
    • R1c, R2C, R3C, and R4C are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 1 or 2;
    • when A and B are both phenyl, then n is 1, Rc′ is chloro, and Rc is selected from the group consisting of a fluoro at the ortho or meta positions, a chloro at the ortho, meta or para positions, a methoxy at the ortho or meta positions, a methyl at the para position, a dichloro at the meta and para positions, or a dichloro at the ortho and para positions,
    • when A is pyridyl, then n is 1 and B is phenyl with a para substituted chloro group;
    • when A is C1-C4 alkyl, then n is 1 and B is phenyl with a para substituted chloro group; and
    • when B is pyrrolidine, then n is 2 and A is phenyl.


Yet other embodiments provide a compound having formula (ID).




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl, or 5-6 membered heteroaryl, each of which is optionally substituted with one or more Rd;
    • B is phenyl optionally substituted with one or more Rd′;
    • D is N or C;
    • Ld is —C(O)NHS(O)2—;
    • Ld′ is —(C(R2d)(R2d′))n—;
    • Ld″ is —(CH2)—;
    • Rd is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 5-10 membered heteroaryl;
    • R2d is at each occurrence H;
    • R2d′ is at each occurrence independently H or C1-C4 alkyl;
    • Rd′ is at each occurrence independently H, —OH, —NH2, halo, —CO2H, —C(O)Me-alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; R1D, R2D, R3D, and R4D are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl;
    • n is 0 or 1;
    • when A and B are both phenyl rings; then both phenyl must be substituted; and
    • when A is a 5-membered heteroaryl, then the heteroaryl must contain 2 heteroatoms.


One embodiment provides a compound having formula (IE):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is phenyl optionally substituted with one or more Re.
    • Re is at each occurrence independently H, —OH, —NH2, halo, —CO2H, alkoxy, cyano or C1-C6 alkyl; and
    • R1E, R2E, R3E, and R4E are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl.


Another embodiment provides a compound having formula (IF):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • A is an isolated pyrrolidine bonded through its nitrogen atom to the carbon chain, and which is optionally substituted with one or more Rf;
    • Rf is H, —OH, halo, alkoxy or C1-C6 alkyl;
    • Rf′ is chloro or bromo;
    • R1F, R2F, R3F, and R4F are at each occurrence independently H, —OH, —NH2, chloro, fluoro, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C1-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, aryl, cycloalkyl, heterocyclyl or 6-10 membered heteroaryl; and
    • n is 1.


Some embodiments provide a compound having formula (IG):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein:

    • R1G is H or fluoro;
    • R2G is H, chloro or fluoro;
    • R3G is H or fluoro;
    • R4G is H;
    • R5G is H or chloro;
    • R6G is H, methoxy or trifluoromethoxy; and
    • R7G is H, chloro, methyl, isopropyl, trifluoromethoxy, trifluoromethyl or difluoromethoxy;
    • with the proviso that when R5G is H and R6G is H, then R7G is isopropyl, trifluoromethoxy or difluoromethoxy;
    • when R5G is chloro, then R6G is H and R7G is trifluoromethyl;
    • when R6G is methoxy, then R7G is chloro; or when R6G is trifluoromethoxy then R5G and R7G are both H;
    • when R7G is trifluoromethyl, then R5G is chloro;
    • when R7G is chloro, then R5G is not chloro, and one of R1G or R2G or R3G is fluoro;
    • when R7G is methyl, then R2G is chloro; and
    • when R7G is difluoromethoxy, then R2G is not fluoro.


Some embodiments provide a compound having formula (IH):




embedded image


or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, wherein.

    • W is




embedded image




    • P is C1-C4 alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rp;

    • Q is cycloalkyl, aryl, heteroaryl or heterocyclyl, each of which is optionally substituted with one or more Rq;

    • T1 is —C(O)NH—, —N(H)C(O)—, —S(O)2CH2C(O)N(H)—, —C(O)N(H)S(O)2—, —S(O)2N(H)—, or —SCH2C(O)—;

    • T2 is —(C(Rt)(Rt′))—;

    • E is —(C(H)Re)n— or (C(H)Re)n—X2—(C(Rt)(Rt′);

    • Z is C or N;

    • X is —X1—(C(Rt)(Rt′)p;

    • X1 is —O—, —NH—, —N(H)C(O)—, —C(O)NH— or —S(═O)2—;

    • X2 is —O—, —C(═O)—, —N(H)C(O)—, —C(O)NH—;

    • Rp and Rq are at each occurrence, independently H, —OH, —NH2, N(Me)2, —NHC(═NH)NH2, —S(═O)2Me, —NHS(═O)2Me, halo, oxo, —CO2H, —C(O)Me, —C(O)NHMe, alkoxy, haloalkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkyl, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • Re is at each occurrence independently H, OH or C1-C4 alkyl;

    • Rt is at each occurrence independently H, C1-C4 alkyl or cycloalkyl;

    • Rt′ is at each occurrence independently H, OH, C1-C4 alkyl, cycloalkyl or Rt and Rt′ together with the atom to which they are bonded form a ring;

    • R1, R2, R3, and R4 are at each occurrence, independently H, —OH, —NH2, halo, —C(O)Me, —CO2H, alkoxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, hydroxylalkly, alkoxyalkyl, aminylalkyl, alkylaminylalkyl, cyanoalkyl, carboxyalkyl, aminylcarbonylalkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl;

    • m is 0, 1, 2 or 3;

    • n is 0, 1, 2 or 3; and p is 0 or 1.





Representative compounds of structure (I), as well as Formulas (IA) through (IH) as applicable, include any one of the compounds listed in Table A below, as well as a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof. To this end, representative compounds are identified herein by their respective “Compound Number”, which is sometimes abbreviated as “Compound No.” or “Cpd. No.”









TABLE A







Representative Compounds








Cpd.



No.
Structure











1


embedded image







2


embedded image







3


embedded image







4


embedded image







5


embedded image







6


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


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


embedded image







57


embedded image







58


embedded image







59


embedded image







60


embedded image







61


embedded image







62


embedded image







63


embedded image







64


embedded image







65


embedded image







6


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


embedded image







103


embedded image







104


embedded image







105


embedded image







106


embedded image







107


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


embedded image







118


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


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


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







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







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







427


embedded image







428


embedded image







429


embedded image







430


embedded image







431


embedded image







432


embedded image







433


embedded image







434


embedded image







435


embedded image







436


embedded image







437


embedded image







438


embedded image







439


embedded image







440


embedded image







441


embedded image







442


embedded image







443


embedded image







444


embedded image







445


embedded image







446


embedded image







447


embedded image







448


embedded image







449


embedded image







450


embedded image







451


embedded image







452


embedded image







453


embedded image







454


embedded image







455


embedded image







456


embedded image







457


embedded image







458


embedded image







459


embedded image







460


embedded image







461


embedded image







462


embedded image







463


embedded image







464


embedded image







465


embedded image







466


embedded image







467


embedded image







468


embedded image







469


embedded image







470


embedded image







471


embedded image







472


embedded image







473


embedded image







474


embedded image







475


embedded image







476


embedded image







477


embedded image







478


embedded image







479


embedded image







480


embedded image







481


embedded image







482


embedded image







483


embedded image







484


embedded image







485


embedded image







486


embedded image







487


embedded image







488


embedded image







489


embedded image







490


embedded image







491


embedded image







492


embedded image







493


embedded image







494


embedded image







495


embedded image







496


embedded image







497


embedded image







498


embedded image







499


embedded image







500


embedded image







501


embedded image







502


embedded image







503


embedded image







504


embedded image







505


embedded image







506


embedded image







507


embedded image







508


embedded image







509


embedded image







510


embedded image







511


embedded image







512


embedded image







513


embedded image







514


embedded image







515


embedded image







516


embedded image







517


embedded image







518


embedded image







519


embedded image







520


embedded image







521


embedded image







522


embedded image







523


embedded image







524


embedded image







525


embedded image







526


embedded image







527


embedded image







528


embedded image







529


embedded image







530


embedded image







531


embedded image







532


embedded image







533


embedded image







534


embedded image







535


embedded image







536


embedded image







537


embedded image







538


embedded image







539


embedded image







540


embedded image







541


embedded image







542


embedded image







543


embedded image







544


embedded image







545


embedded image







546


embedded image







547


embedded image







548


embedded image







549


embedded image







550


embedded image







551


embedded image







552


embedded image







553


embedded image







554


embedded image







555


embedded image







556


embedded image







557


embedded image







558


embedded image







559


embedded image







560


embedded image







561


embedded image







562


embedded image







563


embedded image







564


embedded image







565


embedded image







566


embedded image







567


embedded image







568


embedded image







569


embedded image







570


embedded image







571


embedded image







572


embedded image







573


embedded image







574


embedded image











In certain embodiments, the invention provides a pharmaceutical composition comprising a compound of structure (I) or any one of formulas (IA) through (IH), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, together with at least one pharmaceutically acceptable carrier, diluent, or excipient. For example, the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which can be in the form of an ampoule, capsule, sachet, paper, or other container. When the active compound is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound. The active compound can be adsorbed on a granular solid carrier, for example contained in a sachet. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid, or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose, and polyvinylpyrrolidone. Similarly, the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.


As used herein, the term “pharmaceutical composition” refers to a composition containing one or more of the compounds described herein, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof, formulated with a pharmaceutically acceptable carrier, which can also include other additives, and manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); for administration to a pediatric subject (e.g., solution, syrup, suspension, elixir, powder for reconstitution as suspension or solution, dispersible/effervescent tablet, chewable tablet, lollipop, freezer pops, troches, oral thin strips, orally disintegrating tablet, orally disintegrating strip, and sprinkle oral powder or granules); or in any other formulation described herein. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington: The Science and Practice of Pharmacy, 21st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005) and in The United States Pharmacopeia: The National Formulary (USP 36 NF31), published in 2013.


In some embodiments, the pharmaceutical composition comprising a compound of structure (I) or any one of formulas (IA) through (IH), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, with at least one pharmaceutically acceptable carrier, diluent, or excipient further comprises a second therapeutic agent. In one embodiment, the second therapeutic agent is a liver disease therapeutic agent. In one embodiment, the liver disease therapeutic agent is ursodeoxycholic acid (UDCA), norUrsodeoxycholic acid, cholestyramine, stanozolol, naltrexone, rifampicin, Alisol B 23-acetate (AB23A), curcumin, dihydroartemisinin, fenofibrate, bezafibrate, metronidazole, methotrexate, colchicine, metformin, betaine, glucagon, naltrexone, a farnesoid X-receptor (FXR) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a thyroid hormone receptor beta (TRO) agonist, or any combination thereof.


Examples of FXR agonists that may be used in the pharmaceutical compositions described herein include obeticholic acid, Turofexorate isopropyl (WAY-362450), 3-(2,6-dichlorophenyl)-4-(3′-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064), PX20606 (PX-102), PX-101, INT-767, INT-787, TERN-101, altenusin, tropifexor (LJN452), nidufexor, turofexorate isopropyl, fexaramine, silymarin, silybin, hedragonic acid, cafestol, Cilofexor (GS-9674 or Px-104), EDP-305, BAR704, BAR502, EYP-001, RDX-023, AGN-242266, HPG-1860, MET-409, AGN-242256, EP-024297, IOT-022, M-480, INV-33, RDX023-02, or any combination thereof.


In one embodiment, an FXR agonist is a bile acid or analog thereof (e.g., obeticholic acid, INT-767, INT-787, turofexorate isopropyl (WAY-362450), BAR502, hedragonic acid or BAR704) or a non-bile acid agonist (e.g., EDP-305, tropifexor, nidufexor, cilofexor, GW4064, Turofexorate isopropyl, fexaramine, PX20606 (PX-102), TERN-101, altenusin, silymarin, silybin, hedragonic acid, BAR502, EYP-001, RDX023-2, AGN-242266, HPG-1860, MET-409, EP-024297, M-480, or cafestol). In one embodiment, a PPAR agonist is a PPAR-alpha agonist, a PPAR-gamma agonist, a PPAR-delta agonist, a PPAR-alpha/gamma dual agonist, a PPAR alpha/delta dual agonist, a PPAR gamma/delta dual agonist, a PPAR alpha/gamma/delta pan agonist, or any combination thereof.


Examples of PPAR alpha agonists that may be used in the pharmaceutical compositions described herein include fenofibrate, ciprofibrate, pemafibrate, gemfibrozil, clofibrate, binifibrate, clinofibrate, clofibric acid, nicofibrate, pirifibrate, plafibride, ronifibrate, theofibrate, tocofibrate, and SRI 0171.


Examples of PPAR gamma agonists that may be used in the pharmaceutical compositions described herein include rosiglitazone, pioglitazone, deuterium-stabilized R-pioglitazone, efatutazone, ATx08-001, OMS-405, CHS-131, THR-0921, SER-150-DN, KDT-501, GED-0507-34-Levo, CLC-3001, and ALL-4.


Examples of PPAR delta agonists that may be used in the pharmaceutical compositions described herein include GW501516 (endurabol or ({4-[({4-methyl-2-[4-(trifluoromethyl)phenyl]-1,3-thiazol-5-yl}methyl)sulfanyl]-2-methylphenoxy} acetic acid)), MBX8025 (seladelpar or {2-methyl-4-[5-methyl-2-(4-trifluoromethyl-phenyl)-2H-[1,2,3]triazol-4-ylmethylsylfanyl]-phenoxy}-acetic acid), GW0742 ([4-[[[2-[3-fluoro-4-(trifluoromethyl)phenyl]-4-methyl-5-thiazolyl]methyl]thio]-2-methyl phenoxy]acetic acid), L165041, HPP-593, and NCP-1046.


Examples of PPAR alpha/gamma agonists that may be used in the pharmaceutical compositions described herein include saroglitazar, aleglitazar, muraglitazar, tesaglitazar, and DSP-8658.


Examples of PPAR alpha/delta agonists that may be used in the pharmaceutical compositions described herein include elafibranor and T913659.


Examples of PPAR gamma/delta agonists that may be used in the pharmaceutical compositions described herein include a conjugated linoleic acid (CLA) and T3D-959. Examples of PPAR alpha/gamma/delta agonists that may be used in the pharmaceutical compositions described herein include IVA337 (lanifibranor), TTA (tetradecylthioacetic acid), bavachinin, GW4148, GW9135, bezafibrate, lobeglitazone, 2-(4-(5,6-methylenedioxybenzo[d]thiazol-2-yl)-2-methylphenoxy)-2-methylpropanoic acid (MHY2013), and CS038.


Examples of thyroid hormone receptor beta agonists that may be used in the pharmaceutical compositions described herein include sobetirome, eprotirome, GC-24, MGL-3196, MGL-3745, VK-2809, KB141 [3,5-dichloro-4-(4-hydroxy-3-isopropylphenoxy) phenylacetic acid], and MB07811 (2R,4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4′-hydroxy-3′-isopropylbenzyl)phenoxy)methyl]-2-oxido-[1,3,2]-dioxaphosphonane).


As used herein, the term “pharmaceutically acceptable carrier” refers to any ingredient other than the disclosed compounds, or a pharmaceutically acceptable isomer, racemate, hydrate, solvate, isotope or salt thereof (e.g., a carrier capable of suspending or dissolving the active compound) and having the properties of being nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, or waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.


The formulations can be mixed with auxiliary agents which do not deleteriously react with the active compounds. Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances, preserving agents, sweetening agents, or flavoring agents. The compositions can also be sterilized if desired.


The route of administration can be any route which effectively transports the active compound of the invention to the appropriate or desired site of action, such as oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal, or parenteral, including intravenous, subcutaneous and/or intramuscular. In one embodiment, the route of administration is oral. In another embodiment, the route of administration is topical.


Dosage forms can be administered once a day, or more than once a day, such as twice or thrice daily. Alternatively, dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician or drug's prescribing information. Dosing regimens include, for example, dose titration to the extent necessary or useful for the indication to be treated, thus allowing the patient's body to adapt to the treatment, to minimize or avoid unwanted side effects associated with the treatment, and/or to maximize the therapeutic effect of the present compounds. Other dosage forms include delayed or controlled-release forms. Suitable dosage regimens and/or forms include those set out, for example, in the latest edition of the Physicians' Desk Reference, incorporated herein by reference.


Proper dosages for pediatric patients can be determined using known methods, including weight, age, body surface area, and models such as Simcyp® Pediatric Simulation modeling (CERTARA, Princeton, N.J.) which can be used to establish a pharmacokinetic approach for dosing that takes into account patient age, ontogeny of the clearance pathways to eliminate a compound of any one of formulas (IA) through (IH), and body surface area (BSA). In one embodiment, the dosage form is formulated to provide a pediatric dose from about 30% to about 100% of an adult dose, or about 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of an adult dose.


