WO2013166015A1 - Ror modulators and their uses - Google Patents

Ror modulators and their uses Download PDF

Info

Publication number
WO2013166015A1
WO2013166015A1 PCT/US2013/038867 US2013038867W WO2013166015A1 WO 2013166015 A1 WO2013166015 A1 WO 2013166015A1 US 2013038867 W US2013038867 W US 2013038867W WO 2013166015 A1 WO2013166015 A1 WO 2013166015A1
Authority
WO
WIPO (PCT)
Prior art keywords
halogen
alkyl
optionally substituted
substituted
compound
Prior art date
Application number
PCT/US2013/038867
Other languages
French (fr)
Inventor
Anderson Gaweco
Jefferson W. Tilley
John Walker
Samantha PALMER
James Blinn
Original Assignee
Anderson Gaweco
Tilley Jefferson W
John Walker
Palmer Samantha
James Blinn
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Anderson Gaweco, Tilley Jefferson W, John Walker, Palmer Samantha, James Blinn filed Critical Anderson Gaweco
Priority to US14/394,787 priority Critical patent/US9447069B2/en
Priority to EP13785182.0A priority patent/EP2844260A4/en
Publication of WO2013166015A1 publication Critical patent/WO2013166015A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention relates to Retinoic Acid Receptor-Related Orphan Receptor (ROR) regulated diseases and disorders. More particularly, the invention relates to ROR modulators; compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing ROR regulated diseases and disorders.
  • ROR Retinoic Acid Receptor-Related Orphan Receptor
  • RORs Retinoic Acid Receptor-Related Orphan Receptors
  • ROR subfamily consists of three major isoforms: RORa (NR1F1), ROR (NR1F2), and RORy (NR1F3), encoded by the RORA, RORB and RORC genes, respectively.
  • RORs are multidomain proteins that contain four principal domains typical of nuclear receptors: a highly variable N-terminal A/B domain, a highly conserved DNA-binding domain (DBD), a ligand binding domain (LBD) that contains the ligand-dependent activation function-2 (AF- 2), and a hinge domain between the DBD and LBD.
  • RORa RORa isoforms
  • RORyl and RORy2 [RORyt] RORy isoforms
  • RORa and RORy play an important role in the regulation of lipid/glucose homeostasis, cellular metabolism, immune function and circadian rhythms, and have been implicated in the pathogenesis of several autoimmune, inflammatory and metabolic diseases
  • ROR LBD conformational change Such change promotes the recruitment and displacement of regulatory coactivator and corepressor proteins and upon ROR DBD binding to the ROR responsive element of the target genes lead to the induction or inhibition of ROR-regulated gene transcriptional activity. Therefore, small molecule drugs that bind to the nuclear receptor LBDs such as ROR could elicit a variety of pharmacological responses, including activation (agonists), inactivation (antagonists or non-agonists), and for receptors that are constitutively active, ligands can downregulate the constitutive response (inverse agonists).
  • RORyt is the master regulator of human T Helper 17 (T H 17) cell differentiation, function and cytokine production (Ivanov et al. (2006) Cell, 126:1121-1133).
  • T H 17 human T Helper 17
  • cytokine production Ivanov et al. (2006) Cell, 126:1121-1133.
  • TR17 cells The critical role of TR17 cells in the pathogenesis of autoimmune and inflammatory diseases has been established and is conferred by the production of its signature proinflammatory cytokines IL-
  • RORyt could have a broader anti-inflammatory effect on the combined inhibition of all TR17 cytokine production and inflammatory cellular function, and in the induction and expansion of suppressive TR EG cells, important in autoimmune and inflammatory disease resolution, and may also have therapeutic potential in metabolic diseases such as diet-induced insulin resistance known to be regulated by RORy. Since both RORyl and RORyt [RORyl] protein isoforms, contain identical LBDs, small molecule RORy modulators that inhibit RORyt activity will also inhibit RORy. RORa similarly plays an important regulatory role in the pathogenesis of autoimmune and inflammatory disorders, and also in metabolic diseases.
  • RORa critically regulates lipid and glucose homeostasis and cellular metabolism that contribute to the development of metabolic diseases.
  • the therapeutic benefits of inhibiting RORa for metabolic diseases is associated with diminished inflammation and downregulated expression of RORa target genes as seen in RORa- deficient mice.
  • As ligand-dependent transcription factors it is desirable to prepare compounds that modulate RORa and/or RORy activity which can be used in the treatment of RORa- and/or RORy-regulated autoimmune, inflammatory and metabolic diseases.
  • the invention is based in part on the discovery of ROR modulators which interact with RORa and/or RORy and thereby inhibit or induce RORa and/or RORy-activity, and RORa- and/or RORy-regulated target gene and protein expression.
  • the invention is also based on compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing disorders regulated by RORa and/or RORy, comprising the administration of an effective amount of a ROR modulator.
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R-i) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • R 5 is independently H or alkyl, or both R5 can be taken together to form a carbonyl
  • Y is 5-7 membered heteroaromatic ring linked via a carbon atom and optionally substituted; n is 0, or 1; o is 0 or 1 ; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • compounds of the Formula la are described:
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R 4 ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN,
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • compounds of the Formula lb are described:
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and N(R 4 ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating an 0-3 atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R 4 ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C 1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C 1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • compounds of the Formula Id are described:
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R-i) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C 1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C 1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating an additional N, O, S atom; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • the invention is based in part on the discovery of ROR modulators, which interact with RORa and/or RORy and thereby inhibit or induce RORa and/or RORy activity, and RORa- and/or RORy-regulated target gene and protein expression.
  • the invention is also based on compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing disorders regulated by RORa and/or RORy, comprising the administration of an effective amount of a ROR modulator.
  • ROR refers to RORa and/or RORy isoforms
  • RORy refers to all isoforms encoded by the RORC gene which include RORyl and RORyt [RORy2].
  • RORa modulator refers to a chemical compound that modulates, either directly or indirectly, the activity of RORa.
  • RORa modulators include antagonists/non-agonists, inverse agonists and agonists of RORa.
  • RORy modulator refers to a chemical compound that modulates, either directly or indirectly, the activity of RORy.
  • RORy modulators include antagonists/non-agonists, inverse agonists and agonists of RORy.
  • ROR modulator includes any and all possible isomers, stereoisomers, enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, and prodrugs of the ROR modulators described herein.
  • aryl refers to cyclic, aromatic hydrocarbon groups that have 1 to 2 aromatic rings, including monocyclic or bicyclic groups such as phenyl, biphenyl or naphthyl. Where containing two aromatic rings (bicyclic, etc.), the aromatic rings of the aryl group may be joined at a single point (e.g., biphenyl), or fused (e.g., naphthyl).
  • the aryl group may be optionally substituted by one or more substituents, e.g., 1 to 5 substituents, at any point of attachment. The substituents can themselves be optionally substituted.
  • C 1 -C 3 alkyl refers to a straight or branched chain saturated hydrocarbon containing 1-3 carbon atoms.
  • Examples of a C 1 -C 3 alkyl group include, but are not limited to, methyl, ethyl, propyl and isopropyl.
  • C 1 -C 4 alkyl refers to a straight or branched chain saturated hydrocarbon containing 1-4 carbon atoms.
  • Examples of a C 1 -C 4 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, isopropyl, isobutyl, sec-butyl and tert-butyl.
  • C 1 -C 5 alkyl refers to a straight or branched chain saturated hydrocarbon containing 1-5 carbon atoms.
  • Examples of a C 1 -C 5 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, isopropyl, isobutyl, sec -butyl and tert-butyl, isopentyl and neopentyl.
  • Ci-C 6 alkyl refers to a straight or branched chain saturated hydrocarbon containing 1-6 carbon atoms.
  • Examples of a Ci-C 6 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, and neopentyl.
  • the terms "monocyclic or bicyclic aryl,” or an " monocyclic or bicyclic heteroaryl” as used herein include but are not limited to, indolyl, isoindolyl, isoindolinyl, indazolyl, benzofuranyl, benzothienyl, benzothiazolyl, benzothiazolonyl, benzoxazolyl, benzoxazolonyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, benzimidazolonyl, benzotriazolyl, imidazopyridinyl, dihydropurinonyl, pyrrolopyrimidinyl, purinyl, pyrazolopyridinyl, pyrazolopyrimidinyl, phthalimidyl, phthalimidinyl, pyrazinylpyridinyl, pyridinopyrimidinyl, pyrimidin
  • cycloalkyl refers to a cyclic hydrocarbon containing 3-6 carbon atoms.
  • examples of a cycloalkyl group include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. It is understood that any of the substitutable hydrogens on a cycloalkyl can be substituted with halogen, C 1 -C 3 alkyl, hydroxyl, alkoxy and cyano groups.
  • heterocycle refers to a cyclic hydrocarbon containing 3-6 atoms wherein at least one of the atoms is an O, N, or S wherein a monocyclic heterocycle may contain up to two double bonds.
  • heterocycles include, but are not limited to, aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, tetrahydropyran, thiane, imidazolidine, oxazolidine, thiazolidine, dioxolane, dithiolane, piperazine, oxazine, dithiane, and dioxane.
  • a "subject” is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus monkey, and the terms “subject” and “patient” are used interchangeably herein.
  • the invention also includes pharmaceutical compositions comprising an effective amount of a ROR modulator and a pharmaceutically acceptable carrier.
  • a ROR modulator provided as a pharmaceutically acceptable prodrug, hydrate, salt, such as a pharmaceutically acceptable salt, enantiomers, stereoisomers, or mixtures thereof.
  • Representative "pharmaceutically acceptable salts” include, e.g., water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4-diaminostilbene-2, 2 -disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, calcium, calcium edetate, camsylate, carbonate, chloride, citrate, clavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, la
  • an "effective amount" when used in connection with a ROR modulator is an amount effective for treating or preventing a ROR-regulated disease or disorder.
  • carrier encompasses carriers, excipients, and diluents and means a material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body.
  • treating refers to improving at least one symptom of the subject's disorder. Treating can be curing, improving, or at least partially ameliorating the disorder.
  • disorder is used in this disclosure to mean, and is used interchangeably with, the terms disease, condition, or illness, unless otherwise indicated.
  • administer refers to either directly administering a compound or pharmaceutically acceptable salt of the compound or a composition to a subject, or administering a prodrug derivative or analog of the compound or pharmaceutically acceptable salt of the compound or composition to the subject, which can form an equivalent amount of active compound within the subject's body.
  • prodrug means a compound which is convertible in vivo by metabolic means ⁇ e.g., by hydrolysis) to a ROR modulator.
  • Suitable substituents are selected from the following which include, but are not limited to, hydroxyl, halogen, perfluorinated Ci-C 6 alkyl, amine, -C ⁇ -C ⁇ 2 alkyl, -C 2 -C 12 alkene, -C 2 -C 12 alkyne, -(C 1 -C3 alkyl)-(cycloalkyl), aryl, alkyl-aryl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)-0- alkyl, -C(0)NH(alkyl), benzyl, -C(0)NH 2 , -C(0)N(alkyl) 2 , -NHC(0)H, -NHC(0)alkyl, - S0 2 (alkyl), -S0 2 NH 2 , -S0 2 NH(alkyl), -S0 2 N(alkyl) 2 , S, CN, and SCN.
  • ACTB is ⁇ -actin
  • AF-2 activation function-2
  • AIBN is azobisisobutyronitrile
  • Boc and BOC are tert-butoxycarbonyl
  • Boc 2 0 is di-tert-butyl dicarbonate
  • BSA bovine serum albumin
  • CD is cluster of differentiation
  • CDI is 1 , ⁇ -carbonyldiimidazole
  • DBD is DNA- binding domain
  • DCC is N,N'-dicyclohexylcarbodiimide
  • DIEA and DIPEA is N,N- diisopropylethylamine
  • DMAP is 4-dimethylaminopyridine
  • DMEM is Dulbecco's Modified Eagle Medium
  • DMF is N,N-dimethylformamide
  • DMSO is dimethyl sulfoxide
  • DOSS sodium dioctyl sulfosuccinate
  • EC 50 is half maximal effective concentration
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or ⁇ ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • R 5 is independently H, or alkyl, or both R5 can be taken together to form a carbonyl
  • Y is 5-7 membered heteroaromatic ring linked via a carbon atom and optionally substituted; n is 0, or 1; o is 0 or 1 ; p is 1 or 2; and wherein the symbol indicates a single or double bond. or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • Y is selected from thienyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, imidazolyl, pyrazolyl, oxadiazolyl, thiadiazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, and triazinyl.
  • Y can be optionally substituted.
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R 4 ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • compounds of the Formula lb are described:
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R 4 ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4 membered ring, optionally incorporating an 0-3 atoms; wherein the 4-7 membered ring optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • compounds of the Formula Ic are described:
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and N(R 4 ) 2 ;
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R 4 ) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R 4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R 4 ) 2 ;
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and
  • Ri is absent, H, OH, halogen, N(R 4 ) 2 , or C 1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R 4 ) 2 ;
  • R 2 and R 3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R-i) 2 , or R 2 and R 3 , taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C 1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R 4 ) 2 , or R 2 and R 3 , can be taken together to form a carbonyl;
  • Each R4 is independently H, alkyl, or aryl, or two R 4 can be taken together to form a 4-7 membered ring, optionally incorporating an additional N, O, S atom; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or
  • Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
  • suitable compounds of the invention include:
  • suitable compounds of the invention also include:
  • suitable compounds of the invention may also include:
  • the invention also include:
  • methods of inhibiting, preventing or treating a disease, or symptoms of a disease, regulated by RORa and/or RORy comprises administering to a subject in need thereof, a therapeutically-effective amount of a ROR modulator.
  • the disease regulated by RORa and/or RORy is selected from Autoimmune, Inflammatory and Metabolic Diseases, including but not limited to angina pectoris, myocardial infarction, atherosclerosis, cystic fibrosis, gastritis, autoimmune myositis, giant cell arteritis, Wegener's granulomatosis, asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, juvenile rheumatoid arthritis, allergen-induced lung inflammation, allergy, psoriasis, psoriatic arthritis, colitis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Sjogren's syndrome, dry eye, optic neuritis, neuromyelitis optica, myasthenia gravis, Guillain-Barre syndrome, Graves disease, multiple sclerosis, autoimmune uveitis, ankylosing spondylitis, organ transplant rejection, polymyalgia r
  • Also described are methods of modulating RORa and/or RORy activity as an agonist, inverse agonist or antagonist/non-agonist in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
  • Also described are methods of inducing or inhibiting RORa- and/or RORy-regulated target gene expression and protein production in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
  • Also described are methods of regulating corepressor and/or coactivator protein interaction with RORa and/or RORy LBD in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
  • Also described are methods of reducing the amount of RORa- and/or RORy-regulated production of T H 17 cytokines IL-17A, IL-17F, IL-17AF, IL-21, and/or IL-22 in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
  • RORa- and/or RORy-regulated cell proliferation or activation in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
  • the ROR modulators can each be administered in amounts that are sufficient to treat or prevent but are not limited to Autoimmune, Inflammatory and Metabolic Diseases, or prevent the development thereof in subjects.
  • the invention also includes pharmaceutical compositions useful for treating or preventing a ROR regulated disease, or for inhibiting a ROR regulated disease, or more than one of these activities.
  • the compositions can be suitable for internal use and comprise an effective amount of a ROR modulator and a pharmaceutically acceptable carrier.
  • the ROR modulators are especially useful in that they demonstrate very low systemic toxicity or no systemic toxicity.
  • Administration of the ROR modulators can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral (intravenous), intramuscular, intrathecal, intra-vitreal, transdermal, subcutaneous, vaginal, buccal, rectal, topical administration modes or as a drug- eluting stent.
  • systemic or local administration such as oral, nasal, parenteral (intravenous), intramuscular, intrathecal, intra-vitreal, transdermal, subcutaneous, vaginal, buccal, rectal, topical administration modes or as a drug- eluting stent.
  • compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • injectables tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • they can also be administered in intravenous (both bolus and infusion), intraperitoneal, intrathecal, intra-vitreal injection, subcutaneous or intramuscular form, all using forms well known to those skilled in the pharmaceutical arts.
  • Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a ROR modulator and a pharmaceutically acceptable carrier, such as: a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, corn oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets, a
  • emulsifier or dispersing agent such as Tween 80, Labrasol, HPMC, DOSS, caproyl 909, labrafac, labrafil, peceol, transcutol, capmul MCM, capmul PG-12, captex 355, gelucire, vitamin E TGPS or other acceptable emulsifier; and/or g) an agent that enhances absorption of the compound such as cyclodextrin, hydroxypropyl-cyclodextrin, PEG400, PEG200.
  • Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc.
  • the ROR modulator is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension.
  • a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like.
  • Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the ROR modulators.
  • the ROR modulators can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.
  • the pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations
  • the ROR modulators can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines.
  • a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in United States Patent No. 5,262,564, the contents of which are herein incorporated by reference in their entirety.
  • ROR modulators can also be delivered by the use of monoclonal antibodies as individual carriers to which the ROR modulators are coupled.
  • the ROR modulators can also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide -phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the ROR modulators can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • ROR modulators are not covalently bound to a polymer, e.g., a polycarboxylic acid polymer, or a polyacrylate.
  • Parenteral injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.
  • compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1 % to about 80 %, from about 5 % to about 60 %, or from about 1 % to about 20 % of the ROR modulator by weight or volume.
  • the dosage regimen utilizing the ROR modulator is selected in accordance with a variety of factors including type, species, age, weight, sex, race, diet, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular ROR modulator employed.
  • a physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • Effective dosage amounts of the present invention when used for the indicated effects, range from about 0.1 mg to about 5000 mg of the active ingredient per unit dose which could be administered.
  • the compositions are in the form of a tablet that can be scored.
  • Appropriate dosages of the ROR modulators can be determined as set forth in Goodman, L. S.; Gilman, A. The Pharmacological Basis of Therapeutics, 5th ed.; MacMillan: New York, 1975, pp. 201-226, the contents of which are hereby incorporated by reference.
  • ROR modulators can be administered in a single daily dose, or the total daily dosage can be administered in divided doses of two, three or four times daily. Furthermore, ROR modulators can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration can be continuous rather than intermittent throughout the dosage regimen. Other illustrative topical preparations include creams, ointments, lotions, aerosol sprays and gels, wherein the concentration of the ROR modulator ranges from about 0.1 % to about 15 %, w/w or w/v.
  • the ROR modulators can also each be administered in amounts that are sufficient to treat or prevent ROR-associated diseases.
  • These diseases include, but are not limited to, Autoimmune, Inflammatory and Metabolic diseases, either individually or in combination with one or more agents and or methods for treating and preventing these ROR-regulated diseases.
