NZ722413B2 - Dihydropyrrolopyridine inhibitors of ror-gamma - Google Patents

Dihydropyrrolopyridine inhibitors of ror-gamma Download PDF

Info

Publication number
NZ722413B2
NZ722413B2 NZ722413A NZ72241315A NZ722413B2 NZ 722413 B2 NZ722413 B2 NZ 722413B2 NZ 722413 A NZ722413 A NZ 722413A NZ 72241315 A NZ72241315 A NZ 72241315A NZ 722413 B2 NZ722413 B2 NZ 722413B2
Authority
NZ
New Zealand
Prior art keywords
alkyl
halo
compound
alkoxy
hydroxy
Prior art date
Application number
NZ722413A
Other versions
NZ722413A (en
Inventor
David A Claremon
Lawrence Wayne Dillard
Chengguo Dong
Yi Fan
Lanqi Jia
Zhijie Liu
Stephen D Lotesta
Andrew Marcus
Suresh B Singh
Colin M Tice
Original Assignee
Vitae Pharmaceuticals Llc
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 Vitae Pharmaceuticals Llc filed Critical Vitae Pharmaceuticals Llc
Priority claimed from PCT/US2015/013699 external-priority patent/WO2015116904A1/en
Publication of NZ722413A publication Critical patent/NZ722413A/en
Publication of NZ722413B2 publication Critical patent/NZ722413B2/en

Links

Abstract

Provided are novel compounds of Formula (I): pharmaceutically acceptable salts thereof, and pharmaceutical compositions thereof, which are useful in the treatment of diseases and disorders mediated by RORy. Also provided are pharmaceutical compositions comprising the novel compounds of Formula (I) and methods for their use in treating one or more inflammatory, metabolic, autoimmune and other diseases or disorders. nd methods for their use in treating one or more inflammatory, metabolic, autoimmune and other diseases or disorders.

Description

(12) Granted patent specificaon (19) NZ (11) 722413 (13) B2 (47) Publicaon date: 2021.12.24 (54) DIHYDROPYRROLOPYRIDINE INHIBITORS OF ROR-GAMMA (51) Internaonal Patent Classificaon(s): C07D 471/04 A61K 31/437 A61P 29/00 (22) Filing date: (73) Owner(s): 2015.01.30 VITAE PHARMACEUTICALS, LLC (23) Complete caon filing date: (74) Contact: 2015.01.30 Pizzeys Patent and Trade Mark Attorneys P ty Ltd (30) Internaonal Priority Data: US 61/970,637 2014.03.26 (72) Inventor(s): US 61/935,162 2014.02.03 DONG, Chengguo FAN, Yi (86) Internaonal Applicaon No.: SINGH, Suresh, B.
ZHENG, Yajun ZHUANG, Linghang (87) Internaonal Publicaon : DILLARD, Lawrence, Wayne WO/2015/116904 JIA, Lanqi YUAN, Jing CLAREMON, David, A.
LIU, Zhijie A, Stephen, D.
, Andrew TICE, Colin, M.
ZHAO, Wei (57) Abstract: Provided are novel compounds of Formula (I): pharmaceucally acceptable salts f, and pharmaceucal composions thereof, which are useful in the treatment of diseases and disorders mediated by RORy. Also provided are pharmaceucal composions comprising the novel compounds of Formula (I) and methods for their use in treang one or more inflammatory, B2 metabolic, autoimmune and other diseases or disorders. 722413 DIHYDROPYRROLOPYRIDINE INHIBITORS OF ROR-GAMMA RELATED ATIONS This application claims the benefit of the filing date of US. Provisional Application No. ,162, filed February 3, 2014, and US. Provisional Application No. 61/970,637, filed March 26, 2014. The entire contents of the aformentioned ations are incorporated by reference .
TECHNICAL FIELD OF THE INVENTION The present invention is directed to novel ic acid receptor—related orphan receptor gamma ” or “ROR—gamma”) inhibitors, processes for their preparation, pharmaceutical compositions containing these inhibitors, and their use in the treatment of inflammatory, metabolic, autoimmune and other diseases mediated by RORy.