In one embodiment, the invention provides an oral pharmaceutical composition comprising a compound of structure (I) or any one of formulas (IA) through (IH), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, together with at least one pharmaceutically acceptable oral carrier, diluent, or excipient. In another embodiment, the invention provides a topical pharmaceutical composition comprising a compound of structure (I) or any one of formulas (IA) through (IH), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof, together with at least one pharmaceutically acceptable topical carrier, diluent, or excipient. For example, the oral pharmaceutical composition is provided to treat cholestatic pruritus, wherein the dosage regimen is, for example, once a day. In one embodiment, the topical pharmaceutical composition is provided to treat atopic dermatitis.


In another embodiment, there are provided methods of making a composition of a compound described herein including formulating a compound of the invention with a pharmaceutically acceptable carrier or diluent. In some embodiments, the pharmaceutically acceptable carrier or diluent is suitable for oral administration. In some such embodiments, the methods can further include the step of formulating the composition into a tablet or capsule. In other embodiments, the pharmaceutically acceptable carrier or diluent is suitable for parenteral administration. In some such embodiments, the methods further include the step of lyophilizing the composition to form a lyophilized preparation. In some embodiments, the composition is formulated into a pediatric dosage form suitable for treating a pediatric subject.


In certain embodiments, the invention provides a compound having structure (I) or any one of formulas (IA) through (IH), or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof. Such compounds can be synthesized using standard synthetic techniques known to those skilled in the art. For example, compounds of the present invention can be synthesized using appropriately modified synthetic procedures set forth in the following Examples and Reaction Schemes.


To this end, the reactions, processes, and synthetic methods described herein are not limited to the specific conditions described in the following experimental section, but rather are intended as a guide to one with suitable skill in this field. For example, reactions may be carried out in any suitable solvent, or other reagents to perform the transformation[s] necessary. Generally, suitable solvents are protic or aprotic solvents which are substantially non-reactive with the reactants, the intermediates or products at the temperatures at which the reactions are carried out (i.e., temperatures which may range from the freezing to boiling temperatures). A given reaction may be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction, suitable solvents for a particular work-up following the reaction may be employed.


All reagents, for which the synthesis is not described in the experimental part, are either commercially available, or are known compounds or may be formed from known compounds by known methods by a person skilled in the art. The compounds and intermediates produced according to the methods of the invention may require purification. Purification of organic compounds is well known to a person skilled in the art and there may be several ways of purifying the same compound. In some cases, no purification may be necessary. In some cases, the compounds may be purified by crystallization. In some cases, impurities may be stirred out using a suitable solvent. In some cases, the compounds may be purified by chromatography, particularly flash column chromatography, using purpose-made or prepacked silica gel cartridges and eluents such as gradients of solvents such as heptane, ether, ethyl acetate, acetonitrile, ethanol and the like. In some cases, the compounds may be purified by preparative HPLC using methods as described.


Purification methods as described herein may provide compounds of the present invention which possess a sufficiently basic or acidic functionality in the form of a salt, such as, in the case of a compound of the present invention which is sufficiently basic, a trifluoroacetate or formate salt, or, in the case of a compound of the present invention which is sufficiently acidic, an ammonium salt. A salt of this type can either be transformed into its free base or free acid form, respectively, by various methods known to a person skilled in the art, or be used as salts in subsequent biological assays. It is to be understood that the specific form of a compound of the present invention as isolated and as described herein is not necessarily the only form in which said compound can be applied to a biological assay in order to quantify the specific biological activity.


Chemical names were generated using the ChemDraw naming software (Version 17.0.0.206) by PerkinElmer Informatics, Inc. In some cases, generally accepted names of commercially available reagents were used in place of names generated by the naming software.


EXAMPLES

General Methods 1H NMR (400 MHz) spectra were obtained in solution of deuterochloroform (CDCl3), deuteromethanol (CD3OD) or dimethyl sulfoxide-D6 (DMSO-D6). HPLC retention times, purities and mass spectra (LCMS) were obtained using one of the following methods:


Method 1: Agilent 1260 Infinity II System equipped with an Agilent Poroshell 120 EC-18, 2.7 m, 4.6×100 mm column, using H2O with 0.1% formic acid as mobile phase A, and MeCN with 0.1% formic acid as mobile phase B. The gradient was 10-95% mobile phase B over 12 min, held at 95% for 2 min, then returned to 10% mobile phase B over 1 min. The flow rate was 1 mL/min. An ESI detector in negative mode was used.


Method 2: Agilent 1260 Infinity II System equipped with an Agilent Poroshell 120 EC-18, 2.7 m, 4.6×100 mm column, using H2O with 0.1% formic acid as mobile phase A, and MeCN with 0.1% formic acid as mobile phase B. The gradient was 10-95% mobile phase B over 12 min then held at 95% for 2 min, then return to 10% mobile phase B over 1 min. The flow rate was 1 mL/min. An ESI detector in positive mode was used.


Method 3: Agilent 1100 HPLC system equipped with an Agilent Eclipse XDB-C 18, 3.5μ, 4.6×150 mm column, using water with 0.05% TFA as mobile phase A, and methanol with 0.05% TFA as mobile phase B with a flow rate of 1 mL/minute. Using a gradient of 5% B (95% A) to 95% B over 12 minutes, held at 95% B for 3 minutes and then back to 5% B over 1 minute. An APCI detector in positive mode was used.


Method 4: Agilent 1100 HPLC system equipped with a BEH C18, 1.7 μM, 2.1×50 mm column using a low pH buffer gradient of 5% to 100% of MeCN in H2O (10 mM NH4HCO2; pH 4) over 10 min at 0.7 mL/min. A Waters Micromass ZQ ESI detector was used.


Method 5: SHIMADZU LCMS-2020 equipped with Kinetex® EVO C18 2.1×30 mm 5 μm column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.15 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.04 min. The flow rate was 2 mL/min. An ESI detector in positive mode was used.


Method 6: SHIMADZU LCMS-2020 equipped with Kinetex® EVO C18 2.1×20 mm 2.6 μm column, using H2O with 0.0375% TFA as mobile phase A, and MeCN with 0.01875% TFA as mobile phase B. The gradient was 5-95% mobile phase B for 0.8 min, held at 95% for 0.15 min, then returned to 5% mobile phase B for 0.01 min, held at 5% for 0.04 min. The flow rate was 2 mL/min. An ESI detector in positive mode was used.


The pyridine, dichloromethane (DCM), tetrahydrofuran (THF), and toluene used in the procedures were from Aldrich Sure-Seal bottles kept under nitrogen (N2). All reactions were stirred magnetically, and temperatures are external reaction temperatures. Chromatographies were typically carried out using a Combiflash Rf flash purification system (Teledyne Isco) equipped with Redisep (Teledyne Isco) Rf Gold Normal-Phase silica gel (SiO2) columns or by using a similar system.


Preparative HPLC purifications were typically performed using one of the following systems: 1) Waters System equipped with a Waters 2489 uv/vis detector, an Aquity QDA detector, a Waters xBridge Prep C18 5 μm OBD, 30×150 mm column, and eluting with various gradients of H2O/MeCN (0.1% formic acid) at a 30 mL/min flow rate, 2) Teledyne Isco ACCQPrep® HP150 UV system equipped with a Waters xBridge Prep C18 5 μm OBD, 30×150 mm column, and eluting with various gradients of H2O/MeCN (0.1% formic acid) at a 42.5 mL/min flow rate, or 3) column: Phenomenex Synergi C18 150×30 mm-4 m; mobile phase: [H2O (0.225% formic acid)-MeCN]; B %: 55%-85%, 12 min) and were typically concentrated using a Genevac EZ-2.


The following additional abbreviations are used: ethyl acetate (EA), triethylamine (TEA), dimethyl sulfoxide (DMSO), silica gel (SiO2), azobisisobutyronitrile (AIBN), diisobutylaluminium hydride (DIBAL), trifluoroacetic acid (TFA), 4-dimethylaminopyridine (DMAP), diphenylphosphoryl azide (DPPA), benzoyl peroxide (BPO), 1,1′-bis(diphenylphosphino)ferrocene (dppf), bis(pinacolato)diboron (B2pin2), tetrahydrofuran (THF), 1,4-diazabicyclo[2.2.2]octane bis(sulfur dioxide) adduct (DABSO), hexafluorophosphate azabenzotriazole tetramethyl uronium (HATU), hydroxybenzotriazole (HOBt), N-methyl morpholine (NMN), N-Bromosuccinimide (NBS), diisopropylethyl amine (DIPEA), diethyl azodicarboxylate (DEAD), diisopropyl azodicarboxylate (DIAD), 2-[2-(dicyclohexylphosphino)phenyl]-N-methylindole (CM-Phos), triflic acid (TfOH), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC), isopropanol (IPA), dimethylformamide (DMF), dimethyl acetamide (DMA), dichloromethane (DCM), 1,2-dichloroethane (DCE), acetonitrile (MeCN or ACN), 1,1′-thiocarbonyldiimidazole (TCDI), petroleum ether (PE), not determined (ND), retention time (RT), molecular weight (mw), room temperature (rt), hour (h), and not applicable (N/A).


Example 1
Synthesis of Compound 316



embedded image




embedded image


Step 1-1. Synthesis of methyl 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylate (INT-1A)



embedded image


To a stirring solution of methyl 1H-indazole-3-carboxylate (300 mg, 1 Eq, 1.70 mmol) in THE (10 mL) at 0° C. was slowly added potassium tert-butoxide (210 mg, 1.2 Eq, 1.87 mmol) in two portions separated by 10 minutes. The reaction mixture was warmed to room temperature and stirred for 1 h. After 1 h, the reaction mixture was cooled back to 0° C. and 1-(bromomethyl)-2,4-dichlorobenzene (490 mg, 1.2 Eq., 2.04 mmol) was added. After 15 minutes, the reaction mixture was warmed to 50° C. After heating at 50° C. for 15 h, the reaction mixture was cooled to room temperature and H2O (10 mL) was added. The aqueous layer was extracted with EtOAC (3×15 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. The crude mixture was purified by SiO2 gel chromatography (0->50% 10% MeOH in EtOAc and hexanes) to yield 443 mg (78%) of methyl 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylate (INT-1A) as a white solid.


Step 1-2. Synthesis of 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylic acid (Compound 316)



embedded image


To a stirring solution of methyl 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylate (INT-1A) (443 mg, 1.0 Eq., 1.32 mmol) in THE (5 mL) at room temperature was added 1M NaOH (aq.) (5.3 mL, 4.0 Eq., 5.3 mmol). After stirring for 14 h, the reaction mixture was concentrated in vacuo. To the crude residue was added H2O (10 mL) followed by 3M HCl (3 mL). The aqueous layer was extracted with EtOAc (3×15 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford 207 mg (49% yield) of 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylic acid (Compound 316) as a white solid; LCMS (m/z) calculated for C15H10Cl2N2O2: 320.0; found 321.0 [M+H]+, tR=9.13 min (Method 2). 1H NMR (400 MHz, DMSO-d6) δ 13.12 (br s, 1H), 8.12 (d, J=8 Hz, 1H), 7.81 (d, J=8 Hz, 1H), 7.70 (br s, 1H), 7.50 (t, J=8 Hz, 1H), 7.37 (m, 2H), 6.96 (d, J=8 Hz, 1H), 5.84 (s, 2H).


The compounds listed in Table 1 were made using the procedures of Scheme 1.















TABLE 1






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method























embedded image


316
9.13
321.2
321.0
[M + H]+
2







embedded image


317
9.851
354.7
353.0
[M − H]
1







embedded image


318
8.493
321.2
321.0
[M + H]+
2







embedded image


319
8.744
286.72
287.1
[M + H]+
2







embedded image


320
9.139
321.16
321.0
[M + H]+
2







embedded image


321
8.292
336.3
334.8
[M − H]
1







embedded image


322
8.438
304.71
305.1
[M + H]+
2







embedded image


323
4.791
294.4
295.2
[M + H]+
2







embedded image


324
9.52
300.74
301.1
[M + H]+
2







embedded image


325
8.273
318.28
317.2
[M − H]
1







embedded image


326
8.572
304.71
305.1
[M + H]+
2







embedded image


327
8.064
304.71
305.1
[M + H]+
2







embedded image


328
9.248
336.27
337.1
[M + H]+
2







embedded image


329
8.608
316.74
317.1
[M + H]+
2







embedded image


330
9.663
300.74
301.1
[M + H]+
2







embedded image


331
8.059
270.26
271.1
[M + H]+
2







embedded image


332
11.493
334.2
335.1
[M + H]+
2







embedded image


533
12.67
448.89
448.7
[M + H]+
3







embedded image


534
12.62
476.94
476.9
[M + H]+
3







embedded image


535
11.69
447.9
447.9
[M + H]+
3







embedded image


536
11.86
492.9
493
[M + H]+
3







embedded image


537
12.35
454.51
455.1
[M + H]+
3







embedded image


538
11.72
470.53
471
[M + H]+
3









Example 2
Synthesis of Compound 43



embedded image




embedded image


Step 2-1. Synthesis of 1-(4-chloro-3-methoxybenzyl)-1H-indazole-3-carbonyl chloride (Compound 2-1)



embedded image


To 1-(4-chloro-3-methoxybenzyl)-1H-indazole-3-carboxylic acid (Compound 1-14) (25 mg, 1 Eq., 79 μmol) was added SOCl2 in a vial. The vial was capped and heated at 70° C. After heating at 70° C. for 3 h, the reaction mixture was concentrated in vacuo.


To the crude residue was added toluene (2 mL) and the reaction mixture was concentrated in vacuo. This was repeated twice more with toluene (2 mL) to give 1-(4-chloro-3-methoxybenzyl)-1H-indazole-3-carbonyl chloride (INT-2A) (26 mg) yield (100%) as a light yellow oil that was carried on without further purification.


Step 2-2. Synthesis of 1-(4-chloro-3-methoxybenzyl)-N-(2-(pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide (Compound 43)



embedded image


To a stirring solution of 1-(4-chloro-3-methoxybenzyl)-1H-indazole-3-carbonyl chloride (INT-2A) (26 mg, 1.0 Eq, 78 μmol) in DCM (1 mL) at room temperature was added Et3N (54 μL, 5.0 Eq, 390 μmol) followed by a solution of 2-(pyridin-3-yl)ethan-1-amine (9.5 mg, 1.0 Eq, 78 μmol) in DCM (0.2 mL). After stirring for 14 h, the reaction mixture was concentrated in vacuo. The crude residue was purified by reversed phase preparatory HPLC (20->35% 0.1% formic acid in MeCN and 0.1% formic acid in H2O) to yield 6.5 mg (20%) of 1-(2,4-dichlorobenzyl)-1H-indazole-3-carboxylic acid (Compound 43); LCMS (m/z) calculated for C23H11ClN4O2: 420.1; found 421.1 [M+H]+, tR=7.073 min (Method 2). 1H NMR (400 MHz, CDCl3) δ 8.40 (d, J=8 Hz, 1H), 7.65 (d, J=8 Hz, 1H), 7.40 (m, 1H), 7.30 (m, 5H), 7.14 (m, 1H), 6.69 (m, 2H), 3.79 (m, 5H), 6.96 (d, J=8 Hz, 1H), 5.84 (s, 2H), 3.02 (t, J=8 Hz, 1H).


The compounds listed in Table 2 were made using the procedures of Scheme 2.