  • Compounds according to the present invention may be administered in combination with the following non-limiting examples of therapeutic agents and methods for treating and preventing these ROR-regulated diseases in any combination that may include, but are not limited to any of the following: glucocorticoids, nonsteroidal antiinflammatory drugs (NSAIDs) (non-limiting examples include acetominophen, aspirin, capsaicin, diclofenac, diclofenac/misoprostol, efenamic acid, etodolac, felbinac, fenoprofen, flurbiprofen, ketoprofen, ibuprofen, indomethacin, ketorolac, loxoprofen, meclofenamate, meloxicam, nabumetone, naproxen, nimesulide, oxaprozin, piroxicam, sulindac, tolmetin), cyclooxygenase (COX)-2 inhibitors (non-limiting examples
  • Alkylation can be accomplished by standard methods, such as treatment of the quinoline derivatives with NaH or K 2 CO 3 followed by a benzyl halide such as a substituted benzyl bromide.
  • a protecting group on the functionality on the benzyl group. This can be chosen such that it can be later removed, once the key steps, coupling and or alkylation are complete.
  • the use and selection of protecting groups is well documented in the organic synthesis literature. A particularly useful guide in this process is Greene 's Protective Groups in Organic Synthesis by Peter G. M. Wuts and T. W. Greene, 4 th ed., Wiley, 2007.
  • Het-W in which Het is a 5-7-membered heteroaromatic compound, which may incorporate a protecting group as appropriate, and W is a functional group capable of participating in a transition metal catalyzed cross-coupling reaction such as a Suzuki reaction.
  • Skilled organic chemists will understand how to select the particular choice of X and Y for a given desired transformation and incorporate the appropriate protection/deprotection methods, where needed. See for example: N. Kudo et al, Angew. Chem. Int. Ed., 2006, 45, 1282-1284 and Dvorak, C. A.; et al, Journal of Organic Chemistry 2005, 70, 4188-4190; Barder, T. E., et al. J. Am. Chem. Soc. 2005, 127, 4685-4696.
  • the aniline may be acylated with an acryloyl derivative followed by cyclization in the presence of a strong acid, such as H 2 SO 4 to give compounds of formula XIII or XIV.
  • a strong acid such as H 2 SO 4
  • These compounds can be converted to compound XII by a series of reductions and the products can be alkylated to give XV-XVII as described above. If protecting groups are employed, they can be inserted and removed at appropriate points.
  • an alkyl alcohol can be added ortho to the aniline nitrogen atom using technics well known in organic synthesis, for example using an organometallic coupling reaction with an alkenyl or alkynyl alcohol, followed by reduction, if necessary.
  • These alkyl alcohols can then undergo oxidatetive cyclization in the presence of Rh complexes to give compounds of formula XIII which can then be further elaborated to compounds of the invention using standard techniques. See for example Fuita, K.-i., et al, Organic Lett. 2004 6, 2785.
  • an indole derivative of formula XIX in which Q" is a leaving group suitable for use in a Suzuki reaction or other organo metallic coupling reaction may be alkylated with a substituted benzyl halide, incorporating protecting group as necessary, in the presence of a suitable base, for example NaH to give a N-benzyl derivative XXII.
  • Suzuki or other suitable coupling with a heterocyclic partner as described above then can give the heterocyclic substituted indole derivative, which can be deprotected, if necessary.
  • an indole of formula XIX may be reduced by treatment with a suitable reducing agent, for example, in the case where Q" is a halogen atom, sodium cyanoborohydride in the presence of acetic acid, to give an indoline derivative XX.
  • a suitable reducing agent for example, in the case where Q" is a halogen atom, sodium cyanoborohydride in the presence of acetic acid, to give an indoline derivative XX.
  • it may be appropriate to convert the Q" group to a different leaving group for example from a halide to a borate ester.
  • a coupling reaction with a suitably functionalized heteroaromatic species can then furnish the target compound of the invention XXIV, after deprotection, if necessary.
  • indoline systheses with an indoline derivative already incorporating a heteroaromatic ring, such as XXV.
  • an alkylation as described above for XVIII with a benzyl halide can furnish a compound of formula XXIV directly or after deprotection, as appropriate.
  • a carbonyl derivative such as XXVI, XXVII or XXVIII
  • Y H, OH, NHR 4 , or OR 5 , wherein R 4 is H, lower alkyl or OR 6 , wherein is H or lower alkyl and R5 is lower alkyl or another substituent suitable for the displacement chemistry associated with the intended heterocycle construction.
  • Such intermediates can be alkylated as above with the appropriate benzyl derivatives to give the benyzlated derivatives XXIX, XXX and XXXI, followed by elaboration of the carbonyl derivative to the desired heterocyclic derivative, XXXII, XXXIII, and XXXIV using the chemistry appropriate to the target heterocycle.
  • the sequence of steps necessary to carry out these transformations is well established in the chemistry literature. The sequence of the steps may be altered to suit the particular selection of target, available starting materials and experimental convenience.
  • a functionalized bycylic core such as IV, V, VI, XIX or XX that can be converted to a carbonyl containing compound such as XXIX, XXX, or XXXI using standard organometallic catalyzed acylation reactions.
  • reaction mixture was concentrated under reduced pressure to afford the crude product.
  • the crude product was diluted with water and extracted with dicloromethane (2 x 500 mL). The combined organic layers were washed with water, then brine, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford crude 2-(4-(bromomethyl)phenyl)-l , l ,l ,3,3,3-hexafluoropropan-2-ol.
  • Reaction step 3 Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-l ,2,3,4-tetrahydroquinoline-6-carbaldehyde, 6
  • reaction mixture was diluted with water and extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • the crude product was purified by preparative HPLC followed by preparative TLC; to afford 1,1,1 ,3 ,3 ,3 -hexafluoro-2-(4-((6-(3 -methyl- 1 H-pyrazol-5 -yl)-3 ,4-dihydroquinolin- 1 (2H)- yl)methyl)phenyl)propan-2-ol, 8 as an off white solid.
  • reaction mixture was diluted with ice cold water and extracted with ethyl acetate (2 x 100 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • the crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 10 % EtOAc/Pet ether to give tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole-l-carboxylate, 10 as a white solid.
  • reaction mixture was poured into ice cold water and extracted with ethyl acetate (2 x 200 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was washed with pet ether followed by dichloromethane to afford 6-bromo-l-(4- (1,1,1 ,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-3,4-dihydroquinolin-2(lH)-one, 14.
  • Reaction step 3 Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-2-oxo-l,2,3,4-tetrahydroquinoline-6-carbonitrile, 15
  • reaction mixture was cooled to RT, quenched with aq. ammonia and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • the crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 10 % EtOAc/Pet ether to give l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-2-oxo-l ,2,3,4-tetrahydroquinoline-6-carbonitrile, 15 that was used directly in the next step.
  • Reaction step 4 Preparation of l-(4-(l , l ,l ,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-6-(3-methyl-lH-l ,2,4-triazol-5-yl)-3,4-dihydroquinolin-2(lH)-one, 16
  • reaction mixture was cooled to RT and concentrated under reduced pressure.
  • the crude product was diluted with water and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • Reaction step 5 Preparation of l , l ,l ,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-l ,2,4- triazol-5 -yl)-3 ,4-dihydroquinolin- 1 (2H)-yl)methy l)phenyl)propan-2-ol, 17
  • reaction mixture was cooled to RT, the pH was adjusted to 10 with NaHC0 3 solution, and it was extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • the crude product was purified by preparative HPLC to afford 1,1,1 ,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-l ,2,4-triazol-5-yl)-3,4-dihydroquinolin- l(2H)-yl)methyl)phenyl)propan-2-ol, 17 as a brown semi-solid.
  • Reaction step 1 Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-2-oxo-l,2,3,4-tetrahydroquinoline-6-carbaldehyde, 18
  • reaction mixture was diluted with water and extracted with ethyl acetate (2 x 80 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • Reaction step 3 Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-6-(3-methyl-lH-pyrazol-5-yl)-3,4-dihydroquinolin-2(lH)-one, 20
  • reaction mixture was diluted with ethyl acetate and washed with ice cold water. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford crude product. A solution of hydrochloric acid in dioxane (1 mL) was added to the crude product and the mixture was stirred for 30 min. The reaction mixture was concentrated under reduced pressure. The crude product was diluted with ethyl acetate and washed with saturated sodium bicarbonate solution.
  • reaction mixture was concentrated under reduced pressure to afford the crude product.
  • the crude product was purified over silica gel (60-120 mesh) column chromatography by eluting with 50 % EtO Ac/Pet ether to give l-(4- (1,1,1 ,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-6-(3-methyl-lH-l ,2,4-triazol-5-yl)- 3,4-dihydroquinolin-2(lH)-one, 16 as an off-white solid.
  • reaction mixture was quenched with ice and extracted with ethyl acetate (2 x 300 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • the crude product was purified by column chromatorgraphy over silica gel (100- 200 mesh) eluting with 10 % EtO Ac/Pet ether to give 2-(4-((5-bromoindolin-l- yl)methyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 23 as a pale yellow liquid.
  • Reaction step 5 Preparation of l ,l , l ,3,3,3-hexafluoro-2-(4-((5-(3-methyl-lH-pyrazol- 5-yl)indolin- 1 -yl)methyl)phenyl)propan-2-ol, 26
  • reaction mixture was cooled to RT, diluted with ice cold water and extracted with ethyl acetate (2 x 80 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was triturated with ethyl acetate (60 mL) to afford 1,1,1,3,3,3- hexafluoro-2-(4-((5 -(3 -methyl- lH-pyrazol-5-yl)indolin- 1 -yl)methyl)phenyl)propan-2-ol, 26 as a pale yellow solid.
  • reaction mixture was concentrated under reduced pressure, diluted with ethyl acetate and washed with water. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. After column purification the product was isolated with 60 % purity to afford 2-(4-((5-(lH-pyrazol-4- yl)indolin-l-yl)methyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 27 as a white solid.
  • Reaction step: 1 Preparation of methyl l-(4-(l, 1,1,3, 3,3-hexafluoro-2-hydroxypropan- 2- l)benzyl)indoline-5-carboxylate, 28.
  • reaction mixture was filtered through a celite bed and the eluate extracted with ethyl acetate (2 x 150 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure.
  • reaction mixture was evaporated under reduced pressure, the residue was diluted with cold water, acidified to pH 2 with 2N HCl solution and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with water and brine solution, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to give l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carboxylic acid as an off white solid.
  • reaction mixture was diluted with water and extracted with ethyl acetate (4 x 50 mL). The organic layer was dried over sodium sulfate and evaporated under reduced pressure.
  • the crude product was purified over silica gel (100-200 mesh) column chromatography by eluting with 80% ethyl acetate/ pet ether to afford N- acetyl- l-(4-(l,l,l ,3 ,3 ,3 -hexafluoro-2-hydroxypropan-2-yl)benzyl)indoline-5 -carbohydrazide,
  • Reaction step 4 Preparation of 1,1, 1,3,3, 3-hexafluoro-2-(4-((5-(5-methyl-l, 3,4- oxadiazol-2- l)indolin- 1 -yl)methyl)phenyl)propan-2-ol, 31
  • Methyl 4-((5-bromoindolin-l-yl)methyl)benzoate can be prepared by alkylation of 5- bromoindoline with methyl (4-bromomethyl)benzoate in the presence of NaH in THF using the general procedure described in Example 7, step 1.
  • 4-((5-bromoindolin-l-yl)methyl)benzamide, 33 can be prepared by hydrolysis of the methyl ester of 32 by treatment with NaOH in EtOH until hydrolysis is complete followed by activation of the resulting acid with isobutyl chloro formate in the presence of NMP in THF at
  • 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 34 can be prepared by deoxygenation of a solution of 4-((5-bromoindolin-l-yl)methyl)benzamide, 33, tert-butyl 4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 and CS 2 CO 3 in DMF: water 2: 1 by the passage of argon for 20 min followed by the introduction of Pd(PPh 3 ) 4 and heating the mixture to 100 °C for 2 h. Treatment of the crude product with HC1 in dioxane then can provide 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 34.
  • Methyl 4-((5-bromoindolin-l-yl)methyl)benzoate can be prepared by alkylation of 5- bromoindoline with methyl (4-bromomethyl)benzoate in the presence of NaH in THF using the general procedure described in Example 7, step 1.
  • 4-((5-bromoindolin-l-yl)methyl)-N-methylbenzamide, 35 can be prepared by hydrolysis of the methyl ester of 32 by treatment with NaOH in EtOH until hydrolysis is complete followed by treating a DMF solution of resulting acid with CH 3 NH 2 .HCI and HATU with DIPEA at 0 °C and allowing the mixture to stir over night at room temperature.
  • 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 36 can be prepared by deoxygenation of a solution of 4-((5-bromoindolin-l-yl)methyl)-N-methylbenzamide, 35, tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 and CS 2 CO 3 in DMF: water 2: 1 by the passage of argon for 20 min followed by the introduction of Pd(PPh 3 ) 4 and heating the mixture to 100 °C for 2 h. Treatment of the crude product with HCl in dioxane then can provide 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)-N- methylbenzamide, 36.
  • reaction mixture was quenched with saturated ammonium chloride solution at -70 °C and diluted with water.
  • the aqueous solution was extracted with ethyl acetate (2 x 150 mL).
  • the combined organic layers were dried over anhydrous sodium sulphate and evaporated under reduced pressure.
  • the crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 5 % EtO Ac/Pet ether to give l-(/?-tolyl)cyclobutanol, 39 as a colorless liquid.
  • reaction mixture was cooled, poured into ice cold water and extracted with dichloromethane (2 x 200 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 7 % EtO Ac/Pet ether to give l-(4- (bromomethyl)phenyl)cyclobutanol, 40 as a colorless liquid.
  • Step 3 Preparation of l-(4-((5-bromoindolin-l-yl)methyl)phenyl)cyclobutanol, 41 1- -(bromomethyl)phenyl)cyclobutanol
  • l-(4-((5-bromoindolin-l-yl)methyl)phenyl)cyclobutanol, 41 can be prepared by the alkylation of 5-bromo indoline with l-(4-(bromomethyl)phenyl)cyclobutanol, 40 in the presence of NaH in THF using the procedure described in Example 7, step 1.
  • Step 4 Preparation of l-(4-((5-(lH-pyrazol-4-yl)indolin-l- l)methyl)phenyl)cyclobutanol, 42.
  • l-(4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)phenyl)cyclobutanol, 42 can be prepared by coupling of l-(4-((5-bromoindolin-l-yl)methyl)phenyl)cyclobutanol, 41 with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 followed by deprotection by treatment with HC1 in dioxane using the general procedure described in Example 8.
  • (4-(l,l,l-Trifluoro-2-methylpropan-2-yl)phenyl)methanol, 44 can be prepared from l-bromo-4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzene, 43 by palladium catalyzed carbonylation by deoxygenating a solution of l-bromo-4-( 1,1,1 -trifluoro-2-methylpropan-2- yl)benzene, 43, and an excess of Et 3 N in MeOH DMF 2: 1 by the passage of argon for 20 min contained in an autoclave vessel. Pd(PPh 3 ) 4 can be added and the mixture further deoxygenated with argon for 20 min.
  • l-Bromo-4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzene, 45 can be prepared by treatment of a solution of (4-(l,l,l-trifluoro-2-methylpropan-2-yl)phenyl)methanol, 44, in ether with PBr 3 at room temperature for 4 h, followed by quenching and aqueous workup.
  • Step 3 Preparation of 5-bromo-l-(4-(l,l,l-trifluoro-2-methylpropan-2- yl)benzyl)indoline, 46
  • 5-Bromo-l-(4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzyl)indoline, 46 can be prepared from 5-bromoindoline, 22 and l-bromo-4-( 1,1,1 -trifluoro-2-methylpropan-2- yl)benzene, 45 using the general method described in Example 7, step 1.
  • Step 4 Preparation of 5-(lH-pyrazol-4-yl)-l-(4-(l,l,l-trifluoro-2-methylpropan-2- yl)benzyl)indoline, 47.
  • 5-(lH-Pyrazol-4-yl)- 1 -(4-(l , 1 , 1 -trifluoro-2-methylpropan-2-yl)benzyl)indoline, 47 can be prepared from 5-bromo-l-(4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzyl)indoline, 46 using the general method described in Example 8.
  • 5-(lH-Pyrazol-4-yl)-l-(4-(trifluoromethyl)benzyl)indoline, 48 can be prepared by alkylation of 5-bromoindoline, 22 with l-bromomethyl-4-trifluoromethylbenzene using the conditions described in Example 7, part 1 followed by palladium catalyzed coupling of the alkylation product with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole-l-carboxylate, 10 followed by hydrolysis using the reaction conditions described in Example 8.
  • Example 15 Preparation of l-(4-isopropylbenzyl)-5-(lH-pyrazol-4-yl)indoline
  • l-(4-Isopropylbenzyl)-5-(lH-pyrazol-4-yl)indoline, 49 can be prepared by alkylation of 5-bromoindoline, 22 with l-bromomethyl-4-isopropylbenzene using the conditions described in Example 7, part 1 followed by palladium catalyzed coupling of the alkylation product with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l- carboxylate, 10 followed by hydrolysis using the reaction conditions described in Example 8.
  • Example 16 l-(4-(tef"f-butyl)benzyl)-5-(lH-pyrazol-4-yl)indoline, 50
  • l-(4-Isopropylbenzyl)-5-(lH-pyrazol-4-yl)indoline, 50 can be prepared by alkylation of 5-bromoindoline, 22 with l-(bromomethyl)-4-(fert-butyl)benzene using the conditions described in Example 7, part 1 followed by palladium catalyzed coupling of the alkylation product with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l- carboxylate, 10 followed by hydrolysis using the reaction conditions described in Example 8.
  • Example 17 Preparation of 2-(4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)phenyl)- l,l,l,3,3,3-hexafluoropropan-2-ol
  • Step 2 Preparation of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3, 3,3- hexafluoropropan-2-ol, 23.
  • reaction mixture was de -oxygenated by purging argon gas into the reaction mixture for 30 min.
  • PdCl 2 dppf
  • dichloromethane 4.5 g, 5.5 mmol
  • the progress of the reaction was monitored by TLC (TLC system: 40 % EtOAc/Pet ether, R f value: 0.01).
  • reaction mixture was filtered through a celite bed.
  • the filtrate was extracted with ethyl acetate (2 x 500 mL).
  • the combined organic layers were washed with brine and dried over anhydrous sodium sulfate.
  • the solvent was removed under reduced pressure to afford crude compound. This procedure was repeated on an additional 3 x 25 g batches and the crude products were combined for purification.
  • the compounds according to Formula 1 are RORa and/or RORy modulators, and are useful in the treatment of RORa- and/or RORy-regulated diseases.
  • the biological activities of the compounds according to Formula 1 can be determined using any suitable assay for determining the activity of a candidate compound as a RORa and/or RORy modulator, as well as in tissue and in vivo models.
  • Example 18 TH17 cell differentiation.
  • PBMCs Peripheral blood mononuclear cells
  • leukocyte enriched plasma (buffy coat) from healthy volunteers from which PBMCs were isolated by density gradient centrifugation using Ficoll-PaqueTM PLUS (GE Healthcare).
  • Naive CD4+ T cells were isolated using the Na ' ive CD4+ T cell Isolation Kit II (Miltenyi Biotec) as per manufacturer's instructions.
  • CD4+ T cells were plated at 5 x 10 4 cells/well in 96 well plates pre-coated with immobilized human (h)-aCD3 antibody (eBioscience) then differentiated to T H 17 cells using h-aCD28, h-aIFN-y, h-aIL-4, h-IL-6, h-IL-23, h-IL- ⁇ , h- TGF- ⁇ (eBioscience) and maintained in a cell culture incubator at 37°C with 5% C0 2 in TexMACS Medium (Miltenyi Biotec) supplemented with 1% Penicillin/Streptomycin (Lonza) over 5 days.
  • h-aCD3 antibody eBioscience
  • Example 19 TH17 Cytokine Inhibition as measured by ELISA.
  • Human CD4+ T cells were differentiated to T H 17 cells as described in the presence or absence of various concentrations of compounds in a final concentration of 0.1% DMSO in TexMACS medium. Supernatants were collected and stored at -20 C until they were to be assayed for IL-17A, IL-17F, IL-17AF, IL-21 and/or IL-22 levels as determined by Ready- Set-Go ELISA kits (eBioscience) as per manufacturer's instructions. Endpoint absorbance was read at 450 nm and 570 nm according to manufacturer's instructions (eBioscience) using a microplate reader (Perkin Elmer). The percentage of cytokine inhibition was calculated with reference to DMSO treated cells and the IC50S were determined by GraphPad Prism software and presented in the table below.
  • Example 21 Real Time quantitative Reverse Transcription PCR (Real-Time qRT- PCR).
  • IL-17A, IL-17F, IL-21, IL-22, G- CSF, IL-23R and FOXP3 gene expression were determined for compound treated and untreated T H 17 cell samples using Solaris qPCR Gene expression assays (Thermo Scientific) with an iCycler iQ Real-Time PCR Detection System (Bio-Rad).
  • Example 23 Co-activator and Co-repressor Protein Interaction with Human ROR and RORa LBDs.
  • TR-FRET Time-Resolved Fluorescence Resonance Energy Transfer
  • the RORy LBD and RORa LBD each have a basal level of interaction with the co-activator D22 or co-repressor NCoRl-D2 in the absence of ligand, thus it is possible to identify compounds that inhibit or induce RORy and/or RORa co-activator/co-repressor interactions, thus determining RORa, RORy, or RORa/RORy compound selectivity.
  • the assay was performed as per manufacturer's instructions and TR- FRET was assessed 2 and 20 hours post-assay initiation.
  • ⁇ - Values represent EC50 [ ⁇ ] of D22 co-activator displacement from the respective ROR