BACKGROUND OF THE INVENTION Retinoic acid receptor—related orphan receptors (RORs) are a subfamily of transcription factors in the steroid hormone nuclear receptor superfamily (Jetten & J00 (2006) Adv. Dev. Biol. 2006, 16, 313—355). The ROR family consists of ROR alpha (ROROL), ROR beta (RORB) and ROR gamma (RORy), each encoded by a separate gene (in human: RORA, RORB and RORC, respectively; in mouse: ram, rorb and rorc, respectively). RORs contain four principal domains shared by the ty of nuclear receptors: an N—terminal domain, a highly conserved DNA—binding domain (DBD) ting of two zinc finger motifs, a hinge domain, and a ligand binding domain (LBD). Each ROR gene generates several isoforms, differing only in their N—terminal domains. RORY has two isoforms: RORyl and RORy2 (also known as RORyt). RORY refers to RORyl and/or RORyt. RORyl is expressed in a variety of tissues including thymus, muscle, kidney and liver, but RORyt is exclusively expressed in the cells of the immune system, has a critical role in thymopoiesis and the development of several ary lymphoid tissues, and is a key regulator of Thl7 cell differentiation (Jetten, 2009, Nucl. Recept.
Signal., 7:e003, doi:10.1621/nrs.07003, Epub 2009 Apr 3).
Thl7 cells are a subset of T helper cells which entially produce the pro— inflammatory cytokines IL—17A, IL—17F, IL—21 and IL—22. Th17 cells and their effector molecules, such as IL—17, IL—21, IL—22, GM—CSF and CCL20, are associated with the pathogenesis of several autoimmune and inflammatory diseases, such as rheumatoid arthritis, systemic lupus erythematosus, multiple sis, sis, inflammatory bowel disease, allergy and asthma (Maddur et al., 2012, Am. J. Pathol., 181:8—18). Recent findings t a role for IL17 and Th17 cells in the pathogenesis of acne (Thiboutot et al., 2014, J. Invest. Dermatol., WO 16904 2015/013699 134(2):307—10, doi: 10.1038/jid.2013.400; Agak et al., 2014, J. Invest. Dermatol., 134(2):366— 73, doi: 10.1038/jid.2013.334, Epub 2013 Aug 7). Th17 cells are also potent inducers of inflammation associated with triosis, a c inflammatory disease (Hirata et al., 2010, Endocrinol., 68—5476; Hirata et al., 2011, Fertil Steril., (1):113—7, doi: 6/j.fertnstert.2011.04.060, Epub 2011 May 20). Additionally, Th17 cells have a key role in the mouse mune models of experimental autoimmune encephalomyelitis (EAE), collagen—induced arthritis (CIA) and adjuvant—induced arthritis (AIA) (Bedoya et al., 2013, Clin.
Dev. Immunol., 2013:986789. Epub 2013 Dec 26. Th17 cells are activated during inflammatory and autoimmune disease processes and are responsible for recruiting other inflammatory cell types, particularly neutrophils, to mediate pathology in target tissues (Miossec & Kolls, 2012, Nature Rev., 11:763—776; Korn et al., 2009, Annu. Rev. Immunol., 27:485—517). Aberrant Th17 cell function has been ated in a variety of autoimmune diseases, including multiple sclerosis and rheumatoid arthritis. mune disease is believed to arise from the tion of the equilibrium between effector and regulatory T cells (Solt et al., 2012, ACS Chem. Biol., 7: 1515—1519, Epub 2012 July 9). The importance of RORyt to Th17 cell differentiation and the pathogenic role of Th17 cells is evidenced by the fact that RORyt—deficient mice have very few Th17 cells and have a ion in severity of EAE (Ivanov et al., 2006, Cell, 126:1121—1133).
Circadian rhythms are daily cycles of behavioral and physiological changes that are regulated by endogenous circadian clocks. A number of studies have established links between nuclear receptor (including RORy) function and expression, the circadian regulatory circuitry, and the regulation of s physiological processes (Jetten (2009) op. cit).