TABLE 2






Cpd
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method























embedded image


257
10.804
453.5
454.1
[M + H]+
2







embedded image


1
7.851
462.9
463.1
[M + H]+
2







embedded image


2
7.618
448.9
449.1
[M + H]+
2







embedded image


3
7.704
448.9
449.1
[M + H]+
2







embedded image


4
7.577
444.9
445.2
[M + H]+
2







embedded image


5
7.561
482.5
483.2
[M + H]+
1







embedded image


6
7.937
449.0
449.2
[M + H]+
2







embedded image


7
8.044
463.0
463.2
[M + H]+
2







embedded image


8
8.069
483.4
483.1
[M + H]+
2







embedded image


9
9.654
405.9
406.1
[M + H]+
2







embedded image


10
7.818
430.1
431.1
[M + H]+
2







embedded image


11
8.00
462.2
463.2
[M + H]+
2







embedded image


12
8.26
464.1
465.1
[M + H]+
2







embedded image


13
7.48
428.2
429.2
[M + H]+
2







embedded image


14
7.68
464.2
465.1
[M + H]+
2







embedded image


15
6.998
448.1
449.1
[M + H]+
2







embedded image


16
7.787
424.1
425.1
[M + H]+
2







embedded image


17
7.173
408.1
409.1
[M + H]+
2







embedded image


18
7.228
408.1
409.1
[M + H]+
2







embedded image


19
6.821
390.1
391.1
[M + H]+
2







embedded image


20
10.573
405.1
406.1
[M + H]+
2







embedded image


21
6.166
404.1
405.2
[M + H]+
2







embedded image


22
7.594
424.1
425.1
[M + H]+
2







embedded image


23
7.202
390.1
391.1
[M + H]+
2







embedded image


24
7.742
480.2
481.2
[M + H]+
2







embedded image


25
7.194
434.2
435.2
[M + H]+
2







embedded image


26
6.986
453.2
454.2
[M + H]+
2







embedded image


27
7.792
404.1
405.2
[M + H]+
2







embedded image


28
11.27
430.2
431.2
[M + H]+
2







embedded image


29
11.395
450.1
451.1
[M + H]+
2







embedded image


30
7.171
446.2
447.2
[M + H]+
2







embedded image


31
6.992
390.1
391.1
[M + H]+
2







embedded image


32
7.396
404.1
405.1
[M + H]+
2







embedded image


33
7.044
437.1
438.1
[M + H]+
2







embedded image


34
10.249
424.1
425.1
[M + H]+
2







embedded image


35
8.381
456.3
457.2
[M + H]+
2







embedded image


36
11.149
416.2
417.2
[M + H]+
2







embedded image


37
7.365
404.1
405.1
[M + H]+
2







embedded image


38
10.033
390.2
391.2
[M + H]+
2







embedded image


39
6.766
438.2
439.2
[M + H]+
2







embedded image


40
10.004
405.1
406.1
[M + H]+
2







embedded image


41
6.951
417.2
418.2
[M + H]+
2







embedded image


42
6.723
422.2
423.2
[M + H]+
2







embedded image


43
7.073
420.1
421.1
[M + H]+
2







embedded image


44
6.607
404.1
405.1
[M + H]+
2







embedded image


45
10.77
430.1
431.1
[M + H]+
2







embedded image


46
11.476
446.1
447.1
[M + H]+
2







embedded image


47
5.785
381.2
382.2
[M + H]+
2







embedded image


48
9.409
393.1
394.1
[M + H]+
2







embedded image


49
8.759
438.2
439.2
[M + H]+
2







embedded image


50
8.893
470.3
471.2
[M + H]+
2







embedded image


51
6.758
414.2
415.2
[M + H]+
2







embedded image


52
6.898
370.2
371.2
[M + H]+
2







embedded image


53
10.116
404.2
405.2
[M + H]+
2







embedded image


54
11.548
468.1
469.1
[M + H]+
2







embedded image


55
7.276
404.1
405.1
[M + H]+
2







embedded image


56
12.196
484.1
485.1
[M + H]+
2







embedded image


57
6.337
374.2
375.2
[M + H]+
2







embedded image


58
7.055
432.2
433.2
[M + H]+
2







embedded image


59
6.93
400.2
401.2
[M + H]+
2







embedded image


60
10.341
455.2
456.2
[M + H]+
2







embedded image


258
10.774
471.1
471.7
[M + H]+
2







embedded image


61
7.873
420.3
421.2
[M + H]+
2







embedded image


62
6.925
420.1
421.2
[M + H]+
2







embedded image


63
7.023
370.2
371.1
[M + H]+
2







embedded image


259
10.659
496.1
497.1
[M + H]+
2







embedded image


64
7.878
478.2
479.2
[M + H]+
2







embedded image


65
9.719
432.2
433.2
[M + H]+
2







embedded image


66
6.291
386.2
387.2
[M + H]+
2







embedded image


67
6.345
395.2
396.2
[M + H]+
2







embedded image


68
12.102
464.1
465.1
[M + H]+
2







embedded image


69
10.752
448.2
449.2
[M + H]+
2







embedded image


70
6.528
388.2
389.2
[M + H]+
2







embedded image


71
10.421
460.2
459.2
[M − H]
1







embedded image


260
10.312
430.1
431.1
[M + H]+
2







embedded image


72
5.681
381.2
382.2
[M + H]+
2







embedded image


73
8.233
360.2
361.2
[M + H]+
2







embedded image


74
8.031
462.2
463.2
[M + H]+
2







embedded image


75
11.50
420.1
421.2
[M + H]+
2







embedded image


76
9.058
373.2
374.2
[M + H]+
2







embedded image


77
7.187
404.1
405.1
[M + H]+
2







embedded image


78
6.887
370.2
371.2
[M + H]+
2







embedded image


261
11.728
477.0
478.0
[M + H]+
2







embedded image


79
11.14
438.1
439.1
[M + H]+
2







embedded image


80
6.97
408.1
409.1
[M + H]+
2







embedded image


81
10.648
434.2
435.2
[M + H]+
2







embedded image


82
10.899
482.2
483.1
[M + H]+
2







embedded image


83
10.351
401.2
402.2
[M + H]+
2







embedded image


262
11.055
525.1
525.6
[M + H]+
2







embedded image


84
12.27
425.2
426.2
[M + H]+
2







embedded image


263
11.879
532.1
533.0
[M + H]+
2







embedded image


85
10.871
403.2
404.2
[M + H]+
2







embedded image


264
11.661
453.1
454.1
[M + H]+
2







embedded image


86
10.635
458.1
459.1
[M + H]+
2







embedded image


87
6.771
390.1
391.1
[M + H]+
2







embedded image


88
9.134
454.1
455.1
[M + H]+
2







embedded image


265
11.308
525.1
525.6
[M + H]+
2







embedded image


89
6.768
390.1
391.1
[M + H]+
2







embedded image


90
9.300
380.1
381.1
[M + H]+
2







embedded image


266
10.697
475.1
475.7
[M + H]+
2







embedded image


91
7.919
398.2
399.2
[M + H]+
2







embedded image


92
7.167
438.2
439.1
[M + H]+
2







embedded image


267
10.697
475.1
475.7
[M + H]+
2







embedded image


268
11.352
485.1
486.1
[M + H]+
2







embedded image


269
10.405
475.1
475.7
[M + H]+
2







embedded image


270
10.886
471.1
471.7
[M + H]+
2







embedded image


93
11.435
478.2
478.8
[M + H]+
2







embedded image


94
12.054
422.2
423.2
[M + H]+
2







embedded image


95
11.604
450.1
451.1
[M + H]+
2







embedded image


271
10.865
460.0
461.1
[M + H]+
2







embedded image


272
11.675
489.1
489.7
[M + H]+
2







embedded image


96
6.27
374.2
375.2
[M + H]+
2







embedded image


97
8.94
371.2
372.2
[M + H]+
2







embedded image


98
11.999
464.1
465.1
[M + H]+
2







embedded image


99
7.493
422.2
423.1
[M + H]+
2







embedded image


100
9.274
380.1
381.1
[M + H]+
2







embedded image


273
10.158
430.1
431.1
[M + H]+
2







embedded image


274
11.167
489.1
490.1
[M + H]+
2







embedded image


275
11.033
501.1
502.1
[M + H]+
2







embedded image


101
6.320
386.2
387.2
[M + H]+
2







embedded image


102
8.597
391.1
392.1
[M + H]+
2







embedded image


103
10.649
410.2
411.2
[M + H]+
2







embedded image


276
11.411
525.1
525.6
[M + H]+
2







embedded image


277
10.594
515.1
515.7
[M + H]+
2







embedded image


278
10.811
489.1
490.0
[M + H]+
2







embedded image


104
10.37
358.2
359.2
[M + H]+
2







embedded image


279
11.441
439.1
440.1
[M + H]+
2







embedded image


105
11.776
457.1
458.1
[M + H]+
2







embedded image


106
8.617
490.2
491.2
[M + H]+
2







embedded image


280
10.305
482.1
482.7
[M + H]+
2







embedded image


281
11.484
541.1
541.6
[M + H]+
2







embedded image


282
11.085
485.1
486.1
[M + H]+
2







embedded image


283
11.157
505.1
506.0
[M + H]+
2







embedded image


107
7.154
374.2
375.2
[M + H]+
2







embedded image


284
12.076
507.9
508.0
[M + H]+
2







embedded image


108
12.46
458.6
459.2
[M + H]+
2







embedded image


109
7.59
444.2
445.2
[M + H]+
2







embedded image


110
12.445
464.1
465.1
[M + H]+
2







embedded image


111
8.413
412.1
413.1
[M + H]+
2







embedded image


112
8.436
412.2
413.2
[M + H]+
2







embedded image


285
10.602
487.1
487.7
[M + H]+
2







embedded image


113
8.138
374.2
375.2
[M + H]+
2







embedded image


114
7.222
420.1
421.1
[M + H]+
2







embedded image


286
11.079
541.1
541.6
[M + H]+
2







embedded image


115
6.737
370.2
371.2
[M + H]+
2







embedded image


287
11.304
439.1
440.1
[M + H]+
2







embedded image


116
7.963
446.2
447.2
[M + H]+
2







embedded image


288
11.677
453.1
454.1
[M + H]+
2







embedded image


117
11.476
458.1
459.1
[M + H]+
2







embedded image


118
11.969
464.1
465.1
[M + H]+
2







embedded image


119
9.227
419.2
420.1
[M + H]+
2







embedded image


120
11.38
435.2
436.2
[M + H]+
2







embedded image


289
10.479
487.1
487.7
[M + H]+
2







embedded image


121
8.674
391.1
392.1
[M + H]+
2







embedded image


290
11.168
489.1
490.1
[M + H]+
2







embedded image


291
9.916
426.1
427.0
[M + H]+
2







embedded image


122
11.022
359.2
360.2
[M + H]+
2







embedded image


292
10.807
423.1
424.1
[M + H]+
2







embedded image


293
10.723
435.1
436.1
[M + H]+
2







embedded image


294
11.865
525
526
[M + H]+
2







embedded image


295
11.243
485.1
486.1
[M + H]+
2







embedded image


296
11.532
541.1
541.6
[M + H]+
2







embedded image


297
11.361
419.1
420.1
[M + H]+
2







embedded image


298
11.353
419.1
420.1
[M + H]+
2







embedded image


299
9.626
440.1
441.1
[M + H]+
2







embedded image


300
11.181
469.1
470.1
[M + H]+
2







embedded image


301
10.878
468.0
469.0
[M + H]+
2







embedded image


123
11.764
480.1
481.2
[M + H]+
2







embedded image


302
11.993
467.1
468.1
[M + H]+
2







embedded image


303
11.767
505.1
506.1
[M + H]+
2







embedded image


304
9.676
426.1
427.0
[M + H]+
2







embedded image


124
8.034
373.2
374.2
[M + H]+
2







embedded image


125
6.224
374.2
375.2
[M + H]+
2







embedded image


126
9.341
361.2
362.1
[M + H]+
2







embedded image


127
7.33
404.1
405.1
[M + H]+
2







embedded image


305
10.345
440.1
441.1
[M + H]+
2







embedded image


306
12.168
507.1
508.0
[M + H]+
2







embedded image


128
9.697
360.2
361.1
[M + H]+
2







embedded image


307
11.60
457.1
458.0
[M + H]+
2







embedded image


129
7.126
448.2
449.2
[M + H]+
2







embedded image


308
12.268
493.0
493.9
[M + H]+
2







embedded image


130
8.203
362.1
363.1
[M + H]+
2







embedded image


131
11.733
468.1
469.1
[M + H]+
2









Example 3
Synthesis of Compound 136



embedded image




embedded image


Step 3-1. Synthesis of N-(2-(pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide



embedded image


To a stirring solution of 1H-indazole-3-carboxylic acid (100 mg, 1 Eq, 617 μmol) in DMF (1 mL) at 0° C. was added 1-Hydroxybenzotriazole—hydrate (HOBt) (116 mg, 1.1 Eq, 678 μmol) and Dicyclohexylcarbodiimide (DCC) (134 mg, 1.05 Eq, 648 μmol). The reaction mixture was stirred at 0° C. for 1 h. To the reaction mixture was added a precooled solution (0° C.) of 2-(pyridin-3-yl)ethan-1-amine (86.6 mg, 1.15 Eq, 709 μmol) in DMF (1 mL). The reaction mixture was stirred for 2 h at 0° C. and then warmed to room temperature overnight. The reaction mixture was diluted with EtOAc (5 mL), filtered, and extracted with 1M HCl (10 mL). The aqueous phase was basified to a pH ˜13 with 1M NaOH. The mixture was extracted with DCM (3×10 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford 130 mg (79% yield) of N-(2-(pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide (INT-3A) that was used without further purification. 1H NMR (400 MHz, DMSO-d6) δ 8.44 (m, 3H), 8.15 (d, J=8 Hz, 1H), 7.69 (d, J=8 Hz, 1H), 7.60 (d, J=8 Hz, 1H), 7.41 (t, J=8 Hz, 1H), 7.31 (dd, J=8, 4 Hz, 1H), 7.23, (t, J=8 Hz, 1H), 3.57 (m, 2H), 2.93 (m, 2H).


Step 3-2. Synthesis of N-(2-(pyridin-3-yl)ethyl)-1-(4-(trifluoromethoxy)benzyl)-1H-indazole-3-carboxamide (Compound 136)



embedded image


To a stirring solution of crude N-(2-(pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide (INT-3A) (30 mg, 1 Eq, 0.11 mmol) in DMF (1.5 mL) in a vial was added Cs2CO3 (0.11 g, 3 Eq, 0.34 mmol) and 1-(chloromethyl)-4-(trifluoromethoxy) benzene (26 mg, 1.1 Eq, 0.12 mmol). The vial was capped and heated at 70° C. for 14 h. After heating for 14 h, the reaction mixture was concentrated in vacuo to afford crude solid. The crude solid was purified by ISCO reversed phase prep (25-35% 0.1% formic acid in MeCN and 0.1% formic acid in water) to afford 16 mg (32% yield) of N-(2-(pyridin-3-yl)ethyl)-1-(4-(trifluoromethoxy)benzyl)-1H-indazole-3-carboxamide (Compound 136) as a white solid; LCMS (m/z) calculated for C23H19F3N4O2: 440.2; found 440.8 [M+H]+, tR=7.443 min (Method 2). 1H NMR (400 MHz, DMSO-d6) δ 8.54 (d, J=16 Hz, 2H), 8.42 (d, J=8 Hz, 1H), 7.63 (d, J=8 Hz, 1H), 7.30 (m, 9H), 5.59 (s, 2H), 3.77 (q, J=8 Hz, 1H), 3.02 (t, J=8 Hz, 1H).


The compounds listed in Table 3 were made using the procedures of Scheme 3.