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to ROR modulators; compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing diseases associated with ROR.

Description

ROR MODULATORS AND THEIR USES PRIORITY
[0001] This application claims the priority benefit of U.S. provisional application nos. 61/640,401, filed April 30, 2012, 61/736,895, filed December 13, 2012, and 61/779,856, filed March 13, 2013. The entire disclosures of these applications are relied on for all purposes and are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION
[0002] The invention relates to Retinoic Acid Receptor-Related Orphan Receptor (ROR) regulated diseases and disorders. More particularly, the invention relates to ROR modulators; compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing ROR regulated diseases and disorders.
BACKGROUND OF THE INVENTION
[0003] There are high unmet medical needs in the few established therapies for several autoimmune, inflammatory and metabolic diseases. Despite the diverse clinical manifestations of these diseases, Retinoic Acid Receptor-Related Orphan Receptors (RORs) regulate and contribute to the pathogenesis of these diseases through modulation of immune responses and lipid/glucose homeostasis. Only recently has the critical regulatory role of RORs been well-characterized and target validated in several animal models of some of these diseases. RORs are transcription factors which belong to the nuclear hormone receptor superfamily (Jetten (2009) Nucl.Recept.Signal, 7:e003; Jetten et al. (2013) Front Endocrinol. (Lausanne), 4: 1; Jetten & Joo (2006) Adv. Dev. Biol, 16:313-355). The ROR subfamily consists of three major isoforms: RORa (NR1F1), ROR (NR1F2), and RORy (NR1F3), encoded by the RORA, RORB and RORC genes, respectively. RORs are multidomain proteins that contain four principal domains typical of nuclear receptors: a highly variable N-terminal A/B domain, a highly conserved DNA-binding domain (DBD), a ligand binding domain (LBD) that contains the ligand-dependent activation function-2 (AF- 2), and a hinge domain between the DBD and LBD. Each ROR gene through alternative splicing and promoter usage generates several ROR isoforms that differ only in their amino- terminus. In humans, there are four RORa isoforms (RORa 1-4), one ROR i isoform, and two RORy isoforms (RORyl and RORy2 [RORyt]) that are expressed in a highly tissue- specific manner. RORa and RORy play an important role in the regulation of lipid/glucose homeostasis, cellular metabolism, immune function and circadian rhythms, and have been implicated in the pathogenesis of several autoimmune, inflammatory and metabolic diseases
(Burris et al. (2012) Chem.Biol, 19:51-59; Burris et al. (2013) Pharmacol. Rev., 65:710-778;
Huh & Littman (2012) Eur. J. Immunol, 42:2232-2237; Jetten (2009) NuclRecept. Signal,
7:e003; Jetten et al. (2013) Front Endocrinol (Lausanne), 4: 1). Synthetic ligands have been described that interact with the RORa and RORy LBD functioning as a switch that induces a
ROR LBD conformational change. Such change promotes the recruitment and displacement of regulatory coactivator and corepressor proteins and upon ROR DBD binding to the ROR responsive element of the target genes lead to the induction or inhibition of ROR-regulated gene transcriptional activity. Therefore, small molecule drugs that bind to the nuclear receptor LBDs such as ROR could elicit a variety of pharmacological responses, including activation (agonists), inactivation (antagonists or non-agonists), and for receptors that are constitutively active, ligands can downregulate the constitutive response (inverse agonists).
[0004] RORyt is the master regulator of human T Helper 17 (TH17) cell differentiation, function and cytokine production (Ivanov et al. (2006) Cell, 126:1121-1133). The critical role of TR17 cells in the pathogenesis of autoimmune and inflammatory diseases has been established and is conferred by the production of its signature proinflammatory cytokines IL-
17A, IL-17F, IL-17AF, IL-21, IL-22 (Ghoreschi et al. (2010) Nature, 467:967-971; Lee et al.
(2012) Nat.Immunol, 13:991-999; Miossec et al. (2009) N.Engl.J.Med., 361 :888-898;
Miossec & Kolls (2012) Nat.Rev.Drug Discov., 11 :763-776; Zepp et al. (2011) Trends
Immunol, 32:232-239). Although several transcription factors regulate TH17, γ/δ T and innate lymphoid cells as important sources of TH17 cytokines, these cells are distinguished by its specific regulation of RORyt for cytokine transcriptional output and effector functions and to a lesser extent by RORa in humans (Cua & Tato (2010) Nat. Rev. Immunol, 10:479-489;
Huh & Littman (2012) Eur. J. Immunol, 42:2232-223"/ Ivanov et al. (2006) Cell, 126: 1121-
1133; Spits & Di Santo (2011) Nat.Immunol, 12:21-27; Sutton et al. (2012) Eur. J. Immunol,
42:2221-2231). Also, in several autoimmune disease models, there is a relative imbalance of increased TH17 cells over low numbers of immunosuppressive CD4+CD25+Foxp3+ regulatory
T cells [TReg] (Edwards et al. (2011) J.Neurol. , 258: 1518-1527; Littman & Rudensky (2010)
Cell, 140:845-858). Targeting RORyt could have a broader anti-inflammatory effect on the combined inhibition of all TR17 cytokine production and inflammatory cellular function, and in the induction and expansion of suppressive TREG cells, important in autoimmune and inflammatory disease resolution, and may also have therapeutic potential in metabolic diseases such as diet-induced insulin resistance known to be regulated by RORy. Since both RORyl and RORyt [RORyl] protein isoforms, contain identical LBDs, small molecule RORy modulators that inhibit RORyt activity will also inhibit RORy. RORa similarly plays an important regulatory role in the pathogenesis of autoimmune and inflammatory disorders, and also in metabolic diseases. RORa critically regulates lipid and glucose homeostasis and cellular metabolism that contribute to the development of metabolic diseases. The therapeutic benefits of inhibiting RORa for metabolic diseases is associated with diminished inflammation and downregulated expression of RORa target genes as seen in RORa- deficient mice. As ligand-dependent transcription factors, it is desirable to prepare compounds that modulate RORa and/or RORy activity which can be used in the treatment of RORa- and/or RORy-regulated autoimmune, inflammatory and metabolic diseases.
SUMMARY OF THE INVENTION
[0005] The invention is based in part on the discovery of ROR modulators which interact with RORa and/or RORy and thereby inhibit or induce RORa and/or RORy-activity, and RORa- and/or RORy-regulated target gene and protein expression. The invention is also based on compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing disorders regulated by RORa and/or RORy, comprising the administration of an effective amount of a ROR modulator.
[0006] In one aspect, compounds of the Formula I are described:
Figure imgf000004_0001
I wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R-i)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4) 2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
R5 is independently H or alkyl, or both R5 can be taken together to form a carbonyl;
Y is 5-7 membered heteroaromatic ring linked via a carbon atom and optionally substituted; n is 0, or 1; o is 0 or 1 ; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0007] In another aspect, compounds of the Formula la are described:
Figure imgf000005_0001
la wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl; Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0008] In another aspect, compounds of the Formula lb are described:
Figure imgf000006_0001
Ib wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating an 0-3 atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
[0009] In another aspect, compounds of the Formula Ic are described:
Figure imgf000007_0001
Ic wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C 1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C 1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0010] In another aspect, compounds of the Formula Id are described:
Figure imgf000007_0002
Id wherein A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R-i)2;
Ri is absent, H, OH, halogen, N(R4)2, or C 1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C 1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating an additional N, O, S atom; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
DETAILED DESCRIPTION OF THE INVENTION
[0011] The invention is based in part on the discovery of ROR modulators, which interact with RORa and/or RORy and thereby inhibit or induce RORa and/or RORy activity, and RORa- and/or RORy-regulated target gene and protein expression. The invention is also based on compositions comprising an effective amount of a ROR modulator; and methods for treating or preventing disorders regulated by RORa and/or RORy, comprising the administration of an effective amount of a ROR modulator.
[0012] The details of the invention are set forth in the accompanying description below. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, illustrative methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications cited in this specification are incorporated herein by reference in their entireties.
DEFINITIONS
[0013] The following definitions are used in connection with the ROR modulators:
[0014] "ROR" refers to RORa and/or RORy isoforms
[0015] "RORa" refers to all isoforms encoded by the RORA gene
[0016] "RORy" refers to all isoforms encoded by the RORC gene which include RORyl and RORyt [RORy2].
[0017] "RORa modulator" refers to a chemical compound that modulates, either directly or indirectly, the activity of RORa. RORa modulators include antagonists/non-agonists, inverse agonists and agonists of RORa.
[0018] "RORy modulator" refers to a chemical compound that modulates, either directly or indirectly, the activity of RORy. RORy modulators include antagonists/non-agonists, inverse agonists and agonists of RORy.
[0019] The term "ROR modulator" includes any and all possible isomers, stereoisomers, enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, and prodrugs of the ROR modulators described herein.
[0020] The articles "a" and "an" are used in this disclosure to refer to one or more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
[0021] The term "and/or" is used in this disclosure to mean either "and" or "or" unless indicated otherwise.
[0022] Unless otherwise specifically defined, the term "aryl" refers to cyclic, aromatic hydrocarbon groups that have 1 to 2 aromatic rings, including monocyclic or bicyclic groups such as phenyl, biphenyl or naphthyl. Where containing two aromatic rings (bicyclic, etc.), the aromatic rings of the aryl group may be joined at a single point (e.g., biphenyl), or fused (e.g., naphthyl). The aryl group may be optionally substituted by one or more substituents, e.g., 1 to 5 substituents, at any point of attachment. The substituents can themselves be optionally substituted. [0023] "C1-C3 alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-3 carbon atoms. Examples of a C1-C3 alkyl group include, but are not limited to, methyl, ethyl, propyl and isopropyl.
[0024] "C1-C4 alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-4 carbon atoms. Examples of a C1-C4 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, isopropyl, isobutyl, sec-butyl and tert-butyl.
[0025] "C1-C5 alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-5 carbon atoms. Examples of a C1-C5 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, isopropyl, isobutyl, sec -butyl and tert-butyl, isopentyl and neopentyl.
[0026] "Ci-C6 alkyl" refers to a straight or branched chain saturated hydrocarbon containing 1-6 carbon atoms. Examples of a Ci-C6 alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, and neopentyl.
[0027] The terms "monocyclic or bicyclic aryl," or an " monocyclic or bicyclic heteroaryl" as used herein include but are not limited to, indolyl, isoindolyl, isoindolinyl, indazolyl, benzofuranyl, benzothienyl, benzothiazolyl, benzothiazolonyl, benzoxazolyl, benzoxazolonyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, benzimidazolonyl, benzotriazolyl, imidazopyridinyl, dihydropurinonyl, pyrrolopyrimidinyl, purinyl, pyrazolopyridinyl, pyrazolopyrimidinyl, phthalimidyl, phthalimidinyl, pyrazinylpyridinyl, pyridinopyrimidinyl, pyrimidinopyrimidinyl, cinnolinyl, quinoxalinyl, quinazolinyl, quinolinyl, isoquinolinyl, quinolonyl, isoquinolonyl, phthalazinyl, benzodioxyl, indolinyl, dihydroquinolinyl, tetrahydroquinolinyl, dihydroisoquinolyl, tetrahydroisoquinolinyl, benzoazepinyl, benzodiazepinyl, benzoxapinyl, benzoxazepinyl, phenyl, naphthyl, pyrrolyl, furyl, thienyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, imidazolyl, pyrazolyl, oxadiazolyl, thiadiazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, and triazinyl.
[0028] The term "cycloalkyl" refers to a cyclic hydrocarbon containing 3-6 carbon atoms. Examples of a cycloalkyl group include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. It is understood that any of the substitutable hydrogens on a cycloalkyl can be substituted with halogen, C1-C3 alkyl, hydroxyl, alkoxy and cyano groups.
[0029] The term "heterocycle" as used herein refers to a cyclic hydrocarbon containing 3-6 atoms wherein at least one of the atoms is an O, N, or S wherein a monocyclic heterocycle may contain up to two double bonds. Examples of heterocycles include, but are not limited to, aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, piperidine, tetrahydropyran, thiane, imidazolidine, oxazolidine, thiazolidine, dioxolane, dithiolane, piperazine, oxazine, dithiane, and dioxane.
[0030] A "subject" is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus monkey, and the terms "subject" and "patient" are used interchangeably herein.
[0031] The invention also includes pharmaceutical compositions comprising an effective amount of a ROR modulator and a pharmaceutically acceptable carrier. The invention includes a ROR modulator provided as a pharmaceutically acceptable prodrug, hydrate, salt, such as a pharmaceutically acceptable salt, enantiomers, stereoisomers, or mixtures thereof.
[0032] Representative "pharmaceutically acceptable salts" include, e.g., water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4-diaminostilbene-2, 2 -disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitartrate, borate, bromide, butyrate, calcium, calcium edetate, camsylate, carbonate, chloride, citrate, clavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, magnesium, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, 3-hydroxy-2- naphthoate, oleate, oxalate, palmitate, pamoate (l,l-methene-bis-2-hydroxy-3 -naphthoate, einbonate), pantothenate, phosphate/diphosphate, picrate, polygalacturonate, propionate, p-toluenesulfonate, salicylate, stearate, subacetate, succinate, sulfate, subsalicylate, suramate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate salts.
[0033] An "effective amount" when used in connection with a ROR modulator is an amount effective for treating or preventing a ROR-regulated disease or disorder.
[0034] The term "carrier", as used in this disclosure, encompasses carriers, excipients, and diluents and means a material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body.
[0035] The term "treating", with regard to a subject, refers to improving at least one symptom of the subject's disorder. Treating can be curing, improving, or at least partially ameliorating the disorder. [0036] The term "disorder" is used in this disclosure to mean, and is used interchangeably with, the terms disease, condition, or illness, unless otherwise indicated.
[0037] The term "administer", "administering", or "administration" as used in this disclosure refers to either directly administering a compound or pharmaceutically acceptable salt of the compound or a composition to a subject, or administering a prodrug derivative or analog of the compound or pharmaceutically acceptable salt of the compound or composition to the subject, which can form an equivalent amount of active compound within the subject's body.
[0038] The term "prodrug," as used in this disclosure, means a compound which is convertible in vivo by metabolic means {e.g., by hydrolysis) to a ROR modulator.
[0039] The term "optionally substituted," as used in this disclosure, means a suitable substituent can replace a hydrogen bound to a carbon, nitrogen, or oxygen. When a substituent is oxo {i.e., = O) then 2 hydrogens on the atom are replaced by a single O. Suitable substituents are selected from the following which include, but are not limited to, hydroxyl, halogen, perfluorinated Ci-C6 alkyl, amine, -C\-C\2 alkyl, -C2-C12 alkene, -C2-C12 alkyne, -(C1-C3 alkyl)-(cycloalkyl), aryl, alkyl-aryl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)-0- alkyl, -C(0)NH(alkyl), benzyl, -C(0)NH2, -C(0)N(alkyl)2, -NHC(0)H, -NHC(0)alkyl, - S02(alkyl), -S02NH2, -S02NH(alkyl), -S02N(alkyl)2, S, CN, and SCN. It will be understood by those skilled in the art, with respect to any group containing one or more substituents, that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical, synthetically non-feasible and/or inherently unstable. Furthermore, combinations of substituents and/or variables within any of the Formulae represented herein are permissible only if such combinations result in stable compounds or useful synthetic intermediates wherein stable implies a reasonable pharmologically relevant half-life at physiological conditions.
[0040] The following abbreviations are used herein and have the indicated definitions: ACTB is β-actin, AF-2 is activation function-2, AIBN is azobisisobutyronitrile, Boc and BOC are tert-butoxycarbonyl, Boc20 is di-tert-butyl dicarbonate, BSA is bovine serum albumin, CD is cluster of differentiation, CDI is 1 , Γ-carbonyldiimidazole, DBD is DNA- binding domain, DCC is N,N'-dicyclohexylcarbodiimide, DIEA and DIPEA is N,N- diisopropylethylamine, DMAP is 4-dimethylaminopyridine, DMEM is Dulbecco's Modified Eagle Medium, DMF is N,N-dimethylformamide, DMSO is dimethyl sulfoxide, DOSS is sodium dioctyl sulfosuccinate, EC50 is half maximal effective concentration, EDC and EDCI are l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride, ELISA is enzyme-linked immunosorbent assay, EtOAc is ethyl acetate, FBS is fetal bovine serum, FOXP3 is forkhead box P3, G-CSF is granulocyte colony-stimulating factor, h is hour, HATU is 2-(7-aza-lH- benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate, HIV is human immunodeficiency virus, HOBt is 1-Hydroxybenzotriazole, HPMC is hydroxypropyl methylcellulose, HPRT1 is hypoxanthine phosphoribosyltransferase 1, IC50 is half maximal inhibitory concentration, IFN-γ is interferon gamma, IL is interleukin, IL-23R is interleukin 23 receptor, LAH is lithium aluminum hydride, LBD is ligand binding domain, MIQE is minimum information for publication of quantitative real-time PCR experiments, MTBE is methyl tert-butyl ether, NBS is N-bromosuccinnide, NMP is N-methyl-2-pyrrolidone, oxone is potassium peroxymonosulfate, PBMCs is peripheral blood mononuclear cells, PCR is polymerase chain reaction, Pd/C is palladium on carbon, PGK1 is phosphoglycerate kinase, PPIA is peptidylprolyl isomerase A, REST is Relative Expression Software Tool, RORa is retinoic acid receptor-related orphan receptor alpha, RORy is retinoic acid receptor-related orphan receptor gamma, TBAB is tetrabutylammonium bromide, TBP is terminal binding protein, TFA is trifluoroacetic acid, TFRC is transferrin receptor, TGF-βΙ is transforming growth factor beta 1, TH17 is T helper 17 cell, TGPS is tocopherol propylene glycol succinate, THF is tetrohydrofuran, TLC is thin layer chromatography, TR-FRET is time- resolved fluorescence resonance energy transfer and μΜ is micromolar.
COMPOUNDS
[0041] In one aspect, compounds of the Formula I are described:
Figure imgf000013_0001
I wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or Ν^)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2; R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R )2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
R5 is independently H, or alkyl, or both R5 can be taken together to form a carbonyl;
Y is 5-7 membered heteroaromatic ring linked via a carbon atom and optionally substituted; n is 0, or 1; o is 0 or 1 ; p is 1 or 2; and wherein the symbol indicates a single or double bond. or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
[0042] In some embodiments, Y is selected from thienyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, imidazolyl, pyrazolyl, oxadiazolyl, thiadiazolyl, triazolyl, tetrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, and triazinyl. In some embodiments, Y can be optionally substituted.
[0043] In another aspect, compounds of the Formula la are described:
Figure imgf000014_0001
la wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R4)2; Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0044] In another aspect, compounds of the Formula lb are described:
Figure imgf000015_0001
lb wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl; Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4 membered ring, optionally incorporating an 0-3 atoms; wherein the 4-7 membered ring optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0045] In another aspect, compounds of the Formula Ic are described:
Figure imgf000016_0001
Ic wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0046] In another aspect, compounds of the Formula Id are described:
Figure imgf000017_0001
Id wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and
N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C 1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R-i)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C 1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating an additional N, O, S atom; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or
N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof. [0047] In one embodiment, suitable compounds of the invention include:
Figure imgf000018_0001
-17-
Figure imgf000019_0001
[0048] In another embodiment, suitable compounds of the invention also include:
Figure imgf000021_0001
[0049] In yet another embodiment, suitable compounds of the invention may also include:
Figure imgf000022_0001
[0050] In other embodiments, the invention also include:
Figure imgf000023_0001
-22- [0051] In another aspect, methods of inhibiting, preventing or treating a disease, or symptoms of a disease, regulated by RORa and/or RORy, is provided, which comprises administering to a subject in need thereof, a therapeutically-effective amount of a ROR modulator. In some embodiments, the disease regulated by RORa and/or RORy is selected from Autoimmune, Inflammatory and Metabolic Diseases, including but not limited to angina pectoris, myocardial infarction, atherosclerosis, cystic fibrosis, gastritis, autoimmune myositis, giant cell arteritis, Wegener's granulomatosis, asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, juvenile rheumatoid arthritis, allergen-induced lung inflammation, allergy, psoriasis, psoriatic arthritis, colitis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Sjogren's syndrome, dry eye, optic neuritis, neuromyelitis optica, myasthenia gravis, Guillain-Barre syndrome, Graves disease, multiple sclerosis, autoimmune uveitis, ankylosing spondylitis, organ transplant rejection, polymyalgia rheumatic, systemic lupus erythematosus, cutaneous lupus, lupus nephritis, glomerulonephritis, diabetes mellitus type 1 , pulmonary inflammation, macular degeneration, obesity, non-alcoholic fatty liver disease, steatohepatitis, insulin resistance, diabetes mellitus type 2, glucose intolerance, and metabolic syndrome; and Other Diseases, including but not limited to multiple myeloma, bone disease associated with multiple myeloma, gastric cancer and colon cancer.
[0052] Also described are methods of modulating RORa and/or RORy activity as an agonist, inverse agonist or antagonist/non-agonist in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
[0053] Also described are methods of inducing or inhibiting RORa- and/or RORy-regulated target gene expression and protein production in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
[0054] Also described are methods of regulating corepressor and/or coactivator protein interaction with RORa and/or RORy LBD in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
[0055] Also described are methods of reducing the amount of RORa- and/or RORy-regulated production of TH17 cytokines IL-17A, IL-17F, IL-17AF, IL-21, and/or IL-22 in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
[0056] Also described are methods of inducing or inhibiting, either directly or indirectly,
RORa- and/or RORy-regulated cell proliferation or activation in a subject which comprises administering to a subject in need thereof a pharmaceutically effective amount of a ROR modulator.
[0057] The ROR modulators can each be administered in amounts that are sufficient to treat or prevent but are not limited to Autoimmune, Inflammatory and Metabolic Diseases, or prevent the development thereof in subjects.
[0058] The invention also includes pharmaceutical compositions useful for treating or preventing a ROR regulated disease, or for inhibiting a ROR regulated disease, or more than one of these activities. The compositions can be suitable for internal use and comprise an effective amount of a ROR modulator and a pharmaceutically acceptable carrier. The ROR modulators are especially useful in that they demonstrate very low systemic toxicity or no systemic toxicity.
[0059] Administration of the ROR modulators can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral (intravenous), intramuscular, intrathecal, intra-vitreal, transdermal, subcutaneous, vaginal, buccal, rectal, topical administration modes or as a drug- eluting stent.
[0060] Depending on the intended mode of administration, the compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices. Likewise, they can also be administered in intravenous (both bolus and infusion), intraperitoneal, intrathecal, intra-vitreal injection, subcutaneous or intramuscular form, all using forms well known to those skilled in the pharmaceutical arts.
[0061] Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a ROR modulator and a pharmaceutically acceptable carrier, such as: a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, corn oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and/or polyethylene glycol; for tablets also; c) a binder, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, magnesium carbonate, natural sugars such as glucose or beta- lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, waxes and/or polyvinylpyrrolidone, if desired; d) a disintegrant, e.g. , starches, agar, methyl cellulose, bentonite, xanthan gum, alginic acid or its sodium salt, or effervescent mixtures; e) absorbent, colorant, flavorant and sweetener; f) an emulsifier or dispersing agent, such as Tween 80, Labrasol, HPMC, DOSS, caproyl 909, labrafac, labrafil, peceol, transcutol, capmul MCM, capmul PG-12, captex 355, gelucire, vitamin E TGPS or other acceptable emulsifier; and/or g) an agent that enhances absorption of the compound such as cyclodextrin, hydroxypropyl-cyclodextrin, PEG400, PEG200.
[0062] Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc. For example, the ROR modulator is dissolved in or mixed with a pharmaceutically acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension. Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the ROR modulators.
[0063] The ROR modulators can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.
[0064] In further embodiments, the pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations
[0065] The ROR modulators can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines. In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in United States Patent No. 5,262,564, the contents of which are herein incorporated by reference in their entirety.
[0066] ROR modulators can also be delivered by the use of monoclonal antibodies as individual carriers to which the ROR modulators are coupled. The ROR modulators can also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide -phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the ROR modulators can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels. In one embodiment, ROR modulators are not covalently bound to a polymer, e.g., a polycarboxylic acid polymer, or a polyacrylate.
[0067] Parenteral injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.
[0068] Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1 % to about 80 %, from about 5 % to about 60 %, or from about 1 % to about 20 % of the ROR modulator by weight or volume.
[0069] The dosage regimen utilizing the ROR modulator is selected in accordance with a variety of factors including type, species, age, weight, sex, race, diet, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the particular ROR modulator employed. A physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
[0070] Effective dosage amounts of the present invention, when used for the indicated effects, range from about 0.1 mg to about 5000 mg of the active ingredient per unit dose which could be administered. In one embodiment, the compositions are in the form of a tablet that can be scored. Appropriate dosages of the ROR modulators can be determined as set forth in Goodman, L. S.; Gilman, A. The Pharmacological Basis of Therapeutics, 5th ed.; MacMillan: New York, 1975, pp. 201-226, the contents of which are hereby incorporated by reference.
[0071] ROR modulators can be administered in a single daily dose, or the total daily dosage can be administered in divided doses of two, three or four times daily. Furthermore, ROR modulators can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration can be continuous rather than intermittent throughout the dosage regimen. Other illustrative topical preparations include creams, ointments, lotions, aerosol sprays and gels, wherein the concentration of the ROR modulator ranges from about 0.1 % to about 15 %, w/w or w/v.
[0072] The ROR modulators can also each be administered in amounts that are sufficient to treat or prevent ROR-associated diseases. These diseases include, but are not limited to, Autoimmune, Inflammatory and Metabolic diseases, either individually or in combination with one or more agents and or methods for treating and preventing these ROR-regulated diseases.
[0073] Compounds according to the present invention may be administered in combination with the following non-limiting examples of therapeutic agents and methods for treating and preventing these ROR-regulated diseases in any combination that may include, but are not limited to any of the following: glucocorticoids, nonsteroidal antiinflammatory drugs (NSAIDs) (non-limiting examples include acetominophen, aspirin, capsaicin, diclofenac, diclofenac/misoprostol, efenamic acid, etodolac, felbinac, fenoprofen, flurbiprofen, ketoprofen, ibuprofen, indomethacin, ketorolac, loxoprofen, meclofenamate, meloxicam, nabumetone, naproxen, nimesulide, oxaprozin, piroxicam, sulindac, tolmetin), cyclooxygenase (COX)-2 inhibitors (non-limiting examples include celecoxib, valdecoxib, etoricoxib, lumiracoxib, parecoxib), licofelone (ML3000), disease-modifying antirheumatic drugs (DMARDs), methotrexate, chloroquine, hydroxychloroquine, cyclophosphamide (Cytoxan), inosine monophosphate dehydrogenase (IMPDH) inhibitors (a non-limiting example is mycophenolate mofetil [Cellcept, Myfortic]), sirolimus, everolimus (rapamycin), purine nucleoside phosphorylase inhibitors, de novo purine synthesis inhibitors (non-limiting examples include polygentamate derivatives of methotrexate, antifolate compounds), dihydroorotate dehydrogenase inhibitors (malononitrilamides), prostaglandins PGE2 inhibitors, P2X7 receptor inhibitors, proteinase-activated receptor 2 (PAR-2) inhibitors, inhibitors of activated Complement (non-limiting examples include Eculizumab, Pexelizumab), complement C3/C5 convertase inhibitors (a non-limiting example is Nafamostat mesilate), active convertase inhibitors, complement C5aR antagonists, EP4 agonists, prostaglandin-12 analogs (non-limiting examples include iloprost, cicaprost, treprostinil), Sulphasalazine (SASP), 5 -aminosalicylic acid (5 -ASA), immunomodulator drugs (non-limiting examples include azathioprine (AZA), 6-mercaptopurine (6-MP), methotrexate (MTX)), calcineurin inhibitors (non-limiting examples include cyclosporine, voclosporine, tacrolimus), interleukin-10 (AGO 11), placenta-derived cells (PDA001), mucosal addressin cell adhesion molecule (MAdCAM) inhibitors (PF-00547659), GLP-2 agonists (non-limiting examples include ZP1848, ALX-0600), anti-CD3, CCR9 inhibitors, lenalidomide (Revlimid), recombinant human interleukin-11, CXCR2 Antagonists (a non- limiting example is SB-656933), glucagon-like peptide -2 (GLP-2) analogue (Teduglutide), insulin- like growth factor- 1 (IGF-1) (Increlex), synthetic guanylhydrazone semapimod (CPSI-2364), intracellular adhesion molecule-1 (ICAM-1) inhibitor (alicaforsen), stem cell therapeutics (a non-limiting example is Prochymal), activated protein C (aPC), vitamin D analogs (a non-limiting example is calcipotriene), retinoids (a non-limiting example is tazarotene), phototherapy (non-limiting examples include broadband ultraviolet B light, narrow band ultraviolet B light, psoralen plus ultraviolet A light), methotrexate, cyclosporine, acitretin, CCR6 inhibitors, CCL20 inhibitors, deoxyspergualin, alkylate deoxyribonucleic acid (DNA) agents, tumor necrosis factor (TNF)-alpha inhibitors (non-limiting examples include etanercept, infliximab, adalimumab, certolizumab pegol (Cimzia), golimumab (CNTO-148), inhibitors of TNF-alpha converting enzyme, Janus kinase (JAK) 1, 2 and/or 3 inhibitors (non-limiting examples include Tofacitinib (Xeljanz), INCB-28050, Ruxolitinib), spleen tyrosine kinase (SYK) inhibitors (a non-limiting example is R-788), caspase inhibitor, chemokine receptor antagonists, protein kinase C (pkc) inhibitors (a non-limiting example is Enzastaurin), p38 mitogen-activated protein kinase (MAPK) inhibitors, caspase inhibitors, NF-KB modulators, B cell inhibitors, Hydroxychloroquine, B-lymphocyte stimulator (BLyS) inhibitors (a non-limiting example is belimumab (Benlysta)), membrane -bound and soluble B-cell activating factor inhibitors (a non-limiting example is LY2127399), inhibitors that antagonize the binding of BLyS and APRIL (a proliferation-inducing ligand) cytokines to B cells in order to prevent B-cell maturation and autoantibody production (a non-limiting example is Atacicept), anti-CD 19, CD20 inhibitors (non- limiting examples include Rituximab, Ocrelizumab, Ofatumumab), CD22 inhibitors (a non-limiting example is Epratuzumab), T cell inhibitors (non-limiting examples include Alefacept (Amevive), IPP- 201101), interferon inhibitors (non- limiting examples include MEDI-545, rontalizumab, fontalizumab), toll-like receptor inhibitors, prasterone, estrogen receptor antagonist (fulvestrant), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4)-Ig (a non-limiting example is Abatacept (Orencia)), v-set domain containing T cell activation inhibitor 1 (VTCN1; B7-H4) agonists (AMP- 110), interleukin-1 receptor antagonists (AMG 108, Anakinra [Kineret]), interleukin-1 beta antagonists (non- limiting examples include canakinumab, Xoma 052), soluble IL-1 receptors (a non- limiting example is rilonacept), interleukin-2R antagonists (non-limiting examples include basiliximab (Simulect), daclizumab (Zenapax), interleukin-6 receptor antagonists (non-limiting examples include Tocilizumab [Actemra]), calcipotriene/betamethasone (Taclonex), fumarate (Panaclar/BG- 12), interleukin-15 inhibitors, interleukin-17 and interleukin-17 receptor inhibitors (non- limiting examples include Secukinumab, Brodalumab, Ixekizumab, RG7624), DHODH inhibitors (a non- limiting example is Vidofludimus), interleukin-18 inhibitors, T helper (Th) 17 cell inhibitors, interleukin 12/interleukin 23 inhibitors (non- limiting examples include Ustekinumab [CNTO-1275], briakinumab [ABT-874]), interleukin-22 inhibitors, interleukin- 23 inhibitors (a non- limiting example is ustekinumab), interleukin- 12 inhibitors (a non- limiting example is ustekinumab), alpha interferons, beta interferons [Interferon beta- la (Avonex, Rebif), Interferon beta- lb (Betaseron/Betaferon), Glatiramer acetate (Copaxone), selective adhesion molecule inhibitors, integrin antagonists (Natalizumab [Tysabri], vedolizumab), sphingosine 1 -phosphate receptor (S1P-R) agonists (a non- limiting example is Fingolimod [Gilenya]), fumarate derivative immunomodulators (a non-limiting example is BG-12 [Tecfidera]), laquinimod, anti-LFA-1 (a non-limiting example is Efalizumab [Raptiva]), MBP-8298, cladribin (a non-limiting example is Mylinax), Novantrone, isoxanol dihydroorotate dehydrogenase (DHODH) and tyrosine kinase inhibitor (a non-limiting example is teriflunomide [HMR-1726]), Revimmune (cyclophosphamide), Fampridine SR (4-aminopyridine), Panaclar (dimethylfumarate), MBP8298 (dirucotide, synthetic peptide version of a portion of human myelin basic protein), Campath (alemtuzumab), anti-CD52, Cladribine, purine analogs, Fingolimod (sphingosine 1 -phosphate receptor agonists), Laquinimod, Teriflunomide, de novo pyrimidine synthesis inhibitors (non-limiting examples include brequinar, leflunomide [Arava]), active metabolites of leflunomide, photodynamic therapy [PDT] with verteporfin, Anti-angiogenic factors non-limiting examples include vascular endothelial growth factor A (VEGFA) inhibitors (non-limiting examples include pegaptanib sodium, ranibizumab, bevacizumab), CCR3 inhibitors, anti-CD48, beta 2- agonists, leukotriene modifiers, phosphodiesterase (PDE) inhibitors (non-limiting examples include tetomilast, ibudilast), selective phosphodiesterase-4 (PDE-4) inhibitors (non-limiting examples include rolipram, roflumilast, piclamilast, pentoxifylline), inhibitors targeting IgE (Omalizumab), Th2 cytokine inhibitors (non-limiting examples include suplatast tosilate, sIL- 4R, IL-5 inhibitors), Macrolides, Ketolide, adenosine A2B antagonists, kappa B kinase 2 inhibitors, prostanoid and F2-isoprostane antagonists, Nitric oxide donors, inducible nitric oxide synthase inhibitors, toll-like receptor modulators, Lorcaserin, phentermine, topiramate, bupropion, naltrexone, Anti-CD3, Antithymocyte globulin, serine protease inhibitors (a non- limiting example is alpha- 1 antitrypsin AAT), tyrosine kinase inhibitors, platelet-derived growth factor receptor (PDGFR) inhibitors, insulin, Antigen-Specific Tolerance inducting agents to Type 1 Diabetes (non-limiting examples include Glutamate Decarboxylase 65 and Heat Shock Protein treatments), cannabinoid receptor 1 (CB1) antagonists, long-acting glucagon- like peptide 1 (GLP1) analogues, dipeptidyl peptidase 4 (DPP4) inhibitors, vasoactive intestinal peptide-pituitary adenylate cyclase-activating polypeptide receptor 2 (VPAC2) agonists, Glucokinase activators, Glucagon receptor antagonists, Cytosolic phosphoenolpyruvate carboxykinase (PEPCK) inhibitors, sodium-glucose co-transporter 2 (SGLT2) inhibitors, salsalate, ΙκΒ kinase-β (IK P)-inhibitors, nuclear factor kappa B inhibitors, interleukin-1 (IL-1) receptor antagonists, IL-1 beta-specific antibody, sirtuin 1 (SIRT1) activators, selective peroxisome proliferator-activated receptor (PPAR) modulators (SPPARMs), Πβ-hydroxysteroid dehydrogenase type 1 (l ipHSDl) inhibitors, PPARy ligands (non-limiting examples include rosiglitazone, pioglitazone, troglitazone), thiazolidinediones, glitazones, Warfarin, Coumadin, pradaxa (non-limiting examples include dabigatran etexilate mesylate), anti-thrombotics, Statins, hydroxy-methylglutaryl-coenzyme A reductase (HMG-CoA reductase) inhibitors, ezetimibe, fenofibrates, niacin, amlodipine, Vascular cell-adhesion molecule (VCAM) antagonists, Thromboxane A2 antagonists, prostaglandin D2 receptor 1 antagonists, G-protein-coupled receptor (GPCR) modulators, cannabinoid receptor 1 (also known as CNR1) CB1 receptor antagonists (Rimonabant), cholesteryl ester transfer protein (CETP) inhibitors (JTT-705), chemokine (C-C motif) receptor 2 (CCR2) antagonists, Phospholipase A2 inhibitors, peroxisome proliferator- activated receptor (PPAR) agonists, leucovorin [folinic acid], 5-FU, oxaliplatin, irinotecan, capecitabine, oxaliplatin, bevacizumab, cetuximab, Panitumumab, doxorubicin, mitomycin C (FAM) and combinations thereof, among others.
METHODS OF MAKING
Methods for making the RORa, RORyand RORa/RORy modulators
[0074] Examples of synthetic pathways useful for making ROR modulators of the present invention are set forth in the Examples below and generalized herein. [0075] The starting materials I to III shown in scheme 1 wherein Q is a group capable of participating in a transition metal catalyzed cross coupling reaction, such as a chloride, bromide, iodide or triflate, are commercially available or readily prepared from commercially available compounds. In some cases, it may be desirable to convert Q to a boronic acid derivative prior to the coupling reaction with Het-W.
[0076] Alkylation can be accomplished by standard methods, such as treatment of the quinoline derivatives with NaH or K2CO3 followed by a benzyl halide such as a substituted benzyl bromide. In some cases, it may be desirable to incorporate a protecting group on the functionality on the benzyl group. This can be chosen such that it can be later removed, once the key steps, coupling and or alkylation are complete. The use and selection of protecting groups is well documented in the organic synthesis literature. A particularly useful guide in this process is Greene 's Protective Groups in Organic Synthesis by Peter G. M. Wuts and T. W. Greene, 4th ed., Wiley, 2007.