Obstructive sleep apnea syndrome (OSAS) is a chronic inflammatory disease regulated by T lymphocytes. OSAS patients have a significant increase in eral Th17 cell frequency, IL—17 and RORyt levels (Ye et al., 2012, Mediators Inflamm., 815308, doi: .1155/2012/815308, Epub 2012 Dec 31).
A number of studies have provided evidence of a role of RORs in cancer. Mice deficient in the expression of RORY exhibit a high incidence of thymic lymphomas that metastasize frequently to liver and spleen. High expression of ssociated genes (including RORy) and high levels of Th17 cells in the tumor microenvironment has been shown to correlate with a poor prognosis in various cancers, including lung, gastric, breast and colon cancer (Tosolini et al., 2011, Cancer Res., 71: 1263-1271, doi: 10.1158/0008-5472.CAN-10—2907, Epub 2011 Feb 8; Su et al., 2014, Immunol. Res., 58:118-124, doi: 10.1007/s120268483-y, Epub 2014 Jan 9; Carmi et al., 2011, J. Immunol., 186:3462-3471, doi: 10.4049/jimmunol.1002901, Epub 2011 Feb 7; Chen et al., 2013, Histopathology, 63:225—233, doi: 10.1111/his.12156, Epub 2013 Jun 6).
RORY has also been identified to have a regulatory role in glucose homeostasis, and has been implicated in metabolic syndrome, obesity (Meissburger et al., 2011, EMBO Mol.
Med., 3:637—65 1), hepatosteatosis, insulin ance and diabetes. r support for the role of RORY in the enesis of inflammatory, metabolic, circadian effect, cancer, and autoimmune diseases and disorders can be found in the following references: Chang et al., 2012, J. Exp. Pharmacol., 4:141—148; Jetten et al., 2013, Frontiers Endocrinol., 4:1—8; Huh & n, 2012, Eur. J. Immunol., 42:2232—2237; Martinez et al., 2008, Ann. NY. Acad. Sci., 1143:188—211; Pantelyushin et al., 2012, J. Clin. Invest., 122:2252— 2256; Jetten & Ueda, 2002, Cell Death Differen., 9:1167—1 171; Solt et al., 2010, Curr. Opin.
Lipidol., 21 :204-21 1.
In light of the role that RORY plays in disease pathogenesis, inhibition of RORY actiVity and Th17 cell differentiation and actiVity, including IL17 production, will be of significant therapeutic benefit. It is therefore desirable to prepare compounds that inhibit RORY actiVity and hence have y in the treatment of inflammatory, autoimmune, metabolic, circadian effect, cancer, and other diseases mediated by RORy, such as e.g., , atopic dermatitis, acne, Crohn’s disease, regional enteritis, tive s, Sjogren's syndrome, uveitis, Behcet's disease, dermatomyositis, multiple sclerosis, ankylosing spondylitis, systemic lupus erythematosus, derma, psoriasis, psoriatic arthritis, steroid resistant asthma and rheumatoid arthritis.
SUMMARY OF THE INVENTION It has now been found that compounds described herein, and ceutically acceptable compositions thereof, are effective tors of RORY (see e.g., Table 2). Such compounds include those of Formula (I): c 2 x L1 y \ M\Cy1 (\—)—N | n m N/ R3 (I); or a pharmaceutically acceptable salt thereof, wherein each of R2, R3, R4, X, L1, n, m, Cyl, and Cy2 are as defined and described herein.
WO 16904 The provided compounds, and pharmaceutically able compositions thereof, are inverse agonists or antagonists of RORY and are useful for treating a variety of diseases, disorders or conditions. Such diseases, disorders, or conditions include those described herein.
The provided compounds can be used alone (i.e., as a monotherapy) or in combination with one or more other therapeutic agent effective for treating any of the indications described herein.