TABLE 3






Cpd.
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method























embedded image


408
0.39
421.464
422.2
[M + H]+
6







embedded image


409
0.386
421.464
422.2
[M + H]+
6







embedded image


410
0.529
472.552
473.3
[M + H]+
6







embedded image


411
0.412
474.91
475.2
[M + H]+
5







embedded image


412
0.37
447.502
448.2
[M + H]+
5







embedded image


414
0.369
464.489
465.2
[M + H]+
5







embedded image


415
0.398
443.511
444.2
[M + H]+
5







embedded image


431
0.371
462.517
463.3
[M + H]+
5







embedded image


432
0.432
446.514
447.3
[M + H]+
5







embedded image


433
0.385
440.463
441.2
[M + H]+
5







embedded image


434
0.372
427.468
428.3
[M + H]+
5







embedded image


436
0.447
482.93
483.1
[M + H]+
5







embedded image


437
0.36
430.472
431.2
[M + H]+
5







embedded image


438
0.373
437.463
438.2
[M + H]+
5







embedded image


439
0.344
417.49
418.1
[M + H]+
5







embedded image


440
0.308
387.403
388.1
[M + H]+
5







embedded image


441
0.502
478.479
479.1
[M + H]+
5







embedded image


442
0.419
444.92
445.2
[M + H]+
5







embedded image


443
0.387
456.481
457.2
[M + H]+
5







embedded image


444
0.263
446.559
447.4
[M + H]+
5







embedded image


448
0.279
427.468
428.2
[M + H]+
5







embedded image


452
0.433
450.506
451.2
[M + H]+
5







embedded image


453
0.506
476.54
477.1
[M + H]+
5







embedded image


454
0.288
474.502
448.2
[M + H]+
5







embedded image


455
0.396
447.502
448.2
[M + H]+
5







embedded image


456
0.515
438.535
439.2
[M + H]+
5







embedded image


457
0.497
478.479
479.1
[M + H]+
5







embedded image


460
0.375
450.506
451.2
[M + H]+
5







embedded image


461
0.38
462.517
463.3
[M + H]+
5







embedded image


462
0.398
463.545
464.3
[M + H]+
5







embedded image


463
0.344
462.461
463.2
[M + H]+
5







embedded image


465
0.357
437.467
438.3
[M + H]+
5







embedded image


466
0.306
427.49
428.1
[M + H]+
5







embedded image


467
0.317
428.456
429.3
[M + H]+
5







embedded image


468
0.329
438.491
439.1
[M + H]+
5







embedded image


469
0.389
430.512
431.3
[M + H]+
5







embedded image


470
0.438
474.95
475.2
[M + H]+
5







embedded image


471
0.348
446.462
447.2
[M + H]+
5







embedded image


472
0.39
459.554
460.3
[M + H]+
5







embedded image


481
0.333
458.91
459.1
[M + H]+
5







embedded image


482
0.363
493.35
493.1
[M + H]+
5







embedded image


483
0.411
452.518
453.3
[M + H]+
5







embedded image


484
0.373
454.49
455.2
[M + H]+
5







embedded image


503
0.46
464.485
465.3
[M + H]+
5







embedded image


505
0.483
479.37
479.2
[M + H]+
5







embedded image


507
0.478
476.444
477.5
[M + H]+
5







embedded image


508
0.417
440.463
441.5
[M + H]+
5







embedded image


510
0.375
474.54
475.2
[M + H]+
5







embedded image


511
0.432
442.483
443.3
[M + H]+
5







embedded image


513
0.45
456.389
457.2
[M + H]+
5







embedded image


514
0.461
495.518
496.3
[M + H]+
5







embedded image


515
0.352
400.446
401.1
[M + H]+
5







embedded image


516
0.376
464.489
465.2
[M + H]+
5







embedded image


517
0.452
460.445
461.1
[M + H]+
5







embedded image


518
0.303
436.479
437.3
[M + H]+
5







embedded image


519
0.349
466.505
467.2
[M + H]+
5







embedded image


520
0.425
400.442
401.2
[M + H]+
5







embedded image


521
0.333
436.479
437.3
[M + H]+
5







embedded image


522
0.433
504.477
505.4
[M + H]+
5







embedded image


523
0.495
424.508
425.3
[M + H]+
5







embedded image


524
11.93
448.89
448.8
[M + H]+
3







embedded image


525
12.72
465.34
464.6
[M + H]+
3







embedded image


526
12.36
465.34
464.9
[M + H]+
3







embedded image


527
12.03
448.89
448.8
[M + H]+
3







embedded image


528
11.7
410.481
410.8
[M + H]+
3







embedded image


529
12.73
509.79
508.8
[M + H]+
3







embedded image


543
11.76
460.92
461.1
[M + H]+
3







embedded image


544
8.56
450.506
450.9
[M + H]+
3







embedded image


545
12.21
436.519
436.9
[M + H]+
3







embedded image


546
10.67
455.91
456.2
[M + H]+
3







embedded image


547
11.14
465.34
464.6
[M + H]+
3







embedded image


548
11.52
452.54
452.9
[M + H]+
3







embedded image


549
12.18
444.92
444.6
[M + H]+
3







embedded image


550
12.61
509.79
508.5
[M + H]+
3







embedded image


551
11.36
455.91
455.9
[M + H]+
3







embedded image


552
12.69
556.79
556.5
[M + H]+
3







embedded image


553
12.34
498.89
498.6
[M + H]+
3







embedded image


554
11.91
444.92
444.9
[M + H]+
3







embedded image


555
12.1
444.92
445
[M + H]+
3







embedded image


556
10.88
435.491
436
[M + H]+
3







embedded image


557
12.53
464.529
464.9
[M + H]+
3







embedded image


558
13.4
452.54
452.8
[M + H]+
3







embedded image


559
12.7
450.546
450.8
[M + H]+
3







embedded image


568
8.11
464.533
464.9
[M + H]+
3







embedded image


569
9.17
464.533
464.5
[M + H]+
3







embedded image


570
9.19
464.533
465.2
[M + H]+
3







embedded image


571
12.2
483.33
483
[M + H]+
3







embedded image


572
11.91
466.88
467
[M + H]+
3







embedded image


573
11.91
450.502
451.4
[M + H]+
3







embedded image


574
9.63
468.496
468.9
[M + H]+
3









Example 4
Synthesis of Compound 135



embedded image




embedded image


Step 4-1. Synthesis of N-(2-(1H-indol-3-yl)ethyl)-1-(4-methylbenzyl)-1H-indazole-3-carboxamide (Compound 135)



embedded image


To a vial containing a stirred solution of 1-(4-methylbenzyl)-1H-indazole-3-carboxylica acid (100 mg, 1 Eq, 376 μmol), and DIPEA (194 mg, 0.26 mL, 4 Eq, 1.50 mmol), in DMF (2 mL) was added HATU (157 mg, 1.1 Eq, 413 μmol) and the resulting yellow solution was stirred at room temperature for 5 minutes. Tryptamine (72 mg, 1.2 Eq, 451 μmol) was added, and the resulting yellow-orange solution was stirred overnight at room temperature. After stirring for 17 hours, the reaction mixture (orange solution) was diluted with EtOAc (5 mL) and washed with saturated aqueous NaHCO3 (10 mL). The aqueous layer was extracted with EtOAc (2×10 mL). The combined organics were washed with brine (10 mL), dried over Na2SO4, filtered, and concentrated in vacuo to give crude residue. The crude residue (orange oil) was purified by silica gel column chromatography (0-70% EtOAc and hexanes) to give 131 mg (85% yield) of N-(2-(1H-indol-3-yl)ethyl)-1-(4-methylbenzyl)-1H-indazole-3-carboxamide (Compound 135) as a pale-yellow foam; LCMS (m/z) calculated for C26H24N4O: 408.2; found 409.3 [M+H]+, tR=10.911 min (Method 2).


The compounds listed in Table 4 were made using the procedures of Scheme 4.















TABLE 4






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method























embedded image


149
11.26
389.1
390.6
[M + H]+
3







embedded image


132
12.77
404.1
405.4
[M + H]+
3







embedded image


133
13.34
363.1
364.4
[M + H]+
3







embedded image


134
13.99
412.1
413.2
[M + H]+
3







embedded image


135
10.911
408.2
409.3
[M + H]+
2







embedded image


334
0.501
391.86
391.9
[M + H]+
5







embedded image


335
0.628
423.87
423.9
[M + H]+
5







embedded image


336
0.469
406.87
407
[M + H]+
5







embedded image


337
0.622
419.91
420
[M + H]+
5







embedded image


338
0.491
444.92
445.2
[M + H]+
5







embedded image


339
0.58
424.33
424.1
[M + H]+
5







embedded image


340
0.554
442.95
443.2
[M + H]+
5







embedded image


341
0.561
442.95
443.2
[M + H]+
5







embedded image


342
0.553
433.89
434.1
[M + H]+
5







embedded image


343
0.534
449.94
450.1
[M + H]+
5







embedded image


344
0.596
458.77
458
[M + H]+
5







embedded image


345
0.379
393.88
394.2
[M + H]+
5







embedded image


346
0.396
398.9
399.2
[M + H]+
5







embedded image


347
0.487
424.81
425.1
[M + H]+
5







embedded image


348
0.473
391.86
392.3
[M + H]+
5







embedded image


349
0.52
378.86
379.2
[M + H]+
5







embedded image


350
0.551
395.91
396.1
[M + H]+
5







embedded image


351
0.38
398.89
399.3
[M + H]+
5







embedded image


352
0.402
415.88
4A.1
[M + H]+
5







embedded image


353
0.468
379.85
380.1
[M + H]+
5







embedded image


354
0.543
379.85
380.1
[M + H]+
6







embedded image


355
0.387
404.9
405.1
[M + H]+
5







embedded image


356
0.566
429.9
430.2
[M + H]+
5







embedded image


357
0.405
429.91
430.2
[M + H]+
5







embedded image


358
0.411
492.91
430.2
[M + H]+
5







embedded image


359
0.533
441.92
442.2
[M + H]+
5







embedded image


360
0.557
411.84
412.1
[M + H]+
5







embedded image


361
0.507
430.9
431.1
[M + H]+
5







embedded image


362
0.58
403.91
404.1
[M + H]+
5







embedded image


363
0.57
403.91
404.3
[M + H]+
5







embedded image


364
0.491
420.9
421.2
[M + H]+
5







embedded image


365
0.57
433.94
434.1
[M + H]+
5







embedded image


366
0.53
435.91
436.2
[M + H]+
6







embedded image


367
0.468
410.92
411.1
[M + H]+
5







embedded image


368
0.381
4A.87
417.1
[M + H]+
5







embedded image


369
0.527
442.91
443.2
[M + H]+
5







embedded image


370
0.432
426.9
427.1
[M + H]+
5







embedded image


371
0.418
426.9
427.1
[M + H]+
5







embedded image


372
0.435
426.9
427.2
[M + H]+
6







embedded image


373
0.568
444.96
445.2
[M + H]+
5







embedded image


374
0.487
432.91
433.1
[M + H]+
5







embedded image


375
0.459
377.83
378.1
[M + H]+
5







embedded image


376
0.494
391.86
392.1
[M + H]+
5







embedded image


377
0.498
432.91
433.3
[M + H]+
5







embedded image


378
0.497
453.94
454.2
[M + H]+
5







embedded image


379
0.524
453.94
454.1
[M + H]+
5







embedded image


380
0.541
439.85
440
[M + H]+
5







embedded image


381
0.549
405.88
406.3
[M + H]+
5







embedded image


382
0.505
419.87
420.1
[M + H]+
5







embedded image


383
0.526
422.91
423.2
[M + H]+
5







embedded image


384
0.448
406.89
407.1
[M + H]+
5







embedded image


385
0.48
445.96
446.1
[M + H]+
5







embedded image


386
0.418
415.88
4A.1
[M + H]+
5







embedded image


387
0.499
415.88
4A.1
[M + H]+
5







embedded image


388
0.513
415.88
4A.2
[M + H]+
5







embedded image


389
0.515
430.9
431.2
[M + H]+
5







embedded image


390
0.53
431.94
432.1
[M + H]+
5







embedded image


391
0.499
429.91
430.1
[M + H]+
5







embedded image


392
0.454
406.87
407.1
[M + H]+
5







embedded image


393
0.555
416.87
417.2
[M + H]+
6







embedded image


395
0.452
461.95
462.2
[M + H]+
6







embedded image


396
0.594
463.36
463.1
[M + H]+
6







embedded image


397
0.433
443.94
444.2
[M + H]+
6







embedded image


398
0.456
380.84
381.2
[M + H]+
6







embedded image


399
0.508
432.91
433.1
[M + H]+
6







embedded image


400
0.501
432.91
433.1
[M + H]+
5







embedded image


401
0.465
426.9
427.2
[M + H]+
5







embedded image


403
0.488
410.9
411.2
[M + H]+
5







embedded image


404
0.497
401.85
402.1
[M + H]+
5







embedded image


405
0.441
377.83
378.1
[M + H]+
5







embedded image


406
0.386
406.87
407.2
[M + H]+
5







embedded image


407
0.532
453.94
454.1
[M + H]+
6







embedded image


416
0.563
458.95
459.1
[M + H]+
5







embedded image


418
0.467
406.82
407.2
[M + H]+
5







embedded image


419
0.534
410.9
411.1
[M + H]+
6







embedded image


420
0.484
395.85
396.2
[M + H]+
5







embedded image


421
0.403
429.91
430.1
[M + H]+
5







embedded image


422
0.525
408.89
409.1
[M + H]+
5







embedded image


423
0.457
426.9
427.1
[M + H]+
5







embedded image


424
0.425
440.89
441.2
[M + H]+
5







embedded image


425
0.409
443.94
444.3
[M + H]+
5







embedded image


426
0.408
441.92
442.2
[M + H]+
5







embedded image


427
0.533
417.85
418.2
[M + H]+
5







embedded image


428
0.5A
429.91
430.2
[M + H]+
5







embedded image


429
0.415
441.92
442.3
[M + H]+
5







embedded image


475
0.455
415.88
416.2
[M + H]+
5







text missing or illegible when filed








Example 5
Synthesis of Representative Compound 309



embedded image




embedded image


Step 5-1. Synthesis of 1-(4-(difluoromethoxy)benzyl)-N-tosyl-1H-indazole-3-carboxamide (Compound 309)



embedded image


To a solution of 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) (78 mg, 1.3 Eq, 408 μmol and 4-dimethlaminopyridine (DMAP) (54 mg, 1.4 Eq, 440 μmol) in DCM (1 mL) at 0° C. was added 1-(4-(difluoromethoxy)benzyl)-1H-indazole-3-carboxylic acid (1-10) (100 mg, 1 Eq, 314 μmol). After stirring for 5 minutes, p-toluenesulfonamide (65 mg, 1.2 Eq, 377 μmol) was added. The reaction mixture was warmed to room temperature and stirred overnight. After stirring for 17 hours, The reaction mixture was purified by silica gel column chromatography (0->10% MeOH in DCM) and subsequently lyophilized in MeOH/H2O (4 mL 1:1) to afford 71 mg (48% yield) of 1-(4-(difluoromethoxy)benzyl)-N-tosyl-1H-indazole-3-carboxamide (Compound 309); LCMS (m/z) calculated for C23H19F2N3O4S: 471.1; found 472.2 [M+H]+, tR=10.718 min (Method 2).


The compounds listed in Table 5 were made using the procedures of Scheme 5.















TABLE 5






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method























embedded image


150
11.37
405.1
406.4
[M + H]+
3







embedded image


309
10.718
471.1
472.2
[M + H]+
2







embedded image


310
3.63
440.1
440.1
[M]
4









Example 6
Synthesis of Representative Compound 152



embedded image




embedded image


Step 6-1. methyl 1-(4-chlorobenzyl)-1H-indole-3-carboxylate (INT-6A)



embedded image


A 250 mL round bottom flask containing methyl 1H-indole-3-carboxylate (4.39 g, 1.0 equiv., 37.4 mmol) in DMF (30 mL) was added NaH (60% dispersion in mineral oil, 2.04 g, 1.4 equiv., 51.1 mmol) and then the suspension was stirred at rt for 5 minutes, 1-(bromomethyl)-4-chlorobenzene (7.0 g, 1.3 equiv., 34.1 mmol) was slowly added.


Stirred at rt for 20 minutes. Monitored by LCMS. 20 min: Coupling complete. Quenched with NH4Cl (100 mL) slowly. Product was extracted with EtOAc (300 mL), washed with brine (200 mL), dried over Na2SO4, filtered, and concentrated in vacuo to afford 8.0 g (78% yield) of methyl 1-(4-chlorobenzyl)-1H-indole-3-carboxylate (INT-6A); LCMS (m/z) calculated for C17H14ClNO2: 299.07; found 300.2 [M+H]+, tR=5.37 min (Method 3).


Step 6-2. 1-(4-chlorobenzyl)-1H-indole-3-carboxylic acid (INT-6B)



embedded image


To a 250 mL round bottom flask containing INT-6A (8.0 g, 1.0 equiv., 26.7 mmol) in MeOH (100 mL) was added NaOH (3.2 g, 1.0 equiv., 80.1 mmol) dissolved in H2O (10 mL). The mixture was stirred at rt for 20 h. Monitored by LCMS. No reaction occurred. Additional NaOH (1 g, 0.94 equiv., 25.0 mmol) was added to the reaction and the reaction was heated to 80° C. for 20 h. Concentrated to remove solvent and diluted with H2O (200 mL). The aqueous layer was acidified with 1M HCl. Product was extracted with EtOAc (300 mL), washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated. Diluted with DCM and filtered, collecting the solid to afford 4.0 g (53% yield) 1-(4-chlorobenzyl)-1H-indole-3-carboxylic acid (INT-6B); LCMS (m/z) calculated for C16H12ClNO2: 285.06; found 286.1 [M+H]+, tR=4.82 min (Method 3). 1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H), δ 8.24 (s, 1H), 8.03 (t, J=4 Hz, 1H), 7.52 (t, J=4 Hz, 1H), 7.40 (d, J=8 Hz, 2H), 7.30 (d, J=8 Hz, 2H), 7.20 (t, J=4 Hz, 2H), 5.51 (s, 2H).