[0077] The resulting benzylamines may then be coupled to the heteroaromatic derivatives Het-W in which Het is a 5-7-membered heteroaromatic compound, which may incorporate a protecting group as appropriate, and W is a functional group capable of participating in a transition metal catalyzed cross-coupling reaction such as a Suzuki reaction. Skilled organic chemists will understand how to select the particular choice of X and Y for a given desired transformation and incorporate the appropriate protection/deprotection methods, where needed. See for example: N. Kudo et al, Angew. Chem. Int. Ed., 2006, 45, 1282-1284 and Dvorak, C. A.; et al, Journal of Organic Chemistry 2005, 70, 4188-4190; Barder, T. E., et al. J. Am. Chem. Soc. 2005, 127, 4685-4696.
[0078] Carrying out the coupling reaction will then lead to the target compounds VII to IX, after protecting group removal, if necessary.
Scheme 1
Figure imgf000033_0001
Figure imgf000033_0002
Figure imgf000033_0003
[0079] A process for the preparation of heteroaryl derivatives for which the starting materials not are commercially available or readily synthesized may also be prepared starting with the 4-substituted anilines X and XI, wherein Q' = a heteroaromatic derivative as illustrated in Scheme 2.
[0080] Where Q' is stable to acidic conditions, the aniline may be acylated with an acryloyl derivative followed by cyclization in the presence of a strong acid, such as H2SO4 to give compounds of formula XIII or XIV. These compounds can be converted to compound XII by a series of reductions and the products can be alkylated to give XV-XVII as described above. If protecting groups are employed, they can be inserted and removed at appropriate points.
[0081] Alternatively, an alkyl alcohol can be added ortho to the aniline nitrogen atom using technics well known in organic synthesis, for example using an organometallic coupling reaction with an alkenyl or alkynyl alcohol, followed by reduction, if necessary. These alkyl alcohols can then undergo oxidatetive cyclization in the presence of Rh complexes to give compounds of formula XIII which can then be further elaborated to compounds of the invention using standard techniques. See for example Fuita, K.-i., et al, Organic Lett. 2004 6, 2785.
Figure imgf000035_0001
Figure imgf000035_0002
[0082] Procedures suitable for indole and indoline derivatives are shown in Scheme 3. An indole derivative of formula XVIII in which Q' = a heteroaromatic species, suitably protected, is alkylated with a benzyl halide in the presence of a base, for example NaH or K2CO3, to provide a N-benzyl indole of formula XXI, which can be deprotected as appropriate.
[0083] Alternatively, an indole derivative of formula XIX, in which Q" is a leaving group suitable for use in a Suzuki reaction or other organo metallic coupling reaction may be alkylated with a substituted benzyl halide, incorporating protecting group as necessary, in the presence of a suitable base, for example NaH to give a N-benzyl derivative XXII. Suzuki or other suitable coupling with a heterocyclic partner as described above then can give the heterocyclic substituted indole derivative, which can be deprotected, if necessary. Alternatively, an indole of formula XIX may be reduced by treatment with a suitable reducing agent, for example, in the case where Q" is a halogen atom, sodium cyanoborohydride in the presence of acetic acid, to give an indoline derivative XX. In some cases, it may be appropriate to convert the Q" group to a different leaving group, for example from a halide to a borate ester. Such transformations are well presented in the chemistry literature. A coupling reaction with a suitably functionalized heteroaromatic species can then furnish the target compound of the invention XXIV, after deprotection, if necessary. In some cases, it may be feasible to start indoline systheses with an indoline derivative already incorporating a heteroaromatic ring, such as XXV. In this case an alkylation as described above for XVIII with a benzyl halide can furnish a compound of formula XXIV directly or after deprotection, as appropriate.
Sche
Figure imgf000036_0001
XXIV
[0084] Alternative sequences are also envisioned, in which Q" is a nitrile or converted to a nitrile. Subsequent reactions lead to 1,2,4-triazoles, oxadiazoles and tetrazoles can be carried followed well established literature precedent. Q" can also be or be converted to an alkyne following established chemistry which in turn can be reacted with a substituted azide to give 1,2,3-triazole derivatives.
[0085] In some cases, it may be desireable to construct the heteroaromatic species from a carbonyl derivative such as XXVI, XXVII or XXVIII, Y = H, OH, NHR4, or OR5, wherein R4 is H, lower alkyl or OR6, wherein is H or lower alkyl and R5 is lower alkyl or another substituent suitable for the displacement chemistry associated with the intended heterocycle construction. Such intermediates can be alkylated as above with the appropriate benzyl derivatives to give the benyzlated derivatives XXIX, XXX and XXXI, followed by elaboration of the carbonyl derivative to the desired heterocyclic derivative, XXXII, XXXIII, and XXXIV using the chemistry appropriate to the target heterocycle. In general, the sequence of steps necessary to carry out these transformations is well established in the chemistry literature. The sequence of the steps may be altered to suit the particular selection of target, available starting materials and experimental convenience.
[0086] In many cases, it may be more desirable to start with a functionalized bycylic core such as IV, V, VI, XIX or XX that can be converted to a carbonyl containing compound such as XXIX, XXX, or XXXI using standard organometallic catalyzed acylation reactions.
Figure imgf000038_0001
-37- EXAMPLES
[0087] The disclosure is further illustrated by the following examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure and/or scope of the appended claims.
Compounds
[0088] The following non-limiting compound examples serve to illustrate further embodiments of the ROR modulators. It is to be understood that any embodiments listed in the Examples section are embodiments of the ROR modulators and, as such, are suitable for use in the methods and compositions described above.
[0089] The present invention is not to be limited in scope by the specific embodiments disclosed in the examples, which are intended as illustrations of a few aspects of the invention and any embodiments that are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art and are intended to fall within the scope of the appended claims.
[0090] Preparation of 2- 4-(bromomethyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol
Figure imgf000039_0001
[0091] To a stirred solution of 2-(4-methylphenyl)-l, 1,1,3, 3,3-trifluoropropan-2-ol, 1 (100.0 g, 380 mmol) in CC14 (750 mL) was added NBS (68.6 g, 380 mmol) in portions and AIBN (0.63 g, 0.01 eq.) at RT. The resulting reaction mixture was heated to 80 °C and stirred for 2 h. The progress of the reaction was monitored by TLC (TLC system: 10 % EtO Ac/Pet ether, Rf value: 0.35).
[0092] After completion of the reaction, the reaction mixture was concentrated under reduced pressure to afford the crude product. The crude product was diluted with water and extracted with dicloromethane (2 x 500 mL). The combined organic layers were washed with water, then brine, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford crude 2-(4-(bromomethyl)phenyl)-l , l ,l ,3,3,3-hexafluoropropan-2-ol. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with pet ether to give 2-(4-(bromomethyl)phenyl)-l , l ,l ,3,3,3-hexafluoropropan-2-ol, 2 as a mixture of mono- and di-bromo derivatives. 1HNMR (400 MHz, CDC13) δ: 7.75-7.64 (m, 2 H), 7.48 (d, 2 H, J = 8.4 Hz), 4.5 (s, 2 H), 4.14-4.09 (m, 1 H).
Example 1. Preparation of l,l,l,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-pyrazol-5-yl)- 3,4-dihydroquinolin-l(2H)-yl)methyl)phenyl)propan-2-ol
[0093] Reaction step 1. Preparation of 6-bromo-l , 2,3, 4-tetrahydroquino line, 4
Figure imgf000040_0001
[0094] To a stirred solution of 1 ,2,3,4-tertrahydroquinoline, 3 (10.0 g, 75 mmol) in DMF (100 mL) was added NBS (17.4 g, 98 mmol) in portions at 0 °C and the mixture was stirred for 2 h. The progress of the reaction was monitored by TLC (TLC system: 10 % EtO Ac/Pet ether, Rf value: 0.5).
[0095] After completion of the reaction, the reaction was quenched with water and extracted with ethyl acetate (2 x 200 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford crude product. The crude product was purified over silica gel (100-200 mesh) column chromatography by eluting with 3 % EtO Ac/Pet ether to give 6-bromo-l , 2, 3, 4-tetrahydroquino line, 4 as a light yellow liquid. 1HNMR (400 MHz, CDC13) δ: 7.05-7.01 (m, 2 H), 6.36 (d, 1 H, J = 8.8 Hz), 3.29-3.27 (m, 2 H), 2.72 (t, 2 H, J = 6 Hz), 1.94-1.89 (m, 2 H).
[0096] Reaction step 2. Preparation of 2-(4-((6-bromo-3,4-dihydroquinolin-l (2H)- l)methyl)phenyl)-l , 1 , 1 ,3,3,3-hexafluoropropan-2-ol, 5
Figure imgf000040_0002
[0097] To a stirred solution of 6-bromo-l,2,3,4-tetrahydroquinoline, 4 (4.0 g, 18 mmol) in acetonitrile (100 mL) was added K2C03 (7.8 g, 56 mmol), the mixture was cooled to 0 °C and stirred for 10 min. 2-(4-(bromomethyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 2 (6.3 g, 1.0 eq.) was added dropwise and the reaction mixture was heated to 80 °C for 20 h. The progress of the reaction was monitored by TLC (TLC system: 10 % EtO Ac/Pet ether, Rf value: 0.45). At the end of this period, 30 % the starting compound 4 was present.
[0098] After 70 % completion of reaction, the reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The crude product was diluted with water and extracted with ethyl acetate (2 x 100 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 2 % EtO Ac/Pet ether to give 2-(4-((6-bromo-3,4-dihydroquinolin-l(2H)-yl)methyl)phenyl)- l,l,l,3,3,3-hexafiuoropropan-2-ol, 5 as a white solid. 1HNMR (400 MHz, CDC13) δ: 7.65 (d, 2 H, J= 8.0 Hz), 7.30 (d, 2 H, J = 8.0 Hz), 7.07 (s, 1 H), 7.03 (dd, 1 H, Jx = 8.8, J2 = 2.0 Hz), 6.29 (d, 1 H, J= 8.8 Hz), 4.47 (s, 2 H), 3.36 (t, 2 h, J= 5.6 Hz), 2.79 (t, 2 H, J= 6.4 Hz), 2.01
(m, 2 H).
[0099] Reaction step 3. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-l ,2,3,4-tetrahydroquinoline-6-carbaldehyde, 6
Figure imgf000041_0001
[0100] To a stirred solution of 2-(4-((6-bromo-3,4-dihydroquinolin-l(2H)-yl)methyl)phenyl)- 1,1,1, 3,3, 3-hexafluoropropan-2-ol, 5 (0.2 g, 0.42 mmol) in THF (10 mL) was added a 1M solution of n-BuLi in hexanes (0.85 mL, 0.85 mmol) at -78 °C and the mixture was stirred for 10 min. DMF (0.59 mL, 1.8 eq.) was added dropwise at -78 °C and the reaction mixture slowly allowed to warm to RT and stir for 1 h. The progress of the reaction was monitored by TLC (20 % EtO Ac/Pet ether, Rf value: 0.1) [0101] After completion of the reaction, the reaction mixture was quenched with saturated ammonium chloride solution and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified by silica gel (100-200 mesh) column chromatography eluting with 12-14 % EtOAc/Pet ether to give l-(4-(l, 1,1,3, 3,3-hexafluoro- 2-hydroxypropan-2-yl)benzyl)-l,2,3,4-tetrahydroquinoline-6-carbaldehyde, 6 as a yellow oil. 1HNMR (400 MHz, CDC13) δ: 7.68 (d, 2 H, J = 8.4 Hz), 7.28 (s, 1 H), 7.23 (d, 2 H, J = 8 Hz), 6.41 (d, 1 H, J =8.4 Hz), 4.51 (s, 2 H), 3.51 (s, 1 H) 3.46 (t, 2 H, J= 6 Hz), 2.81 (t, 2 H, J = 6 H), 2.03 (t, 2 H, J= 5.2 Hz).
[0102] Reaction step 4, Preparation of (E)-4-(l-(4-(l, 1,1,3, 3,3-hexafluoro-2-hydroxypropan- 2- l)benzyl)-l,2,3,4-tetrahydroquinolin-6-yl)but-3-en-2-one, 7
Figure imgf000042_0001
[0103] A stirred solution of acetone (0.10 mL, 1.4 mmol), NaOH (0.2 g, 5.0 mmol) and water (4 mL) was cooled to 0 °C. l-(4-(l,l,l,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)benzyl)- l,2,3,4-tetrahydroquinoline-6-carbaldehyde, 6 (0.2 g, 0.48 mmol) dissolved in ethanol (5 mL), was added at 0 °C and the mixture was stirred for 10 min. The resulting reaction mixture was allowed to warm to RT and stir for 8 h. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.25).
[0104] After completion of the reaction, the reaction mixture was diluted with ethyl acetate and washed with water. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to give crude (E)-4-(l-(4-(l, 1,1,3, 3,3- hexafluoro-2-hydroxypropan-2-yl)benzyl)-l,2,3,4-tetrahydroquinolin-6-yl)but-3-en-2-one, 7 as a yellow liquid. LC-MS: 96.7 % at 215 nm (m/z 418 [M+H]).
[0105] Reaction step 5, Preparation of l,l,l,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-pyrazol- 5 -yl)-3 ,4-dihydroquinolin- 1 (2H)-yl)methyl)phenyl)propan-2-ol, 8
Figure imgf000043_0001
[0106] To a solution of (E)-4-(l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)- l,2,3,4-tetrahydroquinolin-6-yl)but-3-en-2-one, 7 (0.45 g, 0.98 mmol) in water (10 mL), NaOH (0.06 g, 1.5 mmol), TBAB (0.475 g, 1.5 mmol), tosyl hydrazine (0.22 g, 3.0 mmol) were added at RT. The resulting reaction mixture was heated to 85 °C for 12 h. The progress of the reaction was monitored by TLC, (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.5)
[0107] After completion of the reaction, the reaction mixture was diluted with water and extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified by preparative HPLC followed by preparative TLC; to afford 1,1,1 ,3 ,3 ,3 -hexafluoro-2-(4-((6-(3 -methyl- 1 H-pyrazol-5 -yl)-3 ,4-dihydroquinolin- 1 (2H)- yl)methyl)phenyl)propan-2-ol, 8 as an off white solid. 1H NMR (400 MHz, DMSO-d6) δ:12.5 (br, s, 1 H), 8.63 (s, 1 H), 7.63 (d, 2 H, J = 8 Hz), 7.39 (d, 2 H, J = 8.4 Hz), 7.29 (s, 1 H), 7.22 (d, 1 H, J = 8.4 Hz), 6.43 (d, 1 H, J= 8.8 Hz), 6.17 (s, 1 H), 4.56 (s, 2 H), 3.40 (t, 2 H, J = 5.2 Hz), 2.78 (t, 2 H, J = 6 Hz), 2.17 (s, 3 H), 1.97 (t, 2 H, J = 5.6 Hz). LC-MS: 95.94 % at 215 nm and 95.36 % at 254 nm (m/z = 470.1, [M+H]+).
Example 2. Preparation of 2-(4-((6-(lH-pyrazol-4-yl)-3,4-dihydroquinolin-l(2H)- yl)methyl)phenyl)- 1 , 1 ,1 ,3,3,3-hexafluoropropan-2-ol
[0108] Reaction step 1. Preparation of tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- - lH-pyrazole- 1 -carboxylate, 10
Figure imgf000043_0002
[0109] To a stirred solution of 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole, 9 (2.0 g, 10 mmol) in DMF (50 mL) was added DMAP (3.14 g, 25 mmol), the mixture was cooled to 0 °C and stirred for 10 min. (Boc)20 (2.9 mL, 13 mmol) was added dropwise and the mixture was stirred for 15 min at same temperature. The resulting reaction mixture was allowed to warm to RT and stir overnight. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.8)
[0110] After completion of the reaction, the reaction mixture was diluted with ice cold water and extracted with ethyl acetate (2 x 100 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 10 % EtOAc/Pet ether to give tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole-l-carboxylate, 10 as a white solid.
[0111] Reaction steps 2 and 3. Preparation of 2-(4-((6-(lH-pyrazol-4-yl)-3,4- dih droquinolin- 1 (2H)-yl)methyl)phenyl)- 1,1,1 ,3 ,3 ,3-hexafluoropropan-2-ol, 11
Figure imgf000044_0001
[0112] To a stirred solution of 2-(4-((6-bromo-3,4-dihydroquinolin-l(2H)-yl)methyl)phenyl)- 1,1,1, 3,3, 3-hexafluoropropan-2-ol 5 (0.8 g, 1.7 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate 10 (0.65 g, 2.2 mmol) in DMF (30 mL) and water (3 mL) was added CS2CO3 (1.67 g, 5.1 mmol) the solution was deoxygenated by the passage of argon for 2 h. retrafo's-triphenylphosphine palladium (0.2 g, 0.17 mmol) was added and the mixture was heated to 80 °C and stirred overnight. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rvalue: 0.2).
[0113] By LC-MS 50% de-Boc product was observed. The reaction mass was filtered through a celite bed and concentrated. The crude product was treated with 2M HCl in dioxane and stirred for 2 h.
[0114] After completion of reaction, the reaction mixture was concentrated under reduced pressure, the residue was diluted with ethyl acetate and washed with saturated NaHC03 solution. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford crude product. The crude product was purified by preparative TLC and to give 2-(4-((6-(lH-pyrazol-4-yl)-3,4-dihydroquinolin-l(2H)- yl)methyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 11 as an off-white solid. 1H NMR (400
MHz, DMSO-d6) δ: 12.68 (s, 1 H), 8.62 (s, 1 H), 7.79 (br, s, 2 H), 7.63 (d, 2 H, J = 8.4 Hz),
7.39 (d, 2 H, J = 8.4 Hz), 7.15 (s, 1 H), 7.09 (dd, 1 H, Ji = 8.4, J2 = 2.4 Hz), 6.41 (d, 2 H, J
= 8.4 Hz), 4.54 (s, 2 H), 3.38 (t, 2 H, J = 5.6 Hz), 2.77 (t, 2 H, J = 6 Hz), 1.98- 1.94 (m, 2
H). LC-MS: 97.22 % at 215 nm and 95.73 % at 254 nm (m/z = 456.1, [M+H]+).
Example 3. Preparation of l,l,l,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-l,2,4-triazol-5- yl)-3,4-dihydroquinolin-l(2H)-yl)methyl)phenyl)propan-2-ol
[0115] Reaction step 1. Preparation of 6-bromo-3,4-dihydroquinolin-2(lH)-one, 13
Figure imgf000045_0001
[0116] 3,4-Dihydroquinolin-2(lH)-one, 12 (25.0 g, 170 mmol) in DMF (50 mL) was added to a stirred solution of NBS (33.2 g, 186 mmol) in DMF (250 mL) at 0 °C and the mixture was stirred for 1 h. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rvalue: 0.35).
[0117] After completion of the reaction, the reaction mixture was quenched with water and then solid formed. The solid was filtered and dried under vacuum to afford the crude product. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 3 % EtO Ac/Pet ether to give 6-bromo-3,4-dihydroquinolin-2(lH)-one, 13 as a white solid. 1H NMR (400 MHz, CDC13) δ: 8.73 (s, 1 H), 7.28 (dd, 2 H, J, = 10.8, J2 = 2.4 Hz), 6.69 (d, 1 H, J = 8.0 Hz), 2.95 (t, 2 H, J = 7.2 Hz), 2.65-2.61 (m, 2 H); LC-MS: 96.7 % at 215 nm (m/z 418 [M+H]).
[0118] Reaction step 2, Preparation of 6-bromo-l-(4-(l, 1,1,3, 3,3-hexafluoro-2- h droxypropan-2-yl)benz l)-3 ,4-dihydroquinolin-2( lH)-one, 14
Figure imgf000045_0002
[0119] A solution of 6-bromo-3,4-dihydroquinolin-2(lH)-one, 13 (10.0 g, 44 mmol) in THF (25 mL) was added to stirred solution of 60% NaH (3.53 g, 88 mmol) in THF (100 mL) at 0 °C and the mixture was stirred for 30 min. A solution of 2-(4-(bromomethyl)phenyl)- 1,1,1, 3,3, 3-hexafluoropropan-2-ol (2) (18.4 g, 55.3 mmol) in THF (25 mL) was added and slowly heated to 65 °C overnight. The progress of the reaction was monitored by TLC (TLC system: 30 % EtO Ac/Pet ether, Rf value: 0.45).
[0120] After completion of the reaction, the reaction mixture was poured into ice cold water and extracted with ethyl acetate (2 x 200 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was washed with pet ether followed by dichloromethane to afford 6-bromo-l-(4- (1,1,1 ,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-3,4-dihydroquinolin-2(lH)-one, 14. 1H NMR (400 MHz, DMSO-d6) δ: 8.65 (s, 1 H), 7.61 (d, 2 H, J = 8.4 Hz), 7.47 (d, 1 H, J = 2.0 Hz), 7.35-7.30 (m, 3 H), 6.85 (d, 1 H, J = 8.4 Hz), 5.17 (s, 2 H), 2.98 (t, 2 H, J= 6.8 Hz), 2.71 (m, 2 H).
[0121] Reaction step 3. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-2-oxo-l,2,3,4-tetrahydroquinoline-6-carbonitrile, 15
Figure imgf000046_0001
[0122] To a stirred solution of 6-bromo-l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-3,4-dihydroquinolin-2(lH)-one 14 (2.0 g, 4.0 mmol) in DMF (10 mL) was added CuCN (1.2 g, 12.0 mmol) in a sealed tube. The resulting reaction mixture was heated to 150 °C for 2 h. The progress of the reaction was monitored by TLC (TLC system: 30 % EtO Ac/Pet ether, Rf value: 0.4).
[0123] After completion of reaction, the reaction mixture was cooled to RT, quenched with aq. ammonia and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 10 % EtOAc/Pet ether to give l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-2-oxo-l ,2,3,4-tetrahydroquinoline-6-carbonitrile, 15 that was used directly in the next step.
[0124] Reaction step 4. Preparation of l-(4-(l , l ,l ,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-6-(3-methyl-lH-l ,2,4-triazol-5-yl)-3,4-dihydroquinolin-2(lH)-one, 16
Figure imgf000047_0001
[0125] To a stirred solution of l-(4-(l , l ,l ,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-2- oxo-l ,2,3,4-tetrahydroquinoline-6-carbonitrile, 15 (0.3 g, 0.70 mmol) in n-butanol (100 mL) was added K2CO3 (0.96 g, 7.0 mmol) and acetylhydrazide (0.33 g, 7.3 mmol) at RT. The resulting reaction mixture was heated to 140 °C and stirred for 40 h. The progress of the reaction was monitored by TLC (TLC system: 5 % MeOH/dicloromethane (KMn04 active), Rf value: 0.5).