ED DESCRIPTION OF CERTAIN EMBODIMENTS 1. General DescriQtion of Compounds of the Invention In certain embodiments, the present invention provides a compound of Formula (1): 2 1 (\—)—N | y In N/ R3 (1); or a pharmaceutically acceptable salt thereof, wherein: R2 and R3 are each independently hydrogen, hydroxy, monocyclic lkyl, monocyclic heterocyclyl, or (C1—C6)alkyl, wherein the (C1—C6)alkyl is ally substituted with l to 2 groups independently selected from hydroxy, halo, and cyano; R4 is en, (C1—C3)alkyl, or :0; X is —C(O)NH— or —NHC(O)—; m is 0, l, or 2; nis 0, l, 2, or 3; L1 is absent or is SOZ or CR7R8; Cy1 is absent or is ed from (C1—C3)alkyl, halo(C1—C3)alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl, wherein the aryl, heteroaryl, heterocyclyl, and cycloalkyl are each optionally tuted with l to 3 groups independently selected from R5; Cy2 is absent or is selected from (C1—C6)alkoxycarbonyl, phenyl(C1—C3)alkoxycarbonyl, halophenyl(C1—C3)alkoxycarbonyl, aryl, heteroaryl, clic cycloalkyl, and monocyclic heterocyclyl, wherein the aryl, heteroaryl, monocyclic lkyl, and moncyclic heterocyclyl are each optionally substituted with l to 3 groups independently selected from R6; R5 and R6 are each independently ed from halo, cyano, nitro, amino, hydroxy, carboxy, (C1—C6)alkyl, heterocyclyl, hydroxy(C1—C6)alkyl, COZH, _3COOH, (C1— C3)all<ylcarbonyloxy, (C3—C6)cycloalkyl, hydroxy(C3—C6)cycloalkyl, (C4—C7)cycloalkylalkyl, (C2— C6)alkenyl, halo(C2—C6)all<enyl, hydroxy(C2—C6)alkenyl, (C2—C6)all<ynyl, (C3—C6)cycloall<yl(C2— C4)a1kyny1, halo(C1—C6)alky1, halo(C3—C6)cycloalky1, halo(C4—C7)cycloa1ky1alky1, (C1—C6)alkoxy, (C3—C6)cycloalkoxy, (C4—C7)cycloa1ky1alkoxy, halo(C1—C6)alkoxy, halo(C3—C6)cycloalkoxy, halo(C4—C7)cycloa1ky1alkoxy, (C1—C6)alky1thio, (C3—C6)cycloalkythio, (C4— C7)cycloa1ky1a1ky1thio, 1—C6)alky1thio, halo(C3—C6)cycloalkythio, halo(C4— loa1ky1a1ky1thio, (C1—C6)alkylsulfinyl, (C3—C6)cycloa1kylsulfiny1, (C4— C7)cycloalkylalkylsulfinyl, halo(C1—C6)alkylsulfony1, halo(C3—C6)cycloalkylsulfiny1, halo(C4— C7)cycloalkylalkylsulfinyl, (C1—C6)a1kylsulfony1, )cycloa1kylsulfony1, (C4— C7)cycloa1ky1alky1sulfonyl, halo(C1—C6)alkylsulfony1, halo(C3—C6)cycloa1kylsulfony1, halo(C4— C7)cycloa1ky1alky1sulfonyl, (C1—C6)alky1amino, di(C1—C6)alky1amino, (C1—C6)alkoxy(C1— C6)alkoxy, halo(C1—C6)alkoxy(C1—C6)alkoxy, (C1—C6)alkoxycarbony1, HZNCO, , (C1- C6)a1ky1aminocarbony1, di(C1—C6)a1ky1aminocarbony1, (C1—C3)alkoxy(C1— C3)a1ky1aminocarbony1, heterocyclylcarbonyl, (C1—C6)a1ky1aminosulfonyl, di(C1— C6)a1ky1aminosulfony1, heterocyclylsulfonyl, (C1—C6)a1ky1carbony1amino, )alky1— carbonylamino(C1—C6)a1ky1, (C1—C6)a1kylsulfony1amino, (C1—C6)a1kylsulfony1amino(C1—C6)a1ky1, (C1—C6)alkoxycarbony1(C1—C6)alkoxy, (C1—C6)alkoxy(C1—C6)alkyl, halo(C1—C6)alkoxy(C1— C6)alky1, hydroxy(C1—C6)alkoxy, aryl, heteroaryl, oxo, amino(C1—C6)alky1, (C1— C6)a1ky1amino(C1—C6)a1ky1, di(C1—C6)a1ky1amino(C1—C6)alky1 