Step 6-3. 1-(4-chlorobenzyl)-N-(2-(2-methylpyridin-3-yl)ethyl)-1H-indole-3-carboxamide (Compound 152)



embedded image


To a solution of INT-6B (30 mg, 1.0 equiv., 0.1 mmol) in DCM (4 ml) was added 2-(2-methylpyridin-3-yl)ethan-1-amine (15.7 mg, 0.12 equiv., 0.12 mmol), EDCI (17.9 mg, 0.12 equiv., 0.0115 mmol) and DMAP (14.2 mg, 0.12 equiv., 0.12 mmol) at rt for overnight. The reaction mixture was diluted with DCM and water. The DCM layer was separated and dried, concentrated and purified by silica gel column chromatography (0-100% EtOAc/Hexane first and then 0-10% MeOH/DCM) to afford 27 mg (64% yield) of 1-(4-chlorobenzyl)-N-(2-(2-methylpyridin-3-yl)ethyl)-1H-indole-3-carboxamide (Compound 152) as a white solid; LCMS (m/z) calculated for C24H22ClN3O: 403.15; found 404.5 [M+H]+, tR=11.27 min (Method 3). 1H NMR (400 MHz, DMSO-d6) δ 8.29 (s, 1H), 8.13 (d, J=4 Hz, 1H), 8.0 (m, 2H), 7.52 (m, 2H), 7.41 (d, J=8 Hz, 2H), 7.23 (d, J=8 Hz, 2H), 7.13 (m, 3H), 5.46 (s, 2H), 3.47 (m, 2H), 2.87 (m, 2H), 2.53 (s, 3H).


The compounds listed in Table 6 were made using the procedures of Scheme 6.















TABLE 6







Purity

Observed

Purity


Structure
Cpd No.
RT (min)
NM
m/z
Ion
Method























embedded image


151
11.5
403.91
404.6
[M + H]+
3







embedded image


152
11.27
403.91
404.5
[M + H]+
3







embedded image


153
11.63
403.91
404.5
[M + H]+
3







embedded image


154
12.95
407.87
408.3
[M + H]+
3







embedded image


155
13.36
419.91
420.2
[M + H]+
3







embedded image


156
11.4
417.94
418.5
[M + H]+
3







embedded image


157
11.25
389.88
390.5
[M + H]+
3







embedded image


158
95
369.468
370.3
[M + H]+
3







embedded image


159
12.92
389.52
390.5
[M + H]+
3







embedded image


160
11.29
369.468
370.4
[M + H]+
3







embedded image


161
11.58
403.91
404.5
[M + H]+
3







embedded image


162
13.66
406.89
407.3
[M + H]+
3







embedded image


163
11.33
389.88
390.5
[M + H]+
3







embedded image


164
13.6
418.92
419.4
[M + H]+
3







embedded image


165
13.63
429.58
430.7
[M + H]+
3







embedded image


166
11.83
421.448
422.2
[M + H]+
3







embedded image


167
10.91
457.908
422.2
[M + H]+
3







embedded image


168
12.93
419.91
420.4
[M + H]+
3







embedded image


169
13.43
407.517
408.4
[M + H]+
3







embedded image


170
13.56
406.89
407.5
[M + H]+
3







embedded image


171
12.64
390.87
391.5
[M + H]+
3







embedded image


172
11.17
369.468
370.4
[M + H]+
3







embedded image


173
11.8
439.438
440.2
[M + H]+
3







embedded image


174
13.78
415.56
416.6
[M + H]+
3







embedded image


175
14.01
402.92
403.5
[M + H]+
3







embedded image


176
11.16
385.467
386.3
[M + H]+
3







embedded image


177
13.84
418.92
419.3
[M + H]+
3







embedded image


178
13.97
402.92
403.6
[M + H]+
3







embedded image


179
13.67
406.89
407.3
[M + H]+
3







embedded image


180
14.08
402.92
403.6
[M + H]+
3







embedded image


181
11.49
416.54
417.5
[M + H]+
3







embedded image


182
13.6
418.92
419
[M + H]+
3







embedded image


183
11.65
389.88
390.5
[M + H]+
3







embedded image


184
9.93
380.45
381.3
[M + H]+
3







embedded image


185
12.84
414.89
415.4
[M + H]+
3







embedded image


186
11.58
439.43
440.3
[M + H]+
3







embedded image


187
12.6
372.472
373.3
[M + H]+
3







embedded image


188
13.01
431.9
432.5
[M + H]+
3







embedded image


189
13.39
457.88
458.6
[M + H]+
3







embedded image


190
13.98
456.89
457.6
[M + H]+
3







embedded image


191
10.17
397.478
398.4
[M + H]+
3







embedded image


192
13.89
425.55
426.3
[M + H]+
3







embedded image


193
10.07
380.451
381.3
[M + H]+
3







embedded image


194
14.1
472.89
473.3
[M + H]+
3







embedded image


195
11.14
389.88
390.5
[M + H]+
3







embedded image


196
14.03
420.91
421.3
[M + H]+
3







embedded image


197
13.38
457.88
458.5
[M + H]+
3







embedded image


198
13.66
424.88
425.3
[M + H]+
3







embedded image


199
13.34
390.434
391.3
[M + H]+
3







embedded image


200
14.1
472.89
473.3
[M + H]+
3







embedded image


201
13.79
369.468
370.4
[M + H]+
3







embedded image


202
10.82
387.458
388.3
[M + H]+
3







embedded image


203
12.82
403.457
404.6
[M + H]+
3







embedded image


204
14.22
455.35
455.4
[M + H]+
3







embedded image


205
14.13
441.33
441.1
[M + H]+
3







embedded image


206
13.94
456.89
457.6
[M + H]+
3







embedded image


207
12.85
404.9
405.4
[M + H]+
3







embedded image


208
13.96
456.89
457.5
[M + H]+
3







embedded image


209
14
474.88
475.3
[M + H]+
3







embedded image


210
13.64
424.88
425.3
[M + H]+
3







embedded image


211
10.72
385.467
386.4
[M + H]+
3







embedded image


212
10.79
387.458
388.2
[M + H]+
3







embedded image


213
11.14
401.485
402.5
[M + H]+
3







embedded image


214
10.73
373.431
374
[M + H]+
3







embedded image


215
12.53
390.87
391.4
[M + H]+
3







embedded image


216
12.3
403.457
404.6
[M + H]+
3







embedded image


217
11.06
403.91
404.5
[M + H]+
3







embedded image


218
13.65
436.91
437.3
[M + H]+
3







embedded image


219
10.69
373.431
374.4
[M + H]+
3







embedded image


220
12.3
405.88
406.5
[M + H]+
3







embedded image


221
13.51
402.469
403.4
[M + H]+
3







embedded image


222
14.05
420.91
421.3
[M + H]+
3







embedded image


223
7.429
463.93
464.2
[M + H]+
3







embedded image


224
13.72
424.88
425.1
[M + H]+
3







embedded image


225
11.29
369.468
370.4
[M + H]+
3







embedded image


226
11.52
385.467
386.4
[M + H]+
3







embedded image


227
11.59
403.91
404.6
[M + H]+
3







embedded image


228
6.54
378.476
379.4
[M + H]+
3







embedded image


229
11.75
355.441
356.4
[M + H]+
3







embedded image


230
11.71
415.92
416.5
[M + H]+
3







embedded image


231
11.51
355.441
356.4
[M + H]+
3







embedded image


232
10.06
397.478
398.3
[M + H]+
3







embedded image


233
11.58
417.94
418.4
[M + H]+
3







embedded image


234
10.53
355.441
356.2
[M + H]+
3







embedded image


235
13.24
431.9
432.4
[M + H]+
3







embedded image


236
13.33
390.434
391.4
[M + H]+
3









Example 7
Synthesis of Representative Compound 311



embedded image




embedded image


Step 7-1. Synthesis of 4-(N-(1-(4-(difluoromethoxy)benzyl)-1H-indazole-3-carbonyl)-sulfamoyl)benzoic acid (311)



embedded image


To a stirring solution of methyl 4-(N-(1-(4-(difluoromethoxy)benzyl)-1H-indazole-3-carbonyl)sulfamoyl)benzoate (15 mg, 1 Eq, 29 μmol) in THF (2 mL) was added 1M sodium hydroxide (0.29 mL, 10 Eq, 0.29 mmol). The vial was capped and heated at 65° C. overnight. After heating at 65° C. for 14 h, the reaction mixture was cooled to room temperature. The THF layer was removed. To the aqueous layer was added 3M HCl (1 mL). The aqueous layer was extracted with EtOAc (3×5 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford crude material. The crude product was purified by ISCO reversed phase prep system (25-35% formic acid in MeCN and 0.1% formic acid in water) to afford 7 mg (48% yield) of 4-(N-(1-(4-(difluoromethoxy)benzyl)-1H-indazole-3-carbonyl)sulfamoyl)benzoic acid (Compound 311) as a white solid; LCMS (m/z) calculated for C23H17F2N3O4S: 501.1; found 499.8 [M+H]+, tR=9.267 min (Method 1). 1H NMR (400 MHz, DMSO-d6) δ 8.17 (m, 4H), 7.99 (d, J=8 Hz, 1H), 7.85 (d, J=8 Hz, 1H), 7.45 (m, 3H), 7.31 (t, J=8 Hz, 1H), 7.15 (m, 3H), 5.80 (s, 2H).


The compounds listed in Table 7 were made using the procedures of Scheme 7.















TABLE 7






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method









embedded image


311
9.267
501.1
499.8
[M − H]
1









Example 8
Synthesis of Representative Compound 137



embedded image




embedded image


Step 8-1. Synthesis of 1-(4-chlorobenzyl)-N-(2-(4-isopropyl-1H-1,2,3-triazol-1-yl)ethyl)-1H-indazole-3-carboxamide (Compound 137)



embedded image


A solution of CuSO4·5H2O (50.0 μL, 2.82 μmol, 0.02 Eq, 0.0564 M in water) and a solution of sodium L-ascorbic acid (50.0 μL, 14.1 μmol, 0.1 Eq, 0.282 M in water) were added to a mixture of N-(2-azidoethyl)-1-[(4-chlorophenyl)methyl]indazole-3-carboxamide (50.0 mg, 1 Eq, 0.141 mmol) and 3-methylbut-1-yne (15.9 μL, 1.1 Eq, 0.155 mmol) in THE (0.62 mL), t-BuOH (0.38 mL) and H2O (1.90 mL) at room temperature under nitrogen. The mixture was then heated at 50° C. for 72 h. After heating at 50° C. for 72 h, the reaction mixture was cooled to room temperature. The mixture was diluted with aqueous NaHCO3 (50 mL), and the aqueous phase was extracted with DCM (3×75 mL). The combined organic phases were washed with brine (25 mL), dried with sodium sulfate, filtered, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (0->20% MeOH and DCM) to provide 40 mg (63% yield) of 1-(4-chlorobenzyl)-N-(2-(4-isopropyl-1H-1,2,3-triazol-1-yl)ethyl)-1H-indazole-3-carboxamide (Compound 137) as a white solid; LCMS (m/z) calculated for C22H23ClN6O: 422.2; found 422.2 [M+H]+, tR=2.52 min (Method 4). 1H NMR (400 MHz, DMSO-d6) δ 8.54 (t, J=5.9 Hz, 1H), 8.16 (dt, J=8.2, 1.1 Hz, 1H), 7.82 (d, J=0.7 Hz, 1H), 7.79 (d, J=8.6 Hz, 1H), 7.45 (ddd, J=8.5, 6.9, 1.1 Hz, 1H), 7.42-7.22 (m, 5H), 5.75 (s, 2H), 4.53 (t, J=6.4 Hz, 2H), 3.73 (q, J=6.2 Hz, 2H), 2.92 (pd, J=6.9, 0.7 Hz, 1H), 1.17 (d, J=6.9 Hz, 6H).


The compounds listed in Table 8 were made using the procedures of Scheme 8.















TABLE 8






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method









embedded image


137
4.66
422.2
422.2
[M]
4







embedded image


138
4.50
420.2
420.2
[M]
4







embedded image


139
4.09
450.2
450.2
[M]
4







embedded image


140
3.86
438.2
438.2
[M]
4







embedded image


141
5.05
456.2
456.2
[M]
4







embedded image


142
3.23
423.2
423.2
[M]
4









Example 9
Synthesis of Representative Compound 143



embedded image




embedded image


Step 9-1. Synthesis of 1-(4-chlorobenzyl)-N-(2-(5-isopropyl-1H-1,2,3-triazol-1-yl)ethyl)-1H-indazole-3-carboxamide (Compound 143)



embedded image


Cp*RuCl(cod) (10.7 mg, 0.0282 mmol) was added to a mixture of N-(2-azidoethyl)-1-[(4-chlorophenyl)methyl]indazole-3-carboxamide (50.0 mg, 1 Eq, 0.141 mmol) and 3-methylbut-1-yne (72 μL, 5 Eq, 0.71 mmol) in PhMe (4 mL) at room temperature under nitrogen. The mixture was stirred at room temperature for 24 h and concentrated in vacuo. The crude residue was purified by reverse phase chromatography (C18) (10-100% with 10 mM ammonium formate (aq) and MeCN) to afford 19 mg (32% yield) of 1-(4-chlorobenzyl)-N-(2-(5-isopropyl-1H-1,2,3-triazol-1-yl)ethyl)-1H-indazole-3-carboxamide (143) as a white solid. LCMS (m/z) calculated for C22H23ClN6O: 422.2; found 422.2 [M], tR=4.7 min (Method 4). 1H NMR (400 MHz, DMSO-d6) δ 8.60 (t, J=6.0 Hz, 1H), 8.17 (dt, J=8.2, 1.0 Hz, 1H), 7.79 (dt, J=8.6, 0.9 Hz, 1H), 7.52 (d, J=0.5 Hz, 1H), 7.50-7.41 (m, 1H), 7.39 (d, J=8.5 Hz, 2H), 7.28 (ddd, J=7.9, 6.9, 0.9 Hz, 1H), 7.25 (d, J=8.5 Hz, 2H), 5.75 (s, 2H), 4.48 (t, J=6.6 Hz, 2H), 3.71 (q, J=6.4 Hz, 2H), 3.11 (p, J=6.8 Hz, 1H), 1.16 (d, J=6.9 Hz, 6H).


The compound listed in Table 9 was made using the procedures of Scheme 9.















TABLE 9






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method









embedded image


143
4.7
422.2
422.2
[M]
4









Example 10
Synthesis of Representative Compound 144



embedded image




embedded image


Step 10-1. Synthesis of 1-(4-chlorobenzyl)-N-(2-(4-(pyridin-3-yl)-1H-1,2,3-triazol-1-yl)ethyl)-1H-indazole-3-carboxamide (Compound 144)



embedded image


A stirring solution of CuSO4·5H2O (750 μL, 0.0423 mmol, 0.2 Eq, 0.0564 M in water) and a solution of sodium L-ascorbic acid (750 μL, 0.211 mmol, 1 Eq, 0.282 M in water) were added to a stirring mixture of N-(2-azidoethyl)-1-[(4-chlorophenyl)methyl]indazole-3-carboxamide (75.0 mg, 1 Eq, 0.211 mmol), tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA) (22.4 mg, 0.2 Eq, 0.0423 mmol) and 3-ethynylpyridine (109 mg, 5 Eq, 1.06 mmol) in THE (1.5 mL) and MeOH (1.5 mL) at room temperature under nitrogen. The mixture was stirred at room temperature for 18 h. The mixture was diluted with NaOH (1M in H2O, 20 mL), and the aqueous phase was extracted with EtOAc (3×20 mL). The combined organics were washed with brine (25 mL), dried over sodium sulfate (Na2SO4), filtered, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (0→7% MeOH in EtOAc) to afford 73 mg (75% yield) of 1-(4-chlorobenzyl)-N— (2-(4-(pyridin-3-yl)-1H-1,2,3-triazol-1-yl)ethyl)-1H-indazole-3-carboxamide (Compound 144) as a solid; LCMS (m/z) calculated for C24H20ClN7O: 457.1; found 457.1 [M], tR=4.13 mi (Method 4). 1H NR (500 MHz, DMSO-d6) δ 9.03 (dd, J=2.2, 0.8 Hz, 1H), 8.73 (s, 1H), 8.62 (t, J=5.9 Hz, 1H), 8.53 (dd, J=4.8, 1.6 Hz, 1H), 8.21-8.17 (m, 1H), 8.16 (d, J=8.2 Hz, 1H), 7.77 (d, J=8.6 Hz, 1H), 7.48-7.43 (m, 2H), 7.38-7.34 (m, 2H), 7.29-7.23 (m, 3H), 5.74 (s, 2H), 4.67 (t, J=6.2 Hz, 2H), 3.81 (q, J=6.0 Hz, 2H).


The compound listed in Table 10 was made using the procedures of Scheme 10.