[0126] After completion of reaction, the reaction mixture was cooled to RT and concentrated under reduced pressure. The crude product was diluted with water and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 70 % EtO Ac/Pet ether to give l-(4-(l ,l , l ,3 ,3 ,3 -hexafluoro-2-hydroxypropan-2-yl)benzyl)-6-(3 -methyl- 1 H- 1 ,2,4-triazol-5 - yl)-3,4-dihydroquinolin-2(lH)-one, 16 as an off white solid. 1H NMR (400 MHz, DMSO-d6) δ: 13.56 (s, 1 H), 8.65 (s, 1 H), 7.84 (s, 1H), 7.71 (d, 1 H, J = 8.0 Hz), 7.62 (d, 2 H, J = 8.0 Hz), 7.37 (d, 2 H, J = 8.4 Hz), 6.97 (d, 1 H, J = 8.4 Hz), 5.21 (s, 2 H), 3.03 (t, 2 H, J= 7.6 Hz), 2.74 (t, 2 H, J= 7.6 Hz), 2.37 (s, 3 H). LC-MS: 96.79 % at 215 nm and 92.43 % at 254 nm (m/z = 485.1 , [M+H]+).
[0127] Reaction step 5. Preparation of l , l ,l ,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-l ,2,4- triazol-5 -yl)-3 ,4-dihydroquinolin- 1 (2H)-yl)methy l)phenyl)propan-2-ol, 17
Figure imgf000048_0001
[0128] To a stirred solution of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-6- (3-methyl-lH-l,2,4-triazol-5-yl)-3,4-dihydroquinolin-2(lH)-one, 16 (0.14 g, 0.20 mmol) in THF (20 niL) was added BH3-DMS (2M) (0.31 niL, 0.6 mmol) at 0 °C and the mixture was stirred for 10 min. The resulting reaction mixture was heated to reflux and was stirred for 2 h. The reaction mixture was quenched with methanol and concentrated under reduced pressure. 2N HC1 (10 mL) was added to crude product and the mixture heated to 100 °C for 1 h. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.5).
[0129] After completion of reaction, the reaction mixture was cooled to RT, the pH was adjusted to 10 with NaHC03 solution, and it was extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified by preparative HPLC to afford 1,1,1 ,3,3,3-hexafluoro-2-(4-((6-(3-methyl-lH-l ,2,4-triazol-5-yl)-3,4-dihydroquinolin- l(2H)-yl)methyl)phenyl)propan-2-ol, 17 as a brown semi-solid. 1H NMR (400 MHz, DMSO- d6) δ: 7.67-7.62 (m, 3 H), 7.51 (s, 1 H), 7.34 - 7.31 (m, 3 H), 6.49 (d, 1 H, J = 8.8 Hz), 4.57 (s, 2 H), 3.49 (s, 1 H), 3.42 (t, 2 H, J= 6 Hz), 2.88 (t, 2 H, J= 6 Hz), 2.47 (s, 3 H), 2.10-2.00 (m, 3 H). LC-MS: 95.06 % at 215 nm and 95.17 % at 254 nm (m/z = 471.1, [M+H]+).
Example 4. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-6- (3-methyl-lH-pyrazol-5-yl)-3,4-dihydroquinolin-2(lH)-one
[0130] Reaction step 1. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-2-oxo-l,2,3,4-tetrahydroquinoline-6-carbaldehyde, 18
Figure imgf000048_0002
[0131] In a steel bomb a mixture of 6-bromo-l-(4-(l , l ,l ,3,3,3-hexafluoro-2-hydroxypropan- 2-yl)benzyl)-3,4-dihydroquinolin-2(lH)-one, 14 (1.0 g, 2.1 mmol) sodium formate (0.09 g, 1.3 mmol) and PdCl2P(Ph3)2 (0.12 g, 0.17 mmol) in DMF (10 mL) was deoxygenated with argon for 20 min and then CO gas was introduced into the bomb, the bomb was sealed. The pressure adjusted to 250 psi and the temperature raised to 100 °C. The reaction mixture was stirred for 30 h at 100 °C. The progress of the reaction was monitored by TLC (TLC system: 50 % EtOAc / Pet ether, Rf value: 0.4).
[0132] After completion of the reaction, the reaction mixture was diluted with water and extracted with ethyl acetate (2 x 80 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 25 % EtO Ac/Pet ether to give l-(4-(l ,l , l ,3,3,3-hexafluoro-2- hydroxypropan-2-yl)benzyl)-2-oxo-l ,2,3,4-tetrahydroquinoline-6-carbaldehyde, 18 as a colorless liquid which was used directly in the next step.
[0133] Reaction step 2. Preparation of (E)-l-(4-(l , l ,l ,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-6-(3 -oxobut- 1 -en- 1 -yl)-3 ,4-dihydroquinolin-2( lH)-one, 19
Figure imgf000049_0001
[0134] To a stirred solution of acetone (0.2 g, 2.7 mmol) and NaOH (0.1 g, 2.5 mmol) in water (4 mL) was added l-(4-(l , 1 , 1 ,3, 3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-2-oxo- l ,2,3,4-tetrahydroquinoline-6-carbaldehyde, 18 (0.1 g, 0.23 mmol) dissolved in EtOH (15 mL) at 0 °C. The resulting reaction mixture was allowed to warm to RT and was stirred overnight. The progress of the reaction was monitored by TLC (TLC system: 50 % EtOAc / Pet ether, Rf value: 0.3).
[0135] After completion of the reaction, reaction the pH was adjusted to 6 with KHS04 solution and extracted with MTBE (2 x 50mL). The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to give (E)-l-(4-
(1 ,1 , 1 ,3 ,3 ,3 -hexafluoro-2-hydroxypropan-2-yl)benzyl)-6-(3 -oxobut- 1 -en- 1 -yl)-3 ,4- dihydroquinolin-2(lH)-one, 19 as a white solid. 1H NMR (400 MHz, CDC13) δ : 7.67 (d, 2 H, J = 8.4 Hz), 7.42 (d, 1 H, J = 16.4 Hz), 7.39 (m, 1 H), 7.33-7.27 (m, 3 H), 6.85 (d, 1 H, J = 8.4 Hz), 6.63 (d, 1 H, J= 16.0 Hz), 5.21 (s, 2H), 3.03 (t, 2 H, J= 8.4 Hz), 2.82 (t, 2 H, J= 8.0 Hz), 2.35 (s, 3 H). LC-MS: 73.98 % (m/z = 472.1, [M+H]+).
[0136] Reaction step 3. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-6-(3-methyl-lH-pyrazol-5-yl)-3,4-dihydroquinolin-2(lH)-one, 20
Figure imgf000050_0001
[0137] To a stirred solution of (E)-l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)-6-(3-oxobut-l-en-l-yl)-3,4-dihydroquinolin-2(lH)-one, 19 (0.15 g, 0.32 mmol) in water (5 niL) was added tosyl hydrazide (0.186 g, 0.38 mmol), TBAB (0.153 g, 0.48 mmol) and NaOH (0.019 g, 0.48 mmol) simultaneously at RT. The resulting reaction mixture was heated to 80 °C and stirred for 2 h. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.5).
[0138] After completion of reaction, brine solution was added to the reaction mixture and it was extracted with ethyl acetate (2 x 50 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with pure ethyl acetate to afford l-(4-(l, 1,1,3, 3,3-hexafluoro-2-hydroxypropan-2 -yl)benzyl)-6-(3- methyl-lH-pyrazol-5-yl)-3,4-dihydroquinolin-2(lH)-one, 20 as an off-while solid. 1H NMR (400 MHz, DMSO-de) δ: 12.44 (s, 1 H), 8.64 (s, 1 H), 7.62 (d, 3 H, J= 8.0 Hz), 7.48 (d, 1 H, J = 8.4 Hz), 7.37 (d, 2 H, J = 8.4 Hz), 6.91 (d, 1 H, J = 8.4 Hz), 6.35 (s, 1 H), 5.20 (s, 2 H), 3.00 (t, 2 H, J = 7.2 Hz), 2.73 (t, 2 H, J = 7.2 Hz), 2.23 (br s, 3 H), 1.23 (s, 1 H). LC-MS: 95.13 % at 215 nm and 92.96 % at 254 nm (m/z = 484.1, [M+H]+).
Example 5. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-6- (lH-pyrazol-4-yl)-3,4-dihydroquinolin-2(lH)-one
Figure imgf000051_0001
[0139] A suspension of 6-bromo-l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)- 3,4-dihydroquinolin-2(lH)-one, 14 (0.5 g, 1.0 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 (0.44 g, 1.5 mmol) in DMF (5 mL) was deoxygenated by passage of argon for 20 min, PdCl2 (dppf) (0.081 g, 0.1 mmol) and potassium acetate (0.30 g, 3.1 mmol) was added at RT. The resulting reaction mixture was heated to 60 °C for 4 h. The progress of the reaction was monitored by TLC (TLC system: 70% % EtOAc / Pet ether, Rf value: 0.1)
[0140] After completion of the reaction, the reaction mixture was diluted with ethyl acetate and washed with ice cold water. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford crude product. A solution of hydrochloric acid in dioxane (1 mL) was added to the crude product and the mixture was stirred for 30 min. The reaction mixture was concentrated under reduced pressure. The crude product was diluted with ethyl acetate and washed with saturated sodium bicarbonate solution. The organic layer was dried over sodium sulfate and concentrated under reduced pressure to afford a crude product that was titurated with diethyl ether to afford l-(4- (1,1,1 ,3 ,3 ,3 -hexafluoro-2-hydroxypropan-2-yl)benzyl)-6-( 1 H-pyrazol-4-yl)-3 ,4- dihydroquinolin-2(lH)-one, 21 as an off-white solid. 1H NMR (400 MHz, DMSO d6) δ: 12.9 (s, 1 H), 8.64 (s, 1 H), 8.08 (br, s, 1 H), 7.83 (br, s, 1 H), 7.61 (d, 2 H, J= 8.4 Hz), 7.48 (d, 1 H, J = 2.0 Hz) 7.40-7.30 (m, 3 H), 7.07 (br, s, 1 H), 6.88 (d, 1 H, J = 6.6 Hz), 5.19 (s, 2 H), 2.98 (t, 2 H, J= 7.6 Hz), 2.71 (t, 2 H, J= 8.0 Hz). LC-MS: 97.46 % at 215 nm and 97.12 % at 254 nm (m/z = 470.1, [M+H]+).
Example 6. Preparation of Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2- hydroxypropan-2-yl)benzyl)-6-(3-methyl-lH-l,2,4-triazol-5-yl)-3,4-dihydroquinolin-
2(lH)-one [0141] K2CO3 (0.97 g, 7.0 mmol) was added to a stirred solution of l-(4-(l, 1,1,3,3,3- hexafluoro-2-hydroxypropan-2-yl)benzyl)-2-oxo- 1 ,2,3 ,4-tetrahydroquinoline-6-carbonitrile, 15 (0.3 g, 0.70 mmol) and acetylhydrazide (0.52 mL, 7.0 mmol) in n-butanol (25 mL) at RT. The resulting reaction mixture was heated to 125 °C and stirred for 3 days. The progress of the reaction was monitored by TLC (TLC system: 5 % MeOH/dicloromethane, Rf value: 0.15).
[0142] After completion of the reaction, the reaction mixture was concentrated under reduced pressure to afford the crude product. The crude product was purified over silica gel (60-120 mesh) column chromatography by eluting with 50 % EtO Ac/Pet ether to give l-(4- (1,1,1 ,3,3,3-hexafluoro-2-hydroxypropan-2-yl)benzyl)-6-(3-methyl-lH-l ,2,4-triazol-5-yl)- 3,4-dihydroquinolin-2(lH)-one, 16 as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ: 13.56 (s, 1 H), 8.65 (s, 1 H), 7.84 (s, 1H), 7.71 (d, 1 H, J = 8.0 Hz), 7.62 (d, 2 H, J = 8.0 Hz), 7.37 (d, 2 H, J = 8.4 Hz), 6.97 (d, 1 H, J = 8.4 Hz), 5.21 (s, 2 H), 3.03 (t, 2 H, J= 7.6 Hz), 2.74 (t, 2 H, J= 7.6 Hz), 2.37 (s, 3 H). LC-MS: 96.79 % at 215 nm and 92.43 % at 254 nm (m/z = 485.1, [M+H]+).
Example 7. Preparation of l,l,l,3,3,3-hexafluoro-2-(4-((5-(3-methyl-lH-pyrazol-5- yl)indolin-l-yl)methyl)phenyl)propan-2-ol
[0143] Reaction step 1. Preparation of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)- 1 1 ,1 ,3,3,3-hexafluoropropan-2-ol, 23
Figure imgf000052_0001
[0144] 60% NaH (4.04 g, 101 mmol) was added to a stirred solution of 2-(4- (bromomethyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 2 (5.0 g, 25.3 mmol) and 5- bromoindoline, 22 (11.02 g, 32.8 mmol) in THF (50 mL) at 0 °C. The resulting reaction mixture was refluxed for 2 h. The progress of the reaction was monitored by TLC (TLC system: 20 % EtOAc / Pet ether, Rf value: 0.55).
[0145] After 70 % completion of reaction, the reaction mixture was quenched with ice and extracted with ethyl acetate (2 x 300 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified by column chromatorgraphy over silica gel (100- 200 mesh) eluting with 10 % EtO Ac/Pet ether to give 2-(4-((5-bromoindolin-l- yl)methyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 23 as a pale yellow liquid. 1H NMR (400 MHz, CDC13) δ: 7.68 (d, 2 H, J = 8.8 Hz), 7.42 (d, 2 H J = 8.4 Hz), 7.17 (s, 1 H), 7.14 (d, 1 H, J= 8.4 Hz), 6.32 (d, 1 H, J= 8.4 Hz), 4.25 (s, 2 H), 3.41 (s, 1H) 3.35 (d, 2 H, J= 8.0 Hz), 2.98 (t, 2 H, J= 8.0 Hz).
[0146] Reaction step 2. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carbaldehyde, 24
Figure imgf000053_0001
[0147] To a stirred solution of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3, 3,3- hexafluoropropan-2-ol, 23 (3.0 g, 6.6 mmol) in THF (15 mL) was added a solution of 1M n- BuLi in hexanes (13.2 mL, 13.2 mmol) at -78 °C and the mixture was stirred for 30 min. DMF (0.29 mL, 11.8 mmol) was added dropwise and the mixture was stirred for 15 min at the same temperature. The resulting reaction mixture was allowed warm to RT and stir for 3 h. The progress of the reaction was monitored by TLC (TLC system: 30 % EtO Ac/Pet ether, Rf value: 0.3).
[0148] After completion of the reaction, the reaction mixture was quenched with saturated ammonium chloride solution and extracted with ethyl acetate (2 x 150 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to give crude l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carbaldehyde, 24 as a liquid. LC-MS: 71.77% at 215 nm (m/z = 404.1 [M+H]+).
[0149] Reaction step 3. Preparation of (E)-4-(l-(4-(l , 1 , 1 ,3, 3,3-hexafluoro-2-hydroxypropan- 2-yl)benzyl)indolin-5-yl)but-3-en-2-one, 25
Figure imgf000054_0001
[0150] To a stirred solution of l-(4-(l, l ,l ,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carbaldehyde 24 (0.3 g, 0.74 mmol) in H20 (10 mL) was added acetone (0.18 mL, 2.2 mmol) dropwise at 0 °C and the mixture was stirred for 10 min. 10% NaOH (4 mL) was added dropwise at 0 °C, the mixture was allowed to warm to RT and stir overnight. The progress of the reaction was monitored by TLC (TLC system: 30 % EtO Ac/Pet ether, Rf value: 0.5).
[0151] After completion of the reaction, the reaction mixture was diluted with ethyl acetate, the organic layer was separated, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to give (E)-4-(l-(4-(l , 1 , 1 ,3, 3,3-hexafluoro-2- hydroxypropan-2-yl)benzyl)indolin-5 -yl)but-3 -en-2-one, 25 as a yellow liquid. 1H NMR (400 MHz, DMSO-de) δ: 7.69 (d, 2 H, J = 7.6 Hz), 7.46-7.38 (m, 3 H), 7.30 (s, 1 H), 7.23 (s, 1 H), 6.52 (d, 1 H, J = 15.6 Hz), 6.42 (d, 1H J = 8.0 Hz), 4.35 (s, 2 H), 3.80 (bs, 1 H), 3.52-3.43 (m, 2 H), 3.05 (t, 2 H, J = 8.4 Hz), 2.33 (s, 3 H).
[0152] Reaction step 5. Preparation of l ,l , l ,3,3,3-hexafluoro-2-(4-((5-(3-methyl-lH-pyrazol- 5-yl)indolin- 1 -yl)methyl)phenyl)propan-2-ol, 26
Figure imgf000055_0001
[0153] A mixture of (E)-4-(l-(4-(l, 1,1,3, 3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indolin-5-yl)but-3-en-2-one, 25 (0.5 g, 1.1 mmol), tosyl hydrazine (0.25 g, 1.3 mmol), TBAB (0.54 g, 1.6 mmol) in THF (50 mL) was added NaOH (0.067 g, 1.6 mmol) in water (4 mL) at RT. The reaction mixture was heated to 100 °C and stirred overnight. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.45).
[0154] After completion of the reaction, the reaction mixture was cooled to RT, diluted with ice cold water and extracted with ethyl acetate (2 x 80 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was triturated with ethyl acetate (60 mL) to afford 1,1,1,3,3,3- hexafluoro-2-(4-((5 -(3 -methyl- lH-pyrazol-5-yl)indolin- 1 -yl)methyl)phenyl)propan-2-ol, 26 as a pale yellow solid. 1H NMR (DMSO-d6) δ: 12.20 (br, s, 1 H), 8.66 (s, 1 H), 8.31 (s, 1 H), 7.66 (d, 2 H, J= 8.0 Hz), 7.49 (d, 2 H, J= 8.4 Hz), 7.37 (br, s, 2 H), 6.58 ( br, s, 1 H), 6.22 (s, 1 H), 4.35 (s, 2 H), 3.31 (s, 2 H), 2.95 (t, 2 H, J= 8.0 Hz), 2.20 (br, s, 3 H). LC-MS: 99.04 % at 215 nm (m/z = 456.1, [M+H]+).
Example 8. Preparation of 2-(4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)phenyl)- l,l,l,3,3,3-hexafluoropropan-2-ol
Figure imgf000055_0002
[0155] To a stirred solution of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3, 3,3- hexafluoropropan-2-ol, 23 (0.4 g, 0.88 mol) and tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate 10 (0.336 g, 1.2 mmol) in DMF (4 mL) and water (2 mL) was added CS2CO3 (0.800 g, 2.5 mmol) the mixture was deoxygenated by the passage of argon for 2 h. Pd(PPh3)4 (0.098 g, 0.084 mmol) was added and the mixture was heated to 100 °C for 2 h. The progress of the reaction was monitored by TLC (TLC system: 50 % EtO Ac/Pet ether, Rf value: 0.22).
[0156] After completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with ethyl acetate and washed with water. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. After column purification the product was isolated with 60 % purity to afford 2-(4-((5-(lH-pyrazol-4- yl)indolin-l-yl)methyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 27 as a white solid.
1H NMR (400 MHz, DMSO-d6) δ: 12.70 (br, s, 1 H), 8.66 (s, 1 H), 7.95 (s, 1 H), 7.74 (s, 1 H), 7.66 (d, 2 H, J = 8.0 Hz), 7.50 (d, 2 H, J = 8.4 Hz), 7.30 (s, 1 H), 7.22 (d, 1 H, J = 8.0 Hz), 6.56 (d, 1 H, J= 8.0 Hz), 4.32 (s, 2 H), 3.28 (t, 2 H, J= 8.0 Hz), 2.95-2.89 (m, 2 H). LC- MS: 96.32% at 215 nm and 86.20% 254 nm (m/z = 440.1 [M-H]+). HPLC: 97.58% 284 nm. Example 9. Preparation of l,l,l,3,3,3-hexafluoro-2-(4-((5-(5-methyl-l,3,4-oxadiazol-2- yl)indolin-l-yl)methyl)phenyl)propan-2-ol
[0157] Reaction step: 1 Preparation of methyl l-(4-(l, 1,1,3, 3,3-hexafluoro-2-hydroxypropan- 2- l)benzyl)indoline-5-carboxylate, 28.
Figure imgf000056_0001
[0158] To a stirred solution of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3, 3,3- hexafluoropropan-2-ol 23 (4.0 g, 8.8 mmol) in MeOH (32 mL) was added Et3N (3.5 mL, 26 mmol) and the solution was deoxygenated for 15 min. Pd(dppf)Cl2 (0.36 g, 0.44 mmol) was added in steel bomb and the mixture was deoxygenated with nitrogen for 15 min in the autoclave vessel. CO gas was passed through the reaction mixture, the vessel sealed and was heated to 100 °C, 150 psi overnight. The progress of the reaction was monitored by TLC (TLC system: 20 % EtO Ac/Pet ether, Rf value: 0.35). [0159] After completion of the reaction, the reaction mixture was filtered through a celite bed and the eluate extracted with ethyl acetate (2 x 150 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 14% EtOAc/Pet ether to afford methyl l-(4-(l,l,l,3,3,3-hexafluoro-2- hydroxypropan-2-yl)benzyl)indoline-5-carboxylate, 28 as a light brown liquid. 1H NMR (400 MHz, CDCls) δ: 7.80 (d, 1 H, J = 7.2 Hz), 7.73 (s, 1 H), 7.68 (d, 2 H J = 8.0 Hz), 7.39 (d, 2 H, J = 8.0 Hz), 6.41 (d, 1 H, J = 8.4 Hz), 4.40 (s, 2 H), 3.84 (s, 3 H) 3.55-3.48 (m, 2 H), 3.06 (t, 2 H, J= 8.8 Hz). LC-MS: 83.99 % at (m/z = 434.1, [M+H]+).
[0160] Reaction step 2. Preparation of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- l)benzyl)indoline-5-carboxylic acid, 29
Figure imgf000057_0001
[0161] To a stirred solution of methyl l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carboxylate, 28 (1.0 g, 2.3 mmol) in THF (5.0 mL) and methanol (5.0 mL) was added 10% NaOH (3 mL) at RT. The resulting reaction mixture was heated to 70 °C for 2 h. The progress of the reaction was monitored by TLC (TLC system: 80 % EtO Ac/Pet ether, Rf value: 0.2)
[0162] After completion of reaction, the reaction mixture was evaporated under reduced pressure, the residue was diluted with cold water, acidified to pH 2 with 2N HCl solution and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with water and brine solution, dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to give l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carboxylic acid as an off white solid.
[0163] Reaction step 3. Preparation of N'-acetyl-l-(4-(l,l,l,3,3,3-hexafluoro-2- hydroxypropan-2-yl)benzyl)indoline-5-carbohydrazide, 30
Figure imgf000058_0001
[0164] To a stirred solution of l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carboxylic acid, 29 (0.2 g, 0.4 mmol) in DMF (4 mL) was added acetyl hydrazine (0.103 g, 1.4 mmol), HATU (0.27 g, 0.7 mmol) at 0 °C and the mixture was stirred for 20 min. DIPEA (0.314 g, 2.0 mmol) was added dropwise and the mixture allowed warm to RT and stir overnight. The progress of the reaction was monitored by TLC (TLC system: 100 % EtOAc (UV & KMn04), Rf value: 0.01).
[0165] After completion of reaction, the reaction mixture was diluted with water and extracted with ethyl acetate (4 x 50 mL). The organic layer was dried over sodium sulfate and evaporated under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography by eluting with 80% ethyl acetate/ pet ether to afford N- acetyl- l-(4-(l,l,l ,3 ,3 ,3 -hexafluoro-2-hydroxypropan-2-yl)benzyl)indoline-5 -carbohydrazide,
30 as a colorless liquid. 1H NMR (400 MHz, DMSO-d6) δ: 10.80 (s, 1 H), 9.83 (s, 1 H), 9.721 (s, 1 H), 8.70 (s, 1H), 7.66 (d, 2 H, J = 8.4 Hz), 7.60 (d, 2 H, J = 8.4 Hz), 7.46 (d, 1 H, J= 4.8 Hz), 7.29 (dd, 1 H, Jx = 10.8, J2 = 5.2 Hz), 6.60 (dd, 1 H, Jx= 8.4, J2 = 4.0 Hz), 4.45 (s, 2 H), 3.42-3.34 (m, 3.0 Hz), 2.97-2.89 (m, 2 H),2.78-2.68 (m, 2 H), 2.50-2.20 (m, 2 H).
[0166] Reaction step 4. Preparation of 1,1, 1,3,3, 3-hexafluoro-2-(4-((5-(5-methyl-l, 3,4- oxadiazol-2- l)indolin- 1 -yl)methyl)phenyl)propan-2-ol, 31
Figure imgf000058_0002
[0167] A mixture of N'-acetyl-l-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2- yl)benzyl)indoline-5-carbohydrazide, 30 (0.180 g, 0.37 mmol) and POCl3 (4 mL) was heated to 130 °C for 6 h. The progress of the reaction was monitored by TLC (TLC system: 70 % EtO Ac/Pet ether, Rf value: 0.4)
[0168] After completion of the reaction, the reaction mixture was evaporated under reduced pressure. The crude product was washed with saturated NaHC03 and extracted with ethyl acetate (3 x 80 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure to afford the crude product. The crude product was purified by prep HPLC to afford l,l,l,3,3,3-hexafluoro-2-(4-((5-(5-methyl- l,3,4-oxadiazol-2-yl)indolin-l-yl)methyl)phenyl)propan-2-ol, 31 as an off- while solid. 1H NMR (400 MHz, DMSO-d6) δ: 8.70 (br, s, 1 H), 7.67 (d, 2 H, J = 8.0 Hz), 7.60 (d, 2 H, J = 6.8 Hz), 7.47 (d, 2 H, J= 8.4 Hz), 6.68 (t, 1 H, J= 6.0 Hz), 4.48 (s, 2 H), 3.49 (t, 2 H, J= 8.4 Hz), 3.04 (t, 2 H, J= 8.0 Hz), 2.5 (s, 3 H). LC-MS: 99.60% at 215 nm (m/z = 458.1 [M+H]+). Example 10. Preparation of 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide
[0169] Step 1. Preparation of methyl 4-((5-bromoindolin-l-yl)methyl)benzoate, 32
Figure imgf000059_0001
[0170] Methyl 4-((5-bromoindolin-l-yl)methyl)benzoate can be prepared by alkylation of 5- bromoindoline with methyl (4-bromomethyl)benzoate in the presence of NaH in THF using the general procedure described in Example 7, step 1.
-((5-bromoindolin-l-yl)methyl)benzamide, 33
Figure imgf000059_0002