amino(C2—C6)alkoxy, (C1— C6)a1ky1amino(C2—C6)alkoxy, di(C1—C6)a1ky1amino(C2—C6)alkoxy, (C1—C6)alky1carbony1, y(C1—C6)a1ky1carbony1, )a1ky1hydroxycarbonyl, (C1—C6)a1ky1hydroxy(C1—C6)a1ky1, (C3—C6)cycloa1ky1carbony1, (C3—C6)cycloalkylaminocarbonyl, {(C3—C6)cycloalky1} { (C1— C6)a1ky1}aminocarbony1, di(C3—C6)cycloalkylaminocarbonyl, (C3—C6)cycloalkylaminosulfonyl, {(C3—C6)cycloalky1}{(C1—C6)a1ky1}aminosu1fony1, di(C3—C6)cycloalky1aminosulfonyl, cyano(C1— C6)alky1, aminocarbony1(C1—C6)a1ky1, (C1—C6)a1ky1aminocarbony1(C1—C6)a1ky1, di(C1— C6)a1ky1aminocarbony1(C1—C6)a1ky1, (C3—C6)cycloa1ky1aminocarbony1(C1—C6)alky1, {(C3— C6)cycloa1ky1} { (C1—C6)a1ky1}aminocarbony1(C1—C6)a1ky1, [(C1—C6)a1ky1(C4—C6)heterocyc1y1](C1— C6)alky1, and di(C3—C6)cycloa1ky1aminocarbony1(C1—C6)a1ky1; and R7 and R8 are each independently hydrogen, hydroxy, (C1—C3)a1ky1, hydroxy(C1—C3)alky1, mono(C1—C3)alky1amino, C3)alky1amino, COZH, _3COOH, moncyclic heterocyclyl, (C1—C3)a1koxycarbony1, (C1—C3)a1ky1(C1—C3)alkoxycarbony1, halophenyl, halopheny1(C1— C3)a1ky1, or quinolin—2(1H)one—4y1—methy1; or R7 and R8, together with the carbon atom to which they are ed, form a 3— to 6— membered cycloalkyl or heterocyclyl.
PCT/U82015/013699 2. Comgounds and Definitions The terms “halo” and “halogen” as used herein refer to an atom selected from fluorine (fluoro, —F), chlorine (chloro, —Cl), bromine (bromo, —Br), and iodine (iodo, —I).
The term “alkyl”, used alone or as a part of a larger moiety such as e.g., “haloalkyl”, means a saturated monovalent ht or branched arbon radical , unless otherwise specified, 1—10 carbon atoms and includes, for example, methyl, ethyl, n—propyl, isopropyl, n— butyl, sec—butyl, isobutyl, tert—butyl, n—pentyl, n—hexyl, n—heptyl, n—octyl, n—nonyl, n—decyl and the like. “Monovalent” means attached to the rest of the molecule at one point.
The term “haloalkyl” or “halocycloalkyl” include mono, poly, and perhaloalkyl groups where the ns are independently selected from fluorine, chlorine, and bromine.
The terms “cycloalkyl” and “cycloaliphatic”, used alone or as part of a larger moiety, refer to a saturated cyclic aliphatic monocyclic or bicyclic ring system, as described herein, haVing from, unless otherwise specified, 3 to 10 carbon ring atoms. Monocyclic cycloalkyl groups include, without limitation, cyclopropyl, utyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, and cyclooctyl. It will be understood that when specified, al substituents on a lkyl or liphatic group may be present on any substitutable position and, include, e.g., the position at which the cycloalkyl or cycloaliphatic group is attached.
The term “carbocycle”, cyclyl”, “carbocyclo”, or “carbocyclic” used alone or as part of a larger moiety refer to saturated, partially saturated, or aromatic ring systems comprising all carbon atoms haVing, unless otherwise specified, a total of 3 to 10 ring members.
It will be understood that when ied, optional substituents on a carbocycle, carbocyclyl, carbocyclo, or