TABLE 10






Cpd
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method









embedded image


144
4.13
457.1
457.1
[M]
4







embedded image


145
3.94
458.1
458.1
[M]
4







embedded image


146
3.32
463.2
463.2
[M]
4







embedded image


147
3.75
527.2
527.2
[M]
4









Example 11
Synthesis of Representative Compound 333



embedded image




embedded image


Step 11-1. Synthesis of methyl 1-(4-chlorobenzyl)-1H-indazole-3-carboxylate (INT-11A)



embedded image


1-(Bromomethyl)-4-chloro-benzene (1.40 g, 1.2 Eq, 6.81 mmol) was added to a mixture of methyl 1H-indazole-3-carboxylate (1.00 g, 1 Eq, 5.68 mmol) and Cs2CO3 (3.70 g, 2 Eq, 11.4 mmol) in DMF (6 mL) at room temperature under nitrogen. The mixture was heated at 80° C. for 18 h. After heating the reaction mixture at 80° C. for 18 h, the reaction mixture was cooled to room temperature and diluted with EtOAc (100 mL) and water (100 mL). The aqueous phase was extracted with EtOAc (3×75 mL). The combined organics were washed with brine (50 mL), dried with sodium sulfate, filtered, and concentrated in vacuo. The crude residue was purified by silica gel chromatography (0->75% EtOAc and hexanes) to afford 1.06 g (62% yield) of methyl 1-(4-chlorobenzyl)-1H-indazole-3-carboxylate (INT-11A) as a white solid; LCMS (m/z) calculated for C16H13ClN2O2: 300.1; found 301.2 [M+H]+, tR=2.59 min (Method 4). 1H NMR (400 MHz, CDCl3) δ 8.25 (dt, J=8.1, 1.1 Hz, 1H), 7.40-7.37 (m, 1H), 7.34 (dd, J=1.8, 1.1 Hz, 1H), 7.32 (q, J=1.2 Hz, 1H), 7.29-7.26 (m, 2H), 7.18-7.12 (m, 2H), 5.67 (s, 2H), 4.06 (s, 3H).


Step 11-2. Synthesis of 1-(4-chlorobenzyl)-1H-indazole-3-carboxylic acid (Compound 333)



embedded image


To a mixture of methyl 1-[(4-chlorophenyl)methyl]indazole-3-carboxylate (INT-11A) (3.95 g, 13.1 mmol) in THF (20 mL) and H2O (20 mL) was added NaOH (1.58 g, 39.4 mmol). The reaction mixture was heated at 60° C. for 18 h. After heating for 18 h at 60° C., the reaction mixture was cooled to room temperature. The mixture was acidified with 12 M HCl (pH ˜ 1). The precipitate was filtered, washed with water (100 mL), and dried to afford 3.60 g (96% yield) of 1-(4-chlorobenzyl)-1H-indazole-3-carboxylic acid (Compound 333) as a white solid; LCMS (m/z) calculated for C15H11ClN2O2: 286.1; found 285.1 [M−H], tR=2.39 min (Method 4). 1H NMR (400 MHz, DMSO-d6) δ 13.10 (s, 1H), 8.10 (dt, J=8.2, 1.0 Hz, 1H), 7.84 (dt, J=8.6, 0.8 Hz, 1H), 7.51-7.44 (m, 1H), 7.42-7.37 (m, 2H), 7.35-7.30 (m, 1H), 7.30-7.25 (m, 2H), 5.78 (s, 2H).


The compound listed in Table 11 was made using the procedures of Scheme 11.















TABLE 11






Cpd
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method









embedded image


333
2.39
286.1
285.1
[M − H]
4









Example 12
Synthesis of Representative Compound 148



embedded image




embedded image


Step 12-1. Synthesis of 1-(4-chlorobenzyl)-1H-indazol-3-amine (INT-12A)



embedded image


1H-Indazol-3-amine (1.00 g, 7.51 mmol) was added to a stirring solution of KOH (843 mg, 15.0 mmol) in DMSO (6.0 mL) at room temperature. After stirring the mixture for 30 min at room temperature, 1-(Bromomethyl)-4-chloro-benzene (1.62 g, 7.89 mmol) was added to the mixture. After stirring at room temperature for 24 h, the mixture was diluted with DCM (100 mL) and water (100 mL). The aqueous phase was extracted with DCM (3×75 mL). The combined organics were washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by silica gel chromatography (0->15% MeOH in DCM) to provide 1.04 g (54% yield) of 1-(4-chlorobenzyl)-1H-indazol-3-amine (INT-12A) as a white solid; LCMS (m/z) calculated for C24H12ClN3: 257.1; found 259.2 [M+H]+, tR=2.33 min (Method 4). 1H NMR (500 MHz, CDCl3) δ 7.55 (dt, J=8.1, 1.0 Hz, 1H), 7.36-7.29 (m, 1H), 7.24 (d, J=8.5 Hz, 2H), 7.18 (dt, J=8.5, 0.9 Hz, 1H), 7.11 (d, J=8.3 Hz, 2H), 7.04 (ddd, J=8.0, 6.9, 0.9 Hz, 1H), 5.32 (s, 2H), 4.08 (s, 2H).


Step 12-2. Synthesis of N-(1-(4-chlorobenzyl)-1H-indazol-3-yl)-3-(pyridin-3-yl)-propanamide (Compound 148)



embedded image


To a stirring mixture of 3-(3-pyridyl)propanoic acid (INT-12A) (100 mg, 0.662 mmol) in DCM (5 mL) at room temperature was added DIPEA (230 μL, 1.32 mmol) followed by HATU (377 mg, 0.992 mmol) and 1-[(4-chlorophenyl)methyl]indazol-3-amine (205 mg, 0.794 mmol). After stirring at room temperature for 18 h, the reaction mixture was concentrated in vacuo. The residue was purified by reverse phase chromatography (C18) (10->100% 0.1% formic acid in MeCN and 0.1% formic acid in H2O) to afford 27 mg (10% yield) of N-(1-(4-chlorobenzyl)-1H-indazol-3-yl)-3-(pyridin-3-yl)propenamide (Compound 148) as a white solid; LCMS (m/z) calculated for C22H19ClN3: 390.1; found 392.6 [M+H]+, tR=3.91 min (Method 4). 1H NMR (400 MHz, DMSO-d6) δ 10.48 (s, 1H), 8.58 (s, 1H), 8.48 (d, J=4.0 Hz, 1H), 7.86 (d, J=7.9 Hz, 1H), 7.72 (dt, J=8.2, 1.0 Hz, 1H), 7.69-7.62 (m, 1H), 7.45 (dd, J=7.9, 4.9 Hz, 1H), 7.41-7.31 (m, 3H), 7.23 (d, J=8.5 Hz, 2H), 7.07 (dd, J=8.3, 6.8 Hz, 1H), 5.54 (s, 2H), 3.00 (t, J=7.5 Hz, 2H), 2.76 (t, J=7.5 Hz, 2H).


The compound listed in Table 12 was made using the procedures of Scheme 12.















TABLE 12






Cpd
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method









embedded image


148
3.91
390.1
392.6
[M + H]+
4









Example 13
Synthesis of Representative Compound 237



embedded image




embedded image


Step 13-1. 1-(4-chlorobenzyl)-1H-indole (INT 13-A)



embedded image


A 250 mL round bottom flask containing methyl 1H-indole (5.0 g, 1.0 equiv., 42.7 mmol) in DMF (15 mL) was added 60% NaH in oil (1.79 g, 1.0 equiv., 42.7 mmol) and then stirred at rt for 5 min. 1-(bromomethyl)-4-chlorobenzene (8.77 g, 1.0 equiv., 42.7 mmol) in DMF (5 mL) was slowly added. Stirred at rt for 20 min. Monitored by LCMS. 20 min: Coupling complete. Quenched with water (100 mL) slowly at 1 h. Product was extracted with DCM (300 mL), washed with brine (200 mL), dried over Na2SO4, filtered, and concentrated. The resulting crude residue was purified by silica gel chromatography using ISCO eluting with (0-10% EtOAc/hexanes) to yield 7.0 g (68%) of 1-(4-chlorobenzyl)-1H-indole (INT-13A). LCMS (m/z) calculated for C15H12ClN: 241.07; found 242.3 [M+H]+, tR=5.70 min (Method 3).


Step 13-2. Methyl 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)thio)acetate (INT 13-B)



embedded image


A 250 mL round bottom flask containing 1-(4-chlorobenzyl)-1H-indole (INT-13A) (3.0 g, 1.0 equiv., 1.24 mmol), and methyl 2-mercaptoacetate (1.32 g, 1.0 equiv., 1.24 mmol) in MeOH (80 mL) and distilled water (20 mL) was added iodine (3.15 g, 2.5 equiv., 3.15 mmol), followed by KI (2.06 g, 1.0 equiv., 1.24 mmol). The mixture was stirred at rt for 16 h and then concentrated in vacuo. The remaining aqueous solution was diluted with saturated aqueous NaHCO3 (100 mL) and product was extracted with EtOAc (200 mL). The organic solution was dried over Na2SO4, filtered and concentrated. The resulting crude residue was purified by silica gel chromatography using ISCO eluting with 0-100% EtOAc/hexanes to yield 2.51 g (59% yield) of methyl 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)thio)acetate (INT-13B). LCMS (m/z) calculated for C18H16ClNO2S: 345.06; found 346.4 [M+H]+, tR=5.52 min (Method 3).


Step 13-3. Methyl 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)acetate (INT-13C)



embedded image


A 100-mL round bottom flask containing methyl 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)thio)acetate (INT-13B) (2.1 g, 1.0 equiv., 6.07 mmol) in DCM (20 ml) was added m-CPBA (2.79 g, 2.0 equiv., 12.1 mmol). The mixture was stirred at rt for 1 h at rt which point LCMS analysis indicated reaction reached completion. The reaction was quenched with saturated aqueous NaHCO3 (100 mL) and extracted with DCM (150 mL). The organic phase was dried over Na2SO4, filtered, and concentrated under reduced pressure. The resulting crude residue was purified by silica gel chromatography using ISCO eluting with 0-100% EtOAc/hexanes to yield 1.52 g (66%) of methyl 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)acetate (INT-13C). LCMS (m/z) calculated for C18H16ClNO4S: 377.05; found 378.3 [M+H]+, tR=4.40 min (Method 3).


Step 13-4. 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)acetic acid (INT-13D)



embedded image


A 250 mL round bottom flask containing methyl 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)acetate (INT-13C) (1.52 g, 1.0 equiv., 4.0 mmol) in MeOH (20 ml) was added NaOH (0.81 g, 5 equiv., 20 mmol) in water (9 mL). The mixture was stirred at 80° C. for 30 minutes at which point LCMS analysis indicated consumption of starting material. PH of the reaction mixture was adjusted to 1 with the addition of 2 N HCl(aq).


The product was extracted with EtOAc (100 mL) and concentrated under vacuo. The crude residue was purified by recrystallization from EtOAc/hexanes to yield 1.3 g (89% yield) of chlorobenzyl)-1H-indol-3-yl)sulfonyl)acetic acid (INT-13D). LCMS (m/z) calculated for C17H14ClNO4S: 363.03; found 364.4 [M+H]+, tR=11.57 min (method 3).


Step 13-5. 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)-N-(2-fluorophenyl)acetamide (Compound 237)



embedded image


A 25-mL round bottom flask containing 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)acetic acid, (INT-13D), (75 mg, 1.0 equiv., 0.21 mmol) and HATU (86 mg, 1.1 equiv., 2.3 mmol) was added DMF (6 mL). The mixture was stirred at rt and 2-fluoroaniline (25 mg, 1.1 equiv., 23 mmol) was added followed by DIEA (83 mg, 3.1 equiv., 64 mmol). The reaction mixture was stirred at rt for 16 h at which point LCMS analysis indicated consumption of starting material. The solution was diluted with EtOAc (100 mL) and washed consecutively with 75 mL each 1 M NaOH (aq), 1 M HCl(aq), and brine. The organic solution was dried over Na2SO4, filtered and concentrated. The resulting crude residue was purified by silica gel chromatography eluting with 0-100% EtOAc/hexanes to afford 33 mg (35% yield) of 2-((1-(4-chlorobenzyl)-1H-indol-3-yl)sulfonyl)—N-(2-fluorophenyl) acetamide (Compound 237). LCMS (m/z) calculated for C23H18ClFN2O3S: 456.07; found 457.5 [M+H]+, tR=11.74 min (Method 3). 1H NMR (400 MHz, DMSO-d6) δ 10.03 (s, 1H), 8.31 (s, 1H), 7.84 (d, J=8 Hz, 1H), 7.77 (d, J=4 Hz, 1H), 7.57 (d, J=8 Hz, 1H), 7.34-7.12 (m, 9H), 5.56 (s, 2H), 4.53 (s, 2H).


The compounds listed in Table 13 were made using the procedures of Scheme 13.















TABLE 13






Cpd.
Purity

Observed

Purity


Structure
No.
RT (min)
MW
m/z
Ion
Method























embedded image


237
12.64
456.92
457.4
[M + H]+
3







embedded image


238
12.73
468.95
469.4
[M + H]+
3







embedded image


239
11.26
439.91
440.3
[M + H]+
3







embedded image


240
12.88
456.92
457.6
[M + H]+
3







embedded image


241
12.32
439.91
440.2
[M + H]+
3







embedded image


242
12.66
438.93
439.1
[M + H]+
3







embedded image


243
10.401
452.95
453.2
[M + H]+
3







embedded image


244
13.01
473.37
473.3
[M + H]+
3







embedded image


245
13.91
456.92
457.6
[M + H ]+
3







embedded image


246
12.87
468.95
469.4
[M + H]+
3







embedded image


247
13.16
452.95
453.4
[M + H]+
3







embedded image


248
12.21
404.91
405.6
[M + H]+
3







embedded image


314
13.92
477.38
477.1
[M + H]+
3







embedded image


249
13.76
507.81
509.5
[M + H]+
3







embedded image


250
11.58
376.86
377.3
[M + H]+
3







embedded image


251
13.65
507.81
507.1
[M + H]+
3







embedded image


252
13.09
390.88
391.4
[M + H]+
3







embedded image


253
31.25
473.37
473.3
[M + H]+
3







embedded image


254
10.49
461.53
462.5
[M + H]+
3







embedded image


255
12.84
452.95
453.3
[M + H]+
3







embedded image


256
13.33
473.37
473.2
[M + H]+
3









Example 14
Synthesis of Representative Compound 312



embedded image




embedded image


Step 14-1. Methyl 1-(4-(difluoromethoxy) benzyl)-1H-indole-3-carboxylate (INT-14A)



embedded image


A 50-mL round bottom flask methyl 1H-indole-3-carboxylate (309 mg, 1.0 equiv., 1.75 mmol) in DMF (5 mL) was added 60% NaH in oil (95 mg, 1.5 equiv., 2.38 mmol) and then stirred at rt for 5 min. To this mixture was added 1-(bromomethyl)-4-(difluoromethoxy) benzene (300 mg, 1.1 equiv., 1.59 mmol). The reaction was stirred at rt for 2 h (additional 20 mg 1-(bromomethyl)-4-(difluoromethoxy)benzene added a 30 min). The reaction was quenched with saturated aqueous NH4Cl (50 mL). Product was extracted with EtOAc (60 mL) and organic phase was washed with brine (30 mL), dried over Na2SO4, filtered, and concentrated. The resulting crude residue was purified by silica gel chromatography using ISCO eluting with 0-10% MeOH/DCM to yield 400 mg (76% yield) of methyl 1-(4-(difluoromethoxy)benzyl)-1H-indole-3-carboxylate (INT-14A). LCMS (m/z) calculated for C18H15F2NO3: 331.10; found 332.4 [M+H]+, tR=4.94 min (method 3).


Step 14-2. 1-(4-(difluoromethoxy)benzyl)-1H-indole-3-carboxylic acid (INT-14B)



embedded image


To a 100-mL round bottom flask containing a solution of methyl 1-(4-(difluoromethoxy)benzyl)-1H-indole-3-carboxylate, (INT-14A), (400 mg, 1.0 equiv., 1.21 mmol) in MeOH (40 mL) was added an aqueous solution of 1 M NaOH (6.0 mL, 5 equiv., 6.0 mmol) and H2O (5 mL). The mixture was stirred at 100° C. for 1.5 h. The reaction was concentrated to remove MeOH, then diluted with water (50 mL). The solution was acidified to pH 1 using 1 M HCl(aq). Product was extracted with EtOAc (60 mL), and the organic solution was washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated to yield 330 mg (86% yield) of 1-(4-(difluoromethoxy)benzyl)-1H-indole-3-carboxylic acid (INT-14B). LCMS (m/z) calculated for C17H13F2NO3: 317.09; found 318.3 [M+H]+, tR=4.38 min (method 3).