[0172] 4-((5-bromoindolin-l-yl)methyl)benzamide, 33 can be prepared by hydrolysis of the methyl ester of 32 by treatment with NaOH in EtOH until hydrolysis is complete followed by activation of the resulting acid with isobutyl chloro formate in the presence of NMP in THF at
-50 °C for 30 min and the gradual introduction of a solution of NH3 in THR. [0173] Step 3. Preparation of 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 34
Figure imgf000060_0001
[0174] 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 34 can be prepared by deoxygenation of a solution of 4-((5-bromoindolin-l-yl)methyl)benzamide, 33, tert-butyl 4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 and CS2CO3 in DMF: water 2: 1 by the passage of argon for 20 min followed by the introduction of Pd(PPh3)4 and heating the mixture to 100 °C for 2 h. Treatment of the crude product with HC1 in dioxane then can provide 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 34.
Example 11. Preparation of 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)- V- methylbenzamide
[0175] Preparation of methyl 4-((5-bromoindolin-l-yl)methyl)benzoate, 32
Figure imgf000060_0002
[0176] Methyl 4-((5-bromoindolin-l-yl)methyl)benzoate can be prepared by alkylation of 5- bromoindoline with methyl (4-bromomethyl)benzoate in the presence of NaH in THF using the general procedure described in Example 7, step 1.
-((5-bromoindolin-l-yl methyl)-N-methylbenzamide, 35
Figure imgf000060_0003
[0178] 4-((5-bromoindolin-l-yl)methyl)-N-methylbenzamide, 35 can be prepared by hydrolysis of the methyl ester of 32 by treatment with NaOH in EtOH until hydrolysis is complete followed by treating a DMF solution of resulting acid with CH3NH2.HCI and HATU with DIPEA at 0 °C and allowing the mixture to stir over night at room temperature.
[0179] Step 3. Preparation of 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)-N- methylbenzamide, 36
Figure imgf000061_0001
[0180] 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)benzamide, 36 can be prepared by deoxygenation of a solution of 4-((5-bromoindolin-l-yl)methyl)-N-methylbenzamide, 35, tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 and CS2CO3 in DMF: water 2: 1 by the passage of argon for 20 min followed by the introduction of Pd(PPh3)4 and heating the mixture to 100 °C for 2 h. Treatment of the crude product with HCl in dioxane then can provide 4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)-N- methylbenzamide, 36.
Example 12. Synthesis of V-(l-(4-(l-hydroxycyclobutyl)benzyl)indolin-5-yl), 42
[0181] Reaction step 1. Preparation of l-(/?-tolyl)cyclobutanol, 39
Figure imgf000061_0002
[0182] To a stirred solution of 4-bromotoluene, 37 (5.0 g, 29 mmol) in THF (70 mL) was added 2.5 M n-BuLi (17 mL, 35 mmol) dropwise at -75 °C and the reaction mixture was stirred for 30 min. Cyclobutanone, 38 (2.6 mL, 35 mmol) was added at the same temperature and the reaction mixture was stirred for 1 h. The progress of the reaction was monitored by TLC (TLC system: 10 % EtO Ac/Pet ether, Rf value: 0.25)
[0183] After completion of the reaction, the reaction mixture was quenched with saturated ammonium chloride solution at -70 °C and diluted with water. The aqueous solution was extracted with ethyl acetate (2 x 150 mL). The combined organic layers were dried over anhydrous sodium sulphate and evaporated under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 5 % EtO Ac/Pet ether to give l-(/?-tolyl)cyclobutanol, 39 as a colorless liquid.
[0184] Reaction step 2. Preparation of l-(4-(bromomethyl)phenyl)cyclobutanol, 40
Figure imgf000062_0001
[0185] To stirred solution of l-(/?-tolyl)cyclobutanol, 39 (3.0 g, 18 mmol) in CC14 (50 mL) was added NBS (3.2 g, 18 mmol) and AIBN (0.03 g, 0.18 mmol) at RT. The resulting reaction mixture was heated to reflux overnight. The progress of the reaction was monitored by TLC (TLC system: 10 % EtO Ac/Pet ether, Rf value: 0.2)
[0186] After completion of the reaction, the reaction mixture was cooled, poured into ice cold water and extracted with dichloromethane (2 x 200 mL). The combined organic layers were dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure. The crude product was purified over silica gel (100-200 mesh) column chromatography eluting with 7 % EtO Ac/Pet ether to give l-(4- (bromomethyl)phenyl)cyclobutanol, 40 as a colorless liquid.
[0187] Step 3. Preparation of l-(4-((5-bromoindolin-l-yl)methyl)phenyl)cyclobutanol, 41 1- -(bromomethyl)phenyl)cyclobutanol
Figure imgf000062_0002
[0188] l-(4-((5-bromoindolin-l-yl)methyl)phenyl)cyclobutanol, 41 can be prepared by the alkylation of 5-bromo indoline with l-(4-(bromomethyl)phenyl)cyclobutanol, 40 in the presence of NaH in THF using the procedure described in Example 7, step 1.
[0189] Step 4. Preparation of l-(4-((5-(lH-pyrazol-4-yl)indolin-l- l)methyl)phenyl)cyclobutanol, 42.
Figure imgf000062_0003
[0190] l-(4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)phenyl)cyclobutanol, 42 can be prepared by coupling of l-(4-((5-bromoindolin-l-yl)methyl)phenyl)cyclobutanol, 41 with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate, 10 followed by deprotection by treatment with HC1 in dioxane using the general procedure described in Example 8.
Example 13. Preparation of 5-(lH-pyrazol-4-yl)-l-(4-(l,l,l-trifluoro-2-methylpropan-2- yl)benzyl)indoline
-(l,l,l-trifluoro-2-methylpropan-2-yl)phenyl)methanol, 44.
Figure imgf000063_0001
[0192] (4-(l,l,l-Trifluoro-2-methylpropan-2-yl)phenyl)methanol, 44 can be prepared from l-bromo-4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzene, 43 by palladium catalyzed carbonylation by deoxygenating a solution of l-bromo-4-( 1,1,1 -trifluoro-2-methylpropan-2- yl)benzene, 43, and an excess of Et3N in MeOH DMF 2: 1 by the passage of argon for 20 min contained in an autoclave vessel. Pd(PPh3)4 can be added and the mixture further deoxygenated with argon for 20 min. Passage of CO gas through the reaction mixture, and adjustment of the CO pressure to 150 psi prior to heating to 100 °C overnight can then lead to methyl 4-(l ,l,l-trifluoro-2-methylpropan-2-yl)benzoate, which can be treated with LAH in THF solution to afford, after an aqueous workup, (4-( 1,1,1 -trifluoro-2-methylpropan-2- yl)phenyl)methanol, 44.
[0193] Step 2. Preparation of l-bromo-4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzene, 45
Figure imgf000063_0002
[0194] l-Bromo-4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzene, 45 can be prepared by treatment of a solution of (4-(l,l,l-trifluoro-2-methylpropan-2-yl)phenyl)methanol, 44, in ether with PBr3 at room temperature for 4 h, followed by quenching and aqueous workup.
[0195] Step 3. Preparation of 5-bromo-l-(4-(l,l,l-trifluoro-2-methylpropan-2- yl)benzyl)indoline, 46
Figure imgf000064_0001
[0196] 5-Bromo-l-(4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzyl)indoline, 46 can be prepared from 5-bromoindoline, 22 and l-bromo-4-( 1,1,1 -trifluoro-2-methylpropan-2- yl)benzene, 45 using the general method described in Example 7, step 1.
[0197] Step 4. Preparation of 5-(lH-pyrazol-4-yl)-l-(4-(l,l,l-trifluoro-2-methylpropan-2- yl)benzyl)indoline, 47.
Figure imgf000064_0002
[0198] 5-(lH-Pyrazol-4-yl)- 1 -(4-(l , 1 , 1 -trifluoro-2-methylpropan-2-yl)benzyl)indoline, 47 can be prepared from 5-bromo-l-(4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzyl)indoline, 46 using the general method described in Example 8.
Exam le 14. Preparation of 5-(lH-pyrazol-4-yl)-l-(4-(trifluoromethyl)benzyl)indoline
Figure imgf000064_0003
3. Dioxane, HCI
[0199] 5-(lH-Pyrazol-4-yl)-l-(4-(trifluoromethyl)benzyl)indoline, 48 can be prepared by alkylation of 5-bromoindoline, 22 with l-bromomethyl-4-trifluoromethylbenzene using the conditions described in Example 7, part 1 followed by palladium catalyzed coupling of the alkylation product with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole-l-carboxylate, 10 followed by hydrolysis using the reaction conditions described in Example 8. Example 15. Preparation of l-(4-isopropylbenzyl)-5-(lH-pyrazol-4-yl)indoline
Figure imgf000065_0001
3. Dioxane, HCI
[0200] l-(4-Isopropylbenzyl)-5-(lH-pyrazol-4-yl)indoline, 49 can be prepared by alkylation of 5-bromoindoline, 22 with l-bromomethyl-4-isopropylbenzene using the conditions described in Example 7, part 1 followed by palladium catalyzed coupling of the alkylation product with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l- carboxylate, 10 followed by hydrolysis using the reaction conditions described in Example 8. Example 16. l-(4-(tef"f-butyl)benzyl)-5-(lH-pyrazol-4-yl)indoline, 50
Figure imgf000065_0002
3. Dioxane, HCI
[0201] l-(4-Isopropylbenzyl)-5-(lH-pyrazol-4-yl)indoline, 50 can be prepared by alkylation of 5-bromoindoline, 22 with l-(bromomethyl)-4-(fert-butyl)benzene using the conditions described in Example 7, part 1 followed by palladium catalyzed coupling of the alkylation product with tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole-l- carboxylate, 10 followed by hydrolysis using the reaction conditions described in Example 8. Example 17. Preparation of 2-(4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)phenyl)- l,l,l,3,3,3-hexafluoropropan-2-ol
[0202] Step 1. Preparation of 5-bromoindoline, 29.
Figure imgf000065_0003
28 22 [0203] NaCNBH3 (213.2 g, 3.38 mol) was added portion wise to a stirred solution of 5- bromoindole (200.0 g, 1.03 mol) in acetic acid (2.0 L) at 10 °C and the mixture was stirred for 2 h. The progress of the reaction was monitored by TLC (TLC system: 20 % EtO Ac/pet ether, Rf value: 0.4).
[0204] After completion of the reaction, the reaction mixture was evaporated under reduced pressure. The crude product was basified with 10% NaOH solution and extracted with ethyl acetate (2 x 1.0 L). The combined organic layers were dried over sodium sulfate and evaporated under reduced pressure. The crude compound was purified over silica gel (100- 200 mesh) column chromatography by eluting with 10 % EtO Ac/Pet ether to afford 5- bromoindoline, 22 as a thick yellow liquid. 1H NMR (400 MHz, CDC13) δ: 7.18 (s, 1 H), 7.08 (d, J= 8.0 Hz, 1H), 6.48 (d, J= 8.4 Hz, 1 H), 3.55 (t, J= 8.4 Hz, 2 H), 3.01 (t, J= 8.4 Hz, 2 H). LC-MS: 96.26 % (m/z = 198.1, [M+H]+).
[0205] Step 2. Preparation of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3, 3,3- hexafluoropropan-2-ol, 23.
Figure imgf000066_0001
[0206] 60%) NaH (60.0 g, 1.52 mol) was added portion wise to a stirred solution of 2-(4- (bromomethyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol (221.7 g, 1.3 mol) and 2- bromoindoline (100.0 g, 0.507 mol) in THF (1.0 L) at 0 °C. The resulting reaction mixture was heated to 80 °C for 3 h. The progress of the reaction was monitored by TLC (TLC system: 30 % EtO Ac / Pet ether, Rf value: 0.65).
[0207] The reaction mixture was quenched with ice cold water and extracted with ethyl acetate (2 x 1.0 L). The combined organic layers were washed with water and brine. The organic layer was dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. The crude compound was purified over silica gel (230-400 mesh) column chromatography eluting with 5 % EtO Ac/Pet ether to afford 2-(4-((5-bromoindolin-l- yl)methyl)phenyl)-l,l,l,3,3,3-hexafluoropropan-2-ol, 23 as a thick yellow liquid. 1H NMR
(400 MHz, CDC13) δ: 7.67 (d, J= 8.0 Hz, 2H), 7.42 (d, J= 8.0 Hz, 2H), 7.17 (s, 1H), 7.12 (d, J= 8.4 Hz, IH), 6.31 (d, J= 8.0 Hz, IH), 4.25 (s, 2H), 3.35 (t, J= 8.0 Hz, 2H), 2.98 (t, J= 8.0 Hz, 2H); LC-MS: 99.28 % {m/z = 454.1, [M+H]+).
[0208] Step 3. Preparation of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3,3,3- hexafluoro ropan-2-ol, 23
Figure imgf000067_0001
[0209] To a stirred solution of 2-(4-((5-bromoindolin-l-yl)methyl)phenyl)-l, 1,1,3, 3,3- hexafluoropropan-2-ol, 23 (25.0 g, 55.2 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate 10 (21.08 g, 71.7 mmol) in DMF (250 mL) and water (75 mL) was added CS2CO3 (53.9 g, 166 mmol) under a nitrogen atmosphere. The reaction mixture was de -oxygenated by purging argon gas into the reaction mixture for 30 min. PdCl2(dppf). dichloromethane (4.5 g, 5.5 mmol) was added to the reaction mixture and the mixture was heated to 100 °C overnight. The progress of the reaction was monitored by TLC (TLC system: 40 % EtOAc/Pet ether, Rf value: 0.01).
[0210] After completion of the reaction, the reaction mixture was filtered through a celite bed. The filtrate was extracted with ethyl acetate (2 x 500 mL). The combined organic layers were washed with brine and dried over anhydrous sodium sulfate. The solvent was removed under reduced pressure to afford crude compound. This procedure was repeated on an additional 3 x 25 g batches and the crude products were combined for purification. The combined crude product was purified by chromatography over neutral alumina eluting with 50 % EtOAc/Pet ether to afford 2-(4-((5-(lH-pyrazol-4-yl)indolin-l-yl)methyl)phenyl)- 1,1,1, 3,3, 3-hexafluoropropan-2-ol, 27, mp 191-195 °C after crystallization from hot ethyl acetate and diethyl ether. 1H NMR (400 MHz, CDC13) δ: 12.68 (bs, IH), 8.63 (s, IH), 7.95 (s, IH), 7.66 (d, J= 8.0 Hz, 2H), 7.50 (d, J= 8.8 Hz, 2H), 7.29 (s, IH), 7.21 (dd, J;= 8.0 Hz, j2=1.6 Hz, IH), 6.55 (d, J= 8.0 Hz, IH), 4.32 (s, 2H), 3.30 (t, J= 8.4 Hz, 2H), 2.93 (t, J= 8.4 Hz, 2H). LC-MS: 98.05 % at 215 nm and 92.29 % at 254 nm (m/z = 441.8, [M+H]+).
Biological Examples [0211] The compounds according to Formula 1 are RORa and/or RORy modulators, and are useful in the treatment of RORa- and/or RORy-regulated diseases. The biological activities of the compounds according to Formula 1 can be determined using any suitable assay for determining the activity of a candidate compound as a RORa and/or RORy modulator, as well as in tissue and in vivo models.
Example 18. TH17 cell differentiation.
[0212] Peripheral blood mononuclear cells (PBMCs) were sourced from freshly prepared leukocyte enriched plasma (buffy coat) from healthy volunteers from which PBMCs were isolated by density gradient centrifugation using Ficoll-Paque™ PLUS (GE Healthcare). Naive CD4+ T cells were isolated using the Na'ive CD4+ T cell Isolation Kit II (Miltenyi Biotec) as per manufacturer's instructions. CD4+ T cells were plated at 5 x 104 cells/well in 96 well plates pre-coated with immobilized human (h)-aCD3 antibody (eBioscience) then differentiated to TH17 cells using h-aCD28, h-aIFN-y, h-aIL-4, h-IL-6, h-IL-23, h-IL-Ιβ, h- TGF-βΙ (eBioscience) and maintained in a cell culture incubator at 37°C with 5% C02 in TexMACS Medium (Miltenyi Biotec) supplemented with 1% Penicillin/Streptomycin (Lonza) over 5 days.
Example 19. TH17 Cytokine Inhibition as measured by ELISA.
[0213] Human CD4+ T cells were differentiated to TH17 cells as described in the presence or absence of various concentrations of compounds in a final concentration of 0.1% DMSO in TexMACS medium. Supernatants were collected and stored at -20 C until they were to be assayed for IL-17A, IL-17F, IL-17AF, IL-21 and/or IL-22 levels as determined by Ready- Set-Go ELISA kits (eBioscience) as per manufacturer's instructions. Endpoint absorbance was read at 450 nm and 570 nm according to manufacturer's instructions (eBioscience) using a microplate reader (Perkin Elmer). The percentage of cytokine inhibition was calculated with reference to DMSO treated cells and the IC50S were determined by GraphPad Prism software and presented in the table below.
Figure imgf000068_0001
3 <10 <10 <10 <10 <10
4 <10 <10 <10
5 <10 <10 <10 <10 <10
6 <10 >10 <10
7 <10 <10 <10 <10 <10
8 <10 <10 <10 <10 <10
9 <10 <10 >10 <10 <10
Example 20. TH17 Cell Viability and Proliferation
[0214] Cells were assayed for viability in the presence or absence of 1 μΜ compound using Prestoblue® Cell Viability Reagent (Life Technologies) as per manufacturer's instructions after 30 minutes incubation of TR17 cells with reagent. Absorbance was measured at 570nm and 600nm using a microplate reader (Perkin Elmer) and data were analyzed as per manufacturer's instructions. Cell viability was determined after 5 days and expressed as a percentage of DMSO treated cells. The percentage (%) cell viability of compound treated TH17 cells are presented in the table below.
Figure imgf000069_0001
[0215] Cellular proliferation was assessed in the presence or absence of 3μΜ compound using Prestoblue® Cell Viability Reagent (Life Technologies) as per manufacturer's instructions using absorbance as the output measured by a microplate reader (Perkin Elmer) after 30 minutes of incubation. A standard curve developed by plotting varying cell numbers versus absorbance was used to assess differences in cellular proliferation between compound treated or untreated cells after the 5 day TR17 differentiation period. Percentage (%) change in TR17 cell number in the presence of compound are presented in the table below.
Figure imgf000070_0001
A: >50% decrease in cell proliferation relative to DMSO treated TR17 cells
B: < 50% decrease in cell proliferation relative to DMSO treated TR17 cells
Example 21. Real Time quantitative Reverse Transcription PCR (Real-Time qRT- PCR).
[0216] RNA was isolated from TH17 cells and cDNA was synthesized using the Verso cDNA kit (Thermo Scientific) as per manufacturer's instructions. IL-17A, IL-17F, IL-21, IL-22, G- CSF, IL-23R and FOXP3 gene expression were determined for compound treated and untreated TH17 cell samples using Solaris qPCR Gene expression assays (Thermo Scientific) with an iCycler iQ Real-Time PCR Detection System (Bio-Rad). Individual gene expression was normalized to a panel of six reference genes (ACTB, TBP, HPRTl, PGKl, TFRC and PPIA) (Thermo Scientific) and analyzed by REST 2009 (Qiagen) in accordance to MIQE guidelines (Bustin et al. (2009) Clin.Chem, 55:611-622). The effects of compounds on TH17 cytokine gene expression are presented as fold change in mRNA levels of compound treated TR17 cells relative to DMSO treated cells in the table below. Fold change in mRNA levels of compound treated ΤΉ17 cells relative
to DMSO treated cells
Compound
from IL-17A IL-17F IL-22 IL-21 IL-23R FOXP3
Example
1 A B B B A C
2 B B B B A C
3 B B B B A A
4 B B B B A A
5 A A A A A A
6 B B B A C C
A: No change in gene expression observed relative to DMSO treated Thl7 cells
B: Decreased gene expression observed relative to DMSO treated Thl7 cells
C: Increased gene expression observed relative to DMSO treated Thl7 cells Example 22. RORy LBD Binding.
[0217] Compound binding to the human RORy LBD was assessed using the Human RORy Assay System (INDIGO Biosciences Inc.) as per manufacturer's instructions. Binding to the RORy LBD was assessed using gradient concentrations of the indicated compounds in a final concentration of 0.5% DMSO, where IC50S were generated using Graphpad Prism and presented in the table below.
Figure imgf000071_0001
Figure imgf000072_0001
Example 23. Co-activator and Co-repressor Protein Interaction with Human ROR and RORa LBDs.
[0218] The Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) assay kit (Life Technologies) was used to measure compound efficacy to disrupt or enhance ligand- mediated interaction of the co-activator and co-repressor peptides with the purified GST- tagged (i) RORy LBD or (ii) RORa LBD. The RORy LBD and RORa LBD each have a basal level of interaction with the co-activator D22 or co-repressor NCoRl-D2 in the absence of ligand, thus it is possible to identify compounds that inhibit or induce RORy and/or RORa co-activator/co-repressor interactions, thus determining RORa, RORy, or RORa/RORy compound selectivity. The assay was performed as per manufacturer's instructions and TR- FRET was assessed 2 and 20 hours post-assay initiation. Compounds were identified to (i) have no impact on either co-activator displacement or co-activator recruitment from the assessed LBD (ii) promote co-activator displacement from the assessed LBD (iii) promote co-repressor recruitment to the assessed LBD or (iv) concurrently promote co-activator displacement from the assessed LBD and co-repressor recruitment to the assessed LBD. The EC50 potency of compounds tested for co-activator displacement and co-repressor recruitment to RORy and RORa are presented in the table below.
Figure imgf000072_0002
7 <30 <30 <30 <30 <30 <30 >30 <30
8 <30 <30 <30 >30 >30 <30 <30 >30
9 >30 >30 <30 >30 >30 <30 >30 <30
Δ - Values represent EC50 [μΜ] of D22 co-activator displacement from the respective ROR
LBD
& - Values represent EC50 [μΜ] of NCoRl-D2 co-repressor recruitment to the respective ROR LBD
[0219] The present invention is not to be limited in scope by the specific embodiments disclosed in the examples which are intended as illustrations of a few aspects of the invention and any embodiments that are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art and are intended to fall within the scope of the appended claims. All patents, patent applications, and publications cited herein are hereby incorporated by reference in their entireties.
EQUIVALENTS
[0220] Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.