Step 14-3. 1-(4-(difluoromethoxy)benzyl)-N-(o-tolylsulfonyl)-1H-indole-3-carboxamide (Compound 312)



embedded image


To a 50-mL round bottom flask containing 1-(4-(difluoromethoxy)benzyl)-1H-indole-3-carboxylic acid, (INT-14B), (50 mg, 1.0 equiv., 0.16 mmol) was added SOCl2 (5 mL) and stirred at rt for 5 min. Volatile was removed under vacuo, and the residue was added 2-methylbenzenesulfonamide (30 mg, 1.1 equiv., 0.17 mmol), DMAP (21 mg, 1.1 equiv., 0.17 mmol), and anhydrous DCM (5 mL). At 1 h, 2-methylbenzenesulfonamide (30 mg, 1 equiv., 0.17 mmol) was added followed by Et3N (50 μmol). After an additional 1 h, solvents were removed under vacuo. The residue was diluted with EtOAc (50 mL) and washed consecutive with 1 M HCl (aq., 30 mL), 1 M NaOH (aq., 30 mL), saturated NaHCO3 (aq., 30 mL), and brine (aq., 30 mL), dried over Na2SO4, filtered, and concentrated. The resulting crude residue was purified by silica gel chromatography using ISCO eluting with 0-100% EtOAc/hexanes. Product fractions were combined, concentrated and the residue was dissolved in CH3CN/H2O and lyophilized to yield 26 mg (36%) of 1-(4-(difluoromethoxy)benzyl)-N-(o-tolylsulfonyl)-1H-indole-3-carboxamide (Compound 312) as a solid. LCMS (m/z) calculated for C24H20F2N2O4S: 470.11; found 471.4 [M+H]+, tR=11.81 min (method 3). 1H NMR (400 MHz, DMSO-d6) δ 12.12 (s, 1H), 8.56 (s, 1H), 8.06 (d, J=8 Hz, 1H), 7.96 (d, J=8 Hz, 1H), 7.61-7.55 (m, 2H), 7.46 (t, J=8 Hz, 1H), 7.41-7.03 (m, 8H), 5.50 (s, 2H), 2.65 (s, 3H).


The compounds listed in Table 14 were made using the procedures of Scheme 14.















TABLE 14






Cpd.
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method









embedded image


312
13.2
470.49
471.4
[M + H]+
3







embedded image


313
13.06
470.49
471.5
[M + H]+
3









Example 15
Synthesis of Compound 315



embedded image




embedded image


Step 15-1. 1H-indol-3-yl carbamimidothioate (INT-15A)



embedded image


To a stirring solution of 1H-indole (2.0 g, 1.0 equiv., 17.0 mmol) and thiourea (1.3 g, 1.0 equiv., 17.0 mmol) in MeOH (60 mL) was added dropwise a solution of potassium iodide (2.8 g, 1.0 equiv., 17.0 mmol) and diiodine (4.3 g, 1.0 equiv., 17.0 mmol) in water (60 mL). Required additional MeOH (20 mL) and H2O (20 mL) to dissolve the iodine. The reaction mixture was stirred at rt for 2 h. TLC of starting indole Rf=0.80 (3:2 EtOAc/hexanes). LCMS showed reaction completion. The mixture was filtered, and the filtrate was concentrated in vacuo to afford 1.6 g (63% yield) of 1H-indol-3-yl carbamimidothioate (INT-15-A) as a white solid that was carried on without further purification. LCMS (m/z) calculated for C9H9N3S: 191.2; found 192.1 [M+H]+, tR=1.63 min (method 2).


Step 15-2. 1H-indole-3-thiol (INT-15B)



embedded image


To 1H-indol-3-yl carbamimidothioate, (INT-15A), (3.3 g, 1.0 equiv., 17.0 mmol) was added 2N NaOH (60 mL). The reaction mixture was heated at 85° C. for 30 min. LCMS showed reaction completion. The mixture was cooled to rt with an ice bath and acidified with 6N HCl (25 mL). The resulting precipitate was filtered, washed with water (2×60 mL), filtered, and dried in vacuo to afford crude product. The crude product was purified by silica gel chromatography (0->50% EtOAc in hexanes) to afford 1.6 g (63% yield) 1H-indole-3-thiol (INT-15B) as a white solid. LCMS (m/z) calculated for C8H7NS: 149.2; found 150.1 [M+H]+, tR=3.97 min (Method 2).


Step 15-3. 2-((1H-indol-3-yl)thio)-1-(pyrrolidin-1-yl)ethan-1-one (INT-15C)



embedded image


To a stirring solution of 1H-indole-3-thiol, (INT-15B), (50.0 mg, 1.0 equiv., 0.34 mmol) in acetonitrile (2 mL) was added potassium carbonate (460.0 mg, 10.0 equiv., 3.4 mmol) followed by 2-chloro-1-(pyrrolidin-1-yl)ethan-1-one (49.0 mg, 10.0 equiv., 0.34 mmol). The vial was capped and heated at 85° C. overnight. LCMS showed reaction completion. Filtered through Celite rinsing with EtOAc. Purified by silica gel chromatography (0-100% 10% MeOH in EtOAc and hexanes) to afford 2-((1H-indol-3-yl)thio)-1-(pyrrolidin-1-yl)ethan-1-one (INT-15C) as a solid. LCMS (m/z) calculated for C14H16N2O2S: 260.3; found 261.1 [M+H]+, tR=6.61 min (Method 2).


Step 15-4. 2-(4(1H-indol-3-yl)thio)-1-(pyrrolidin-1-yl)ethan-1-one (Compound 315)



embedded image


To a stirring solution of 2-((1H-indol-3-yl)thio)-1-(pyrrolidin-1-yl)ethan-1-one, (INT 15-C), (78.7 mg, 1.0 equiv., 0.3 mmol) in 15 mL DMF was added sodium hydride (15 mg, 60% wt, 1.2 equiv., 0.36 mmol) followed by 1-bromo-4-(bromomethyl)benzene (90.7 mg, 1.2 equiv., 0.36 mmol). The reaction mixture was stirred at rt for 4 h. LCMS showed reaction completion. Saturated ammonium chloride was added dropwise, the aqueous layer was extracted with EtOAc (3×3 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford crude product. 56.6 mg colorless oil. Purified by silica gel chromatography (0-70% 10% MeOH in EtOAc and hexanes). Impurity came off right before it but had a much higher Rf Impurity with a similar retention time on the LCMS. 56.6 mg colorless oil. Added ˜0.25 mL EtOAc followed by ˜0.25 mL of Et2O. The product crystallized to yield 57 mg (44% yield) of 2-(4(1H-indol-3-yl)thio)-1-(pyrrolidin-1-yl)ethan-1-one (Compound 315) as a solid. LCMS (m/z) calculated for C21H21BrN2OS: 429.8; found 431.1 [M+H], tR=10.43 min (Method 2).


Example 16
Synthesis of Compound 530



embedded image




embedded image


Step 16-1. Synthesis of 1-(4-chlorobenzyl)-5-methyl-1H-indazole-3-carboxylic acid



embedded image


To a stirring solution of 5-methyl-1H-indazole-3-carboxylic acid (200 mg, 1 Eq, 1.14 mmol) at 0° C. in DMF (2 mL) was added NaH (60% in mineral oil, 84 mg, 3.1 Eq, 3.5 mmol). After stirring for 15 min at rt, a solution of 1-chloro-4-(chloromethyl)benzene (192 mg, 1.05 eq, 1.2 mmol) in DMF (2 mL) was added. After 1 h, the reaction mixture was quenched with H2O (5 mL) and washed with EtOAc (3×10 mL). The aqueous layer was pH adjusted to pH 2 (6 N HCl) and then extracted with EtOAc. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo to afford crude material. The crude material was purified by SiO2 chromatography (0-10% MeOH/DCM) to afford 200 mg (59% yield) of 1-(4-chlorobenzyl)-5-methyl-1H-indazole-3-carboxylic acid (INT-16AA) as a white solid; LCMS (m/z) calculated for C16H13ClN2O2: 300.07; found 301.0 [M+H]+, tR=6.36 min (Method 3).


Step 16-2. Synthesis of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1-(4-chlorobenzyl)-5-methyl-1H-indazole-3-carboxamide



embedded image


To a stirring solution of 1-(4-chlorobenzyl)-5-methyl-1H-indazole-3-carboxylic acid (100 mg, 1 Eq, 0.33 mmol) in DMF (0.5 mL) was added HATU (139 mg, 1.1 Eq, 0.37 mmol). After 15 min, 2-([1,2,4] triazolo[4,3-a]pyridin-3-yl)ethan-1-amine (59 mg, 1.1 eq, 0.37 mmol) and DIEA (129 mg, 3 eq, 1 mmol) were added. After 3 h, the reaction mixture was quenched with water and extracted with EtOAc. The organic layer was separated, dried over sodium sulfate, filtered, and concentrated in vacuo to afford crude material. The crude product was purified by silica gel column chromatography (0-10% MeOH/DCM) to afford 25 mg (17% yield) of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1-(4-chlorobenzyl)-5-methyl-1H-indazole-3-carboxamide (Compound 530) as a white solid; LCMS (m/z) calculated for C24H21ClN6O: 444.1; found 444.9 [M+H]+, tR=12.03 min (Method 3).


The compounds listed in Table 16 were made using the procedures of Scheme 16.















TABLE 16






Cpd.
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method























embedded image


530
12.03
444.92
444.9
[M + H]+
3







embedded image


531
11.79
482.443
483.1
[M + H]+
3







embedded image


539
12.91
465.34
464.5
[M + H]+
3







embedded image


540
11.6
448.89
448.8
[M + H]+








embedded image


541
12.12
509.79
508.9
[M + H]+
3







embedded image


542
10.02
455.91
456.3
[M + H]+
3







embedded image


560
11.92
443.94
443.9
[M + H]+
3







embedded image


561
11.87
443.94
444
[M + H]+
3







embedded image


562
12.62
465.34
464.8
[M + H]+
3







embedded image


563
11.69
460.92
461.1
[M + H]+
3







embedded image


564
12.61
498.89
498.4
[M + H]+
3







embedded image


565
11.99
444.92
444.8
[M + H]+
3







embedded image


566
10.96
509.79
511
[M + H]+
3









Example 17
Synthesis of Compound 532



embedded image




embedded image


Step 17-1. Synthesis of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-5-fluoro-1H-indazole-3-carboxamide



embedded image


To a vial containing 5-fluoro-1H-indazole-3-carboxylic acid (50 mg, 1 Eq, 0.28 mmol) was added thionyl chloride (84 mg, 20 Eq, 5.5 mmol). After stirring for 1 h at 80° C., the reaction mixture was concentrated in vacuo. The resulting residue was diluted with DCM and re-concentrated multiple times. The resulting precipitate was dissolved in DCM (1 mL) and 2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethan-1-amine (45 mg, 1.0 eq, 0.28 mmol) and TEA (84 mg, 3 eq, 0.83 mmol) were added. After 0.5 h, the reaction mixture was filtered to collect the solid precipitate, which was washed with DCM, acetone, water, acetone and DCM, followed by air drying to provide 48 mg (53% yield) of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-5-fluoro-1H-indazole-3-carboxamide (INT-17BA) as a light gray solid; LCMS (m/z) calculated for C16H13FN6O: 324.1; found 325.0 [M+H]+, tR=4.75 min (Method 3).


Step 17-2. Synthesis of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-5-fluoro-1-(naphthalen-2-ylmethyl)-1H-indazole-3-carboxamide



embedded image


Into a vial containing N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-5-fluoro-1H-indazole-3-carboxamide (48 mg, 1.0 eq, 0.15 mmol) was added DMF (1 mL), followed by NaH (60% solution in mineral oil, 6.5 mg, 1.1 eq, 0.16 mml). After bubbling ceased, 2-(bromomethyl)naphthalene (33 mg, 1.0 eq, 0.15 mmol) was added. After 15 min, the reaction was quenched with water and extracted with DCM (3X). The combined organic extracts were dried over sodium sulfate, filtered, and concentrated in vacuo to afford crude material. The crude product was purified by SiO2 column chromatography (0-100% 91% MeOH/in EtOAc/DCM) to afford 24 mg (35% yield) of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-5-fluoro-1-(naphthalen-2-ylmethyl)-1H-indazole-3-carboxamide (Compound 532) as a pale yellow solid; LCMS (m/z) calculated for C27H21FN6O: 464.2; found 465 [M+H]+, tR=12.19 min (Method 3).


The compounds listed in Table 17 were made using the procedures of Scheme 17.















TABLE 17






Cpd.
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method









embedded image


532
12.07
464.504
464.7
[M + H]+
3









Example 18
Synthesis of Representative Compound 402



embedded image




embedded image




embedded image


Step 18-1. Synthesis of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide

Into a solution of 1H-indazole-3-carboxylic acid (5.34 g, 1 eq, 32.91 mmol) in DMF (50 mL) were added EDCI (7.57 g, 1.2 eq, 39.5 mmol), HOBt (5.34 g, 1.2 eq, 39.49 mmol) and TEA (9.99 g, 3 eq, 98.72 mmol, 13.74 mL) at 25° C., followed by 2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethanamine (8.0 g, 1 eq, 32.91 mmol, HBr salt). The mixture was stirred at 25° C. for 2 h. The reaction mixture was filtered, and the filter cake was washed with MTBE (3×100 mL) and H2O (100 mL). The resulting filter cake was collected and dried in vacuo to provide 4 g (40% yield) of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide (INT-18CA) as a white solid; LCMS (m/z) calculated for C16H14N6O: 306.1; found 307.1 [M+H]+, tR=0.28 min (Method 5). 1H NMR (400 MHz, DMSO-d6) δ 13.58 (s, 1H), 8.62 (t, J=6.0 Hz, 1H), 8.51-8.48 (m, 1H), 8.16 (d, J=8.3 Hz, 1H), 7.73 (td, J=1.0, 9.3 Hz, 1H), 7.61 (d, J=8.4 Hz, 1H), 7.44-7.39 (m, 1H), 7.36-7.32 (m, 1H), 7.26-7.22 (m, 1H), 6.97 (dt, J=0.9, 6.7 Hz, 1H), 3.79 (q, J=6.9 Hz, 2H), 3.40 (t, J=7.0 Hz, 2H).


Step 18-2. Synthesis of 2-cyclohexylethyl methanesulfonate



embedded image


Into a solution of 2-cyclohexylethanol (100 mg, 1 eq, 0.78 mmol, 109 μL) in DCM (5 mL) were added DMAP (9.5 mg, 0.1 eq, 0.078 mmol) and TEA (237 mg, 3 eq, 2.34 mmol, 326 μL). Methylsulfonyl methanesulfonate (150 mg, 1.1 eq, 858 μmol) was added at 0° C. and then the mixture was stirred at 25° C. for 2 h. The reaction mixture was diluted with H2O (20 mL) and then extracted with DCM (3×10 mL). The combined organic layers were washed with saturated NaCl (2×10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to afford 160 mg (99% yield) of 2-cyclohexylethyl methanesulfonate as a colorless oil that was used without further purification; TLC (EA, ninhydrin stain) Rf=0.40.


Step 18-3. Synthesis of N-(2-([1,2,4]triazolo[4,3-a] pyridin-3-yl)ethyl)-1-(2-0cyclohexylethyl)-1H-indazole-3-carboxamide



embedded image


Into a solution of N-[2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl]-1H-indazole-3-carboxamide (20 mg, 1 eq, 65.29 μmol) and 2-cyclohexylethyl methanesulfonate (13.47 mg, 1 eq, 65.29 μmol) in NMP (1 mL) was added t-BuOK (18.32 mg, 2.5 eq, 163.23 μmol). The reaction was stirred at 80° C. for 2 The reaction mixture was filtered through a filter membrane. The filtrate was purified by reverse phase prep-HPLC (ACN/H2O with formic acid). The resulting material was lyophilized to afford 11.6 mg (43% yield) of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1H-indazole-3-carboxamide (Compound 402) as a pale brown solid; LCMS (m/z) calculated for C24H28N6O: 416.2; found 417.2 [M+H]+, tR=0.46 min (Method 5). 1H NMR (400 MHz, DMSO-d6) δ=8.49 (d, J=7.1 Hz, 2H), 8.15 (d, J=8.3 Hz, 1H), 7.73 (t, J=8.3 Hz, 2H), 7.47-7.42 (m, 1H), 7.33 (ddd, J=0.9, 6.5, 9.3 Hz, 1H), 7.26 (t, J=7.4 Hz, 1H), 6.98-6.93 (m, 1H), 4.51-4.45 (m, 2H), 3.77 (q, J=6.9 Hz, 2H), 3.39 (br t, J=7.1 Hz, 2H), 1.79-1.71 (m, 4H), 1.65 (br d, J=5.9 Hz, 2H), 1.60 (br d, J=4.0 Hz, 1H), 1.22 (td, J=3.6, 10.8 Hz, 1H), 1.18-1.10 (m, 3H), 1.00-0.89 (m, 2H).