Claims

1. A compound of Formula I :
Figure imgf000074_0001
I wherein
A is aryl or heteroaryloptionally substituted with C1-C4 alkyl, halogen, OH, CN, or N(R4)2;
Ri is absent, H, OH, halogen, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Each R5 is independently H or alkyl, or both R5 can be taken together to form a carbonyl;
Y is 5-7 membered heteroaromatic ring linked via a carbon atom and optionally substituted; n is 0, or 1; o is 0 or 1 ; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof;
2. The compound of claim 1 , wherein Y is selected from substituted or unsubstituted thienyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted isoxazolyl, substituted or unsubstituted isothiazolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted pyrazolyl, substituted or unsubstituted oxadiazolyl, substituted or unsubstituted thiadiazolyl, substituted or unsubstituted triazolyl, substituted or unsubstituted tetrazolyl, substituted or unsubstituted pyridinyl, substituted or unsubstituted pyrimidinyl, substituted or unsubstituted pyrazinyl, substituted or unsubstituted pyridazinyl, and substituted or unsubstituted triazinyl.
The compound of claim 2, wherein Y is substituted or unsubstituted pyrazolyl. The compound of claim 2, wherein Y is substituted or unsubstituted triazolyl. The compound of claim 2, wherein Y is substituted or unsubstituted oxadiazolyl. The compound of claim 2, wherein Y is substituted or unsubstituted pyrimidinyl. The compound of claim 2, wherein Y is substituted or unsubstituted pyridinyl. The compound of claim 2, wherein Y is substituted or unsubstituted pyrazinyl. The compound of claim 1, wherein the compound is selected from the group consisting of:
Figure imgf000076_0001
-75-
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
-78-
Figure imgf000080_0001
-79-
Figure imgf000081_0001
Figure imgf000081_0002
Figure imgf000081_0003
wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH,
Figure imgf000082_0001
Ri is absent, H, OH, halogen, N(R )2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
11. The compound of claim 10, wherein the compound is selected from the group consisting of:
Figure imgf000083_0001
Figure imgf000083_0002
Figure imgf000083_0003
-82-
Figure imgf000084_0001
2. A compound of formula lb:
Figure imgf000085_0001
lb wherein
A is aryl or heteroaryl is optionally substituted with C1-C4 alkyl, halogen,
Figure imgf000085_0002
Ri is absent, H, OH, halogen, N(R )2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R-i)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating an 0-3 atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
13. The compound of claim 12, wherein the compound is selected from the group consisting of:
Figure imgf000086_0001
-85-
Figure imgf000087_0001
Figure imgf000087_0002
Figure imgf000087_0003
Ic wherein
A is aryl or heteroaryl; optionally substituted with C1-C4 alkyl, halogen, OH, CN, and N(R4)2; Ri is absent, H, OH, halogem, N(R4)2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H, halogen, or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two R4 can be taken together to form a 4-7 membered ring, optionally incorporating 0-3 N, O, S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; and p is 1 or 2; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
The compound of claim 14, wherein the compound is selected from the group
Figure imgf000088_0001
16. A compound of formula Id:
Figure imgf000088_0002
Id wherein
A is aryl or heteroaryl optionally substituted with C1-C4 alkyl, halogen, OH, CN, and N(R4)2;
Ri is absent, H, OH, halogen, N(R )2, or C1-C4 alkyl optionally substituted with halogen, OH, O-alkyl, CN, or N(R4)2;
R2 and R3 are independently selected from H or C1-C4 alkyl optionally substituted with halogen, OH, CN, N(R4)2, or R2 and R3, taken together may form a 4-7 membered saturated or unsaturated ring, optionally incorporating 0-3 N, O, or S atoms; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, carbonyl, CN, halogen, or N(R4)2, or R2 and R3, can be taken together to form a carbonyl;
Each R4 is independently H, alkyl, or aryl, or two can be taken together to form a 4-7 membered ring, optionally incorporating an additional N, O, S atom; wherein the 4-7 membered ring is optionally substituted with C1-C4 alkyl, halogen, OH, CN, halogen, or N(R4)2;
Y is 5-7 membered heteroaryl linked via a carbon atom and optionally substituted; p is 1 or 2; and wherein the symbol indicates a single or double bond; or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, and tautomer thereof.
17. The compound of claim 14, wherein the compound is selected from the group consisting of:
Figure imgf000090_0001
18. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier.
19. A method of treating any disease described herein comprising administering an effective amount of a compound of claim 1.
20. The method claim 19, wherein the disease is selcted from the group consisting of angina pectoris, myocardial infarction, atherosclerosis, cystic fibrosis, gastritis, autoimmune myositis, giant cell arteritis, Wegener's granulomatosis, asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, juvenile rheumatoid arthritis, allergen-induced lung inflammation, allergy, psoriasis, psoriatic arthritis, colitis, inflammatory bowel disease,
Crohn's disease, ulcerative colitis, Sjogren's syndrome, dry eye, optic neuritis, neuromyelitis optica, myasthenia gravis, Guillain-Barre syndrome, Graves disease, multiple sclerosis, autoimmune uveitis, ankylosing spondylitis, organ transplant rejection, polymyalgia rheumatic, systemic lupus erythematosus, cutaneous lupus, lupus nephritis, glomerulonephritis, diabetes mellitus type 1 , pulmonary inflammation, macular degeneration, obesity, non-alcoholic fatty liver disease, steatohepatitis, insulin resistance, diabetes mellitus type 2, glucose intolerance, and metabolic syndrome.
PCT/US2013/038867 2012-04-30 2013-04-30 Ror modulators and their uses WO2013166015A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/394,787 US9447069B2 (en) 2012-04-30 2013-04-30 ROR modulators and their uses
EP13785182.0A EP2844260A4 (en) 2012-04-30 2013-04-30 Ror modulators and their uses

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201261640401P 2012-04-30 2012-04-30
US61/640,401 2012-04-30
US201261736895P 2012-12-13 2012-12-13
US61/736,895 2012-12-13
US201361779856P 2013-03-13 2013-03-13
US61/779,856 2013-03-13

Publications (1)

Publication Number Publication Date
WO2013166015A1 true WO2013166015A1 (en) 2013-11-07

Family

ID=49514807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/038867 WO2013166015A1 (en) 2012-04-30 2013-04-30 Ror modulators and their uses

Country Status (3)

Country Link
US (1) US9447069B2 (en)
EP (1) EP2844260A4 (en)
WO (1) WO2013166015A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015112801A1 (en) * 2014-01-24 2015-07-30 Purdue Pharma L.P. Pyridines and pyrimidines and use thereof
WO2015138280A3 (en) * 2014-03-10 2015-11-26 Innov17 Llc Retinoic acid receptor-related orphan receptor modulators and uses thereof
WO2016066597A1 (en) * 2014-10-31 2016-05-06 F. Hoffmann-La Roche Ag New dihydroquinoline pyrazolyl compounds
JP2017502057A (en) * 2014-01-09 2017-01-19 オリオン コーポレーション Bicyclic heterocyclic derivatives as bromodomain inhibitors
EP3116504A4 (en) * 2014-03-10 2017-08-09 Innov17 LLC Retinoic acid receptor-related orphan receptor modulators and uses thereof
EP3172195A4 (en) * 2014-07-25 2018-03-21 Innov17 LLC Sulfonamide retinoic acid receptor-related orphan receptor modulators and uses thereof
US10011566B2 (en) 2015-12-15 2018-07-03 Astrazeneca Ab Compounds
US11034654B2 (en) 2017-06-14 2021-06-15 Astrazeneca Ab 2,3-dihydroisoindole-1-carboxamides useful as ROR-gamma modulators

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
US5739338A (en) * 1996-11-05 1998-04-14 Allergan N-aryl substituted tetrahydroquinolines having retinoid agonist, retinoid antagonist or retinoid inverse agonist type biological activity
US20040265809A1 (en) * 2001-05-07 2004-12-30 Dino Moras Polypeptides derived from retinoic acid-related orphan receptor(ror) and their applications
US20050009870A1 (en) * 2003-07-11 2005-01-13 Sher Philip M. Tetrahydroquinoline derivatives as cannabinoid receptor modulators
US7812017B2 (en) * 2006-07-03 2010-10-12 Biovitrum Ab (Publ.) 4-substituted indole and indoline compounds
WO2011112263A1 (en) * 2010-03-11 2011-09-15 New York University AMIDO COMPOUNDS AS RORγt MODULATORS AND USES THEREOF
WO2012064744A2 (en) * 2010-11-08 2012-05-18 Lycera Corporation Tetrahydroquinoline and related bicyclic compounds for inhibition of rorϒ activity and the treatment of disease

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4581356A (en) * 1983-03-22 1986-04-08 Fujisawa Pharmaceutical Co., Ltd. Triazine derivatives, and pharmaceutical compositions comprising the same
ES2288195T3 (en) * 2001-09-14 2008-01-01 Novo Nordisk A/S NEW LINKS FOR HISB10 ZN2 + SITES OF THE INSULIN HEXAMERO IN THE CONFIGURATION R.
TW200500362A (en) * 2003-04-09 2005-01-01 Japan Tobacco Inc 5-membered heteroaromatic ring compound and pharmaceutical use thereof
WO2006082245A1 (en) * 2005-02-07 2006-08-10 Novo Nordisk A/S Pharmaceutical preparations comprising insulin, zinc ions and a zinc-binding ligand

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
US5739338A (en) * 1996-11-05 1998-04-14 Allergan N-aryl substituted tetrahydroquinolines having retinoid agonist, retinoid antagonist or retinoid inverse agonist type biological activity
US20040265809A1 (en) * 2001-05-07 2004-12-30 Dino Moras Polypeptides derived from retinoic acid-related orphan receptor(ror) and their applications
US20050009870A1 (en) * 2003-07-11 2005-01-13 Sher Philip M. Tetrahydroquinoline derivatives as cannabinoid receptor modulators
US7812017B2 (en) * 2006-07-03 2010-10-12 Biovitrum Ab (Publ.) 4-substituted indole and indoline compounds
WO2011112263A1 (en) * 2010-03-11 2011-09-15 New York University AMIDO COMPOUNDS AS RORγt MODULATORS AND USES THEREOF
WO2012064744A2 (en) * 2010-11-08 2012-05-18 Lycera Corporation Tetrahydroquinoline and related bicyclic compounds for inhibition of rorϒ activity and the treatment of disease

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
BARDER, T. E. ET AL., J. AM. CHEM. SOC., vol. 127, 2005, pages 4685 - 4696
BURNS ET AL., PHARMACOL.REV., vol. 65, 2013, pages 710 - 778
BURRIS ET AL., CHEM.BIOL., vol. 19, 2012, pages 51 - 59
BUSTIN ET AL., CLIN.CHEM, vol. 55, 2009, pages 611 - 622
CUA; TATO, NAT.REV.IMMUNOL., vol. 10, 2010, pages 479 - 489
DVORAK, C.A. ET AL., JOURNAL OF ORGANIC CHEMISTRY, vol. 70, 2005, pages 4188 - 4190
EDWARDS ET AL., J.NEUROL., vol. 258, 2011, pages 1518 - 1527
FUITA, K.-I. ET AL., ORGANIC LETT., vol. 6, 2004, pages 2785
GHORESCHI ET AL., NATURE, vol. 467, 2010, pages 967 - 971
GOODMAN, L. S.; GILMAN, A.: "The Pharmacological Basis of Therapeutics, 5th ed.", 1975, MACMILLAN: NEW YORK, pages: 201 - 226
HUH; LITTMAN, EUR.J.IMMUNOL., vol. 42, 2012, pages 2232 - 2237
IVANOV ET AL., CELL, vol. 126, 2006, pages 1121 - 1133
JETTEN ET AL., FRONT ENDOCRINOL.(LAUSANNE), vol. 4, 2013, pages 1
JETTEN, NUCL.RECEPT.SIGNAL., vol. 7, 2009, pages E003
JETTEN; JOO, ADV.DEV.BIOL., vol. 16, 2006, pages 313 - 355
LEE ET AL., NAT.IMMUNOL., vol. 13, 2012, pages 991 - 999
LITTMAN; RUDENSKY, CELL, vol. 140, 2010, pages 845 - 858
MIOSSEC ET AL., N.ENGL.J.MED., vol. 361, 2009, pages 888 - 898
MIOSSEC; KOLLS, NAT.REV.DRUG DISCOV., vol. 11, 2012, pages 763 - 776
N. KUDO ET AL., ANGEW. CHEM. INT. ED., vol. 45, 2006, pages 1282 - 1284
PETER G. M. WUTS; T. W. GREENE: "Greene 's Protective Croups in Organic Synthesis, 4th ed.", 2007, WILEY
See also references of EP2844260A4 *
SPITS; DI SANTO, NAT.IMMUNOL., vol. 12, 2011, pages 21 - 27
SUTTON ET AL., EUR.J.IMMUNOL., vol. 42, 2012, pages 2221 - 2231
ZEPP ET AL., TRENDS IMMUNOL., vol. 32, 2011, pages 232 - 239

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2017502057A (en) * 2014-01-09 2017-01-19 オリオン コーポレーション Bicyclic heterocyclic derivatives as bromodomain inhibitors
WO2015112801A1 (en) * 2014-01-24 2015-07-30 Purdue Pharma L.P. Pyridines and pyrimidines and use thereof
US10047075B2 (en) 2014-01-24 2018-08-14 Purdue Pharma L.P. Pyridines and pyrimidines and use thereof
JP2017503837A (en) * 2014-01-24 2017-02-02 パーデュー、ファーマ、リミテッド、パートナーシップ Pyridines and pyrimidines and uses thereof
WO2015138280A3 (en) * 2014-03-10 2015-11-26 Innov17 Llc Retinoic acid receptor-related orphan receptor modulators and uses thereof
EP3116866A4 (en) * 2014-03-10 2017-07-26 Innov17 LLC Retinoic acid receptor-related orphan receptor modulators and uses thereof
EP3116504A4 (en) * 2014-03-10 2017-08-09 Innov17 LLC Retinoic acid receptor-related orphan receptor modulators and uses thereof
EP3172195A4 (en) * 2014-07-25 2018-03-21 Innov17 LLC Sulfonamide retinoic acid receptor-related orphan receptor modulators and uses thereof
US9790200B2 (en) 2014-10-31 2017-10-17 Hoffmann-La Roche Inc. Dihydroquinoline pyrazolyl compounds
CN106795138A (en) * 2014-10-31 2017-05-31 豪夫迈·罗氏有限公司 New EEDQ pyrazolyl compounds
WO2016066597A1 (en) * 2014-10-31 2016-05-06 F. Hoffmann-La Roche Ag New dihydroquinoline pyrazolyl compounds
CN106795138B (en) * 2014-10-31 2020-04-14 豪夫迈·罗氏有限公司 Novel dihydroquinoline pyrazolyl compound
US10011566B2 (en) 2015-12-15 2018-07-03 Astrazeneca Ab Compounds
US10526286B2 (en) 2015-12-15 2020-01-07 Astrazeneca Ab Compounds
US10988445B2 (en) 2015-12-15 2021-04-27 Astrazeneca Ab Compounds
US11453644B1 (en) 2015-12-15 2022-09-27 Astrazeneca, Ab Compounds
US11034654B2 (en) 2017-06-14 2021-06-15 Astrazeneca Ab 2,3-dihydroisoindole-1-carboxamides useful as ROR-gamma modulators

Also Published As

Publication number Publication date
US9447069B2 (en) 2016-09-20
US20150099779A1 (en) 2015-04-09
EP2844260A4 (en) 2016-06-29
EP2844260A1 (en) 2015-03-11

Similar Documents

Publication Publication Date Title
US9708268B2 (en) ROR modulators and their uses
US9321750B2 (en) ROR modulators and their uses
US9447069B2 (en) ROR modulators and their uses
US20210292285A1 (en) Heterocyclic compounds as adenosine antagonists
JP6750806B2 (en) Substituted heterocyclyl derivatives as CDK inhibitors
US9050334B2 (en) MIF inhibitors and their uses
US9133164B2 (en) MIF inhibitors and their uses
JP5735987B2 (en) Nitrogen-containing heteroaryl derivatives
JP2019511466A (en) Halo substituted piperidines as orexin receptor modulators
US9771374B2 (en) Benzimidazole retinoic acid receptor-related orphan receptor modulators and uses thereof
US9701663B2 (en) Sulfonamide retinoic acid receptor-related orphan receptor modulators and uses thereof
US20150252022A1 (en) Retinoic acid receptor-related orphan receptor modulators and uses thereof
US9884847B2 (en) Indazole retinoic acid receptor-related orphan receptor modulators and uses thereof
US20150252051A1 (en) Retinoic acid receptor-related orphan receptor modulators and uses thereof
US20170298060A1 (en) Pyrrolopyridine retinoic acid receptor-related orphan receptor modulators and uses thereof
US20170217956A1 (en) Azaindole retinoic acid receptor-related orphan receptor modulators and uses thereof

Legal Events

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

Ref document number: 13785182

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14394787

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013785182

Country of ref document: EP