The compounds listed in Table 18 were made using the procedures of Scheme 18.















TABLE 18






Cpd.
Purity RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method























embedded image


402
0.46
4A.529
417.2
[M + H]+
5







embedded image


485
0.468
460.92
461.3
[M + H]+
5







embedded image


486
0.383
456.506
457.2
[M + H]+
5







embedded image


487
0.507
4A.529
417.3
[M + H]+
5







embedded image


488
0.471
402.502
403.3
[M + H]+
5







embedded image


489
0.5435
438.483
439.2
[M + H]+
5







embedded image


504
0.481
402.502
403.3
[M + H]+
5







embedded image


506
0.47
454.534
455.3
[M + H]+
5









Example 19
Synthesis of Compound 445



embedded image




embedded image


Step 19-1. Synthesis of 2-cyclohexylethyl methanesulfonate



embedded image


A solution of (4-ethynylphenyl)methanol (50 mg, 1 eq, 0.38 mmol) in SOCl2 (1 mL) was stirred at 80° C. for 2 h. The reaction mixture was concentrated in vacuo to afford 50 mg (88% yield) of 1-(chloromethyl)-4-ethynyl-benzene as a yellow solid that was used without further purification; TLC (5:1 Pet ether/EA) Rf=0.70.


Step 19-2 Synthesis of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1-(4-ethynylbenzyl)-1H-indazole-3-carboxamide



embedded image


Into a solution of 1-(chloromethyl)-4-ethynyl-benzene (15 mg, 1 eq, 98 μmol) and N-[2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl]-1H-indazole-3-carboxamide (30 mg, 1 eq, 98 μmol) in NMP (1 mL) was added Cs2CO3 (96 mg, 3 eq, 294 μmol). After stirring at 50° C. for 2 h, the reaction mixture was directly purified using reverse-phase prep HPLC (H2O (formic acid)/ACN) to afford 24 mg (58% yield) of N-(2-([1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-1-(4-ethynylbenzyl)-1H-indazole-3-carboxamide (Compound 445) as an off-white solid; LCMS (m/z) calculated for C25H20N6O: 420.2; found 421.2 [M+H]+, tR=0.41 min (Method 5). 1H NMR (400 MHz, DMSO-d6) δ=8.61 (t, J=6.0 Hz, 1H), 8.49 (d, J=7.0 Hz, 1H), 8.18 (d, J=8.1 Hz, 1H), 7.75 (dd, J=8.9, 19.8 Hz, 2H), 7.48-7.42 (in, 3H), 7.35-7.26 (m, 2H), 7.22 (d, J=8.3 Hz, 2H), 6.94 (t, J=6.8 Hz, 1H), 5.78 (s, 2H), 4.18 (s, 1H), 3.78 (br d, J=6.4 Hz, 2H), 3.40 (t, J=7.1 Hz, 2H).


The compounds listed in Table 19 were made using the procedures of Scheme 19.















TABLE 19







Purity







Cpd.
RT

Observed

Purity


Structure
No.
(min)
MW
m/z
Ion
Method























embedded image


413
0.444
438.491
439.3
[M + H]+
6







embedded image


430
0.334
426.48
427.2
[M + H]+
5







embedded image


435
0.356
474.54
475.2
[M + H]+
5







embedded image


445
0.407
420.476
421.2
[M + H]+
5







embedded image


446
0.369
421.464
422.2
[M + H]+
5







embedded image


447
0.364
424.508
425.2
[M + H]+
5







embedded image


449
0.526
486.579
487.1
[M + H]+
5







embedded image


450
0.755
476.544
477.2
[M + H]+
6







embedded image


451
0.305
439.523
440.3
[M + H]+
6







embedded image


458
0.36
447.502
448.1
[M + H]+
5







embedded image


459
0.456
482.443
483.2
[M + H]+
6







embedded image


473
0.378
476.54
477.2
[M + H]+
5







embedded image


474
0.492
446.514
447.1
[M + H]+
5







embedded image


476
0.291
436.479
437.3
[M + H]+
5







embedded image


477
0.418
454.51
455.1
[M + H]+
5







embedded image


478
0.423
463.501
464.1
[M + H]+
5







embedded image


479
0.479
438.535
439.3
[M + H]+
5







embedded image


480
0.356
464.533
465.3
[M + H]+
5







embedded image


490
0.494
436.519
437
[M + H]+
5







embedded image


491
0.452
438.491
439.2
[M + H]+
5







embedded image


492
0.325
440.511
441.3
[M + H]+
5







embedded image


493
0.445
436.475
437.2
[M + H]+
5







embedded image


494
0.478
436.519
437.3
[M + H]+
5







embedded image


495
0.461
452.54
453.3
[M + H]+
5







embedded image


496
0.48
436.475
437.2
[M + H]+
5







embedded image


497
0.456
453.52
454.1
[M + H]+
5







embedded image


167
0.43
462.517
463.3
[M + H]+
5







embedded image


499
0.499
509.59
510.2
[M + H]+
5







embedded image


500
0.459
507.51
508.3
[M + H]+
5







embedded image


501
0.455
471.46
472.1
[M + H]+
5







embedded image


502
0.361
515.375
5A.9
[M + H]+
5







embedded image


509
0.485
482.443
483.2
[M + H]+
5







embedded image


512
0.473
479.56
480.3
[M + H]+
5









Example 20
MRGPR X4 Activity

HEK cells stably transfected to express human MIRGPR X4 were maintained in an incubator at 37° C. with 5% CO2 and grown in DMEM media with 10% fetal bovine serum (FBS) and 100 each of sodium pyruvate, Glutamax, penicillin/streptomycin, and Geneticin. HEK cells stably transfected to express mouse MRGPR A1 were maintained in the same incubator and grown in DMEM media with 10% FBS, 1% each of sodium pyruvate, Glutamax, penicillin/streptomycin, Geneticin, and 2.2 mg/mL Hygromycin.


Cells were plated in a 384-well assay plate at 20,000 cells per well in 12 μL of Opti-MEM and kept in an incubator overnight. On the day of the assay, compounds solubilized at 10 mM in DMSO were added as a 10-point curve (10 uM final top concentration with 1:3 serial dilutions) using a Tecan D300E digital dispenser. Agonists were diluted in assay buffer (final concentrations of 5.7 mM Tris-HCl, 43 mM NaCl, 50 mM LiCl, pH=8) and 2 μL of the appropriate agonist are added to each well. Final concentrations of agonists were 10 μM bilirubin, 20 μM deoxycholic acid, or 100 μM conjugated bilirubin (obtained from Lee Biosolutions, catalog #910-12). Final concentrations of DMSO were kept consistent across the plate. Plates were incubated in the dark for 1 h at 37° C. and then for 30 minutes at room temperature. IP-1 standards and HTRF detection reagents were added according to the IP-One—Gq Kit purchased from Cisbio (part number 62IPAPEJ) and incubated in the dark for 1 h at room temperature. The plate was read on a Molecular Devices SpectraMax iD5 plate reader. The HTRF ratio was calculated from the raw data and graphed using GraphPad Prism to calculate an IC50 value for each compound.


Activity data for selected MRGPR X4 antagonists (versus 20 μM deoxycholic acid agonist) are displayed in Table B. The activity ranges are denoted as follows: “+++++” denotes antagonist activity <100 nM; “++++” denotes antagonist activity between 100 and 500 nM; “+++” denotes activity between 501 and 1000 nM; “++” denotes activity between 1001 and 2500 nM; and “+” denotes activity >2500 nM












TABLE B








MRGPRX4




Antagonist



Compound No.
Activity (nM)



















1
+++++



2
+++++



3
+++++



4
+++++



5
+++++



6
+++++



7
+++++



8
+++++



9
+++++



10
+++++



11
+++++



12
+++++



13
+++++



14
+++++



15
+++++



16
+++++



17
+++++



18
+++++



19
+++++



20
+++++



21
+++++



22
+++++



23
+++++



24
+++++



25
+++++



26
+++++



27
+++++



28
++++



29
++++



30
++++



31
++++



32
++++



33
++++



34
++++



35
++++



36
++++



37
++++



38
++++



39
++++



40
++++



41
++++



42
++++



43
++++



44
++++



45
++++



46
++++



47
++++



48
++++



49
++++



50
++++



51
++++



52
++++



53
++++



54
++++



55
++++



56
++++



57
++++



58
++++



59
++++



60
++++



61
++++



62
++++



63
++++



64
++++



65
++++



66
++++



67
++++



68
++++



69
++++



70
+++



71
+++



72
+++



73
+++



74
+++



75
+++



76
+++



77
+++



78
+++



79
+++



80
+++



81
+++



82
+++



83
+++



84
+++



85
+++



86
+++



87
+++



88
+++



89
+++



90
+++



91
+++



92
+++



93
++



94
++



95
++



96
++



97
++



98
++



99
++



100
++



101
++



102
++



103
++



104
++



105
++



106
++



107
++



108
++



109
++



110
++



111
++



112
++



113
++



114
++



115
++



116
++



117
++



118
++



119
++



120
++



121
++



122
++



123
+



124
+



125
+



126
+



127
+



128
+



129
+



130
+



131
++++



132
++++



133
++



134
++



135
++



136
++++



137
+++++



138
+++++



139
++++



140
+++



141
+++



142
+



143
++++



144
++++



145
+++



146
+



147
+



148
++



149
+++++



150
++++



151
+++++



152
+++++



153
+++++



154
+++++



155
++++



156
++++



157
++++



158
++++



159
++++



160
++++



161
++++



162
++++



163
++++



164
++++



165
++++



166
++++



167
++++



168
++++



169
++++



170
++++



171
++++



172
++++



173
++++



174
++++



175
++++



176
++++



177
++++



178
++++



179
++++



180
++++



181
++++



182
++++



183
+++



184
+++



185
+++



186
+++



187
+++



188
+++



189
+++



190
+++



191
+++



192
+++



193
+++



194
+++



195
+++



196
+++



197
+++



198
+++



199
+++



200
++



201
++



202
++



203
++



204
++



205
++



206
++



207
++



208
++



209
++



210
++



211
++



212
++



213
++



214
++



215
++



216
++



217
++



218
++



219
++



220
++



221
++



222
++



223
++



224
+



225
+



226
+



227
+



228
+



229
+



230
+



231
+



232
+



233
+



234
+



235
+



236
+



237
++++



238
++++



239
++++



240
++++



241
++++



242
+++



243
+++



244
+++



245
+++



246
++



247
++



248
++



249
++



250
++



251
++



252
++



253
++



254
++



255
+



256
+



257
+



258
++++



259
++++



260
+++



261
+++



262
+++



263
+++



264
+++



265
+++



266
+++



267
+++



268
+++



269
++



270
++



271
++



272
++



273
++



274
++



275
++



276
++



277
++



278
++



279
++



280
++



281
++



282
++



283
++



284
++



285
++



286
++



287
++



288
++



289
++



290
++



291
++



292
++



293
++



294
++



295
++



296
+



297
+



298
+



299
+



300
+



301
+



302
+



303
+



304
+



305
+



306
+



307
+



308
+



309
++



310
+



311
++



312
++



313
+



314
++



315
++



316
++



317
+++



318
++



319
++



320
++



321
++



322
++



323
++



324
+



325
+



326
+



327
+



328
+



329
+



330
+



331
+



332
+



333
++



334
+++



335
+



336
++++



337
+



338
++++



339
+++



340
+



341
+



342
+++



343
+++



344
+



345
+



346
++



347
+



348
++



349
+++



350
++



351
+



352
++++



353
+



354
+



355
+



356
+++



357
+++++



358
+++++



359
+



360
+



361
+++++



362
++



363
+



364
++++



365
+



366
+++++



367
++++



368
++++



369
+



370
+



371
+



372
+



373
+++



374
+



375
+



376
+++



377
+



378
+



379
+



380
+



381
++



382
+++



383
++



384
+



385
+



386
+



387
+



388
+



389
+



390
+



391
++



392
++



393
+



395
+



396
+



397
++++



398
+



399
+



400
++++



401
+++



402
++++



403
++++



404
++



405
+



406
++



407
++



408
++++



409
++



410
+



411
+++++



412
++++



413
++++



414
++++



415
+++



416
+++++



418
+



419
++++



420
++



421
+++



422
+



423
+



424
+++



425
++



426
++



427
+



428
+



429
+++



430
+



431
++



432
+++++



433
+++



434
++



435
+



436
++++



437
++++



438
++++



439
++++



440
+



441
++



442
+



443
+



444
+



445
++++



446
++



447
++++



448
+



449
++++



450
+



451
++++



452
+++



453
++



454
++



455
++++



456
++++



457
+



458
+++



459
++++



460
+++



46
++



462
++



463
+++++



464
++



465
++++



466
++++



467
++++



468
++



469
++



470
+



471
+



472
+



473
++++



474
++++



475
+



476
+



477
+++++



478
+++



479
++++



480
+++



481
+++



482
+++++



483
+++



484
++



485
+++



486
+++



487
++++



488
+++



489
+++



490
+++++



491
++++



492
+



493
+++++



494
+++++



495
+++++



496
+++++



497
+++++



498
++++



499
+++



500
++



501
++++



502
+



503
+++



504
+++



505
++



506
+



507
++++



508
++++



509
++++



510
+



511
++++



512
++++



513
+



514
++++



515
+



516
+



517
++



518
++



519
+++



520
+



521
++



522
+++



523
++



524
+++++



525
+++++



526
+++++



527
+++++



528
+++++



529
+++++



530
+++++



531
+++++



532
+++++



533
+++++



534
+++++



535
+++++



536
+++++



537
+++++



538
+++++



539
+++++



540
+++++



541
+++++



542
+++++



543
++++



544
++++



545
++++



546
++++



547
+++



548
+++



549
++++



550
++++



551
+++++



552
+++++



553
+++++



554
+++++



555
+++++



556
+++++



557
++++



558
+++++



559
+++++



560
++++



561
+++++



562
+++++



563
+++++



564
+++++



565
+++++



566
++++



567
++



568
+++



569
++++



570
+



571
+++++



572
+++++



573
+++++



574
++++










The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary, to employ concepts of the various patents, applications and publications to provide yet further embodiments.


These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.


This application claims the benefit of priority to U.S. Provisional Application No. 63/079,870, filed Sep. 17, 2020, which application is hereby incorporated by reference in its entirety.

Claims
  • 1. A method of modulating a Mas-Related G-Protein Receptor (MRGPR) X4 by contacting the MRGPR X4 with an effective amount of a compound having structure (I):
  • 2. A method for treating an MRGPR X4-dependent condition by administering to a subject in need thereof an effective amount of a compound having structure (I):
  • 3. The method of claim 2, wherein the MRGPR X4-dependent condition is an itch associated condition, a pain associated condition, or an autoimmune disorder.
  • 4-8. (canceled)
  • 9. The method of claim 2, wherein the compound of structure (I) has formula (IA):
  • 10. The method of claim 2, wherein the compound of structure (I) has formula (IB):
  • 11. The method of claim 2, wherein the compound of structure (I) has formula (IC):
  • 12. The method of claim 2, wherein the compound of structure (I) has formula (ID):
  • 13. The method of claim 2, wherein the compound of structure (I) has formula (IE):
  • 14. The method of claim 2, wherein the compound of structure (I) has formula (IF):
  • 15. The method of claim 2, wherein the compound of structure (I) has formula (IG):
  • 16. The method of claim 2, wherein the compound of structure (I) has formula (IH):
  • 17. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having structure (I):
  • 18-25. (canceled)
  • 26. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Table A, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.
  • 27-35. (canceled)
  • 36. A compound having structure (I):
  • 37. The compound of claim 36, wherein the compound is selected from any one of the compounds listed in Table A, or a pharmaceutically acceptable salt, isomer, hydrate, solvate or isotope thereof.
PCT Information
Filing Document Filing Date Country Kind
PCT/US2021/050706 9/16/2021 WO
Provisional Applications (1)
Number Date Country
63079870 Sep 2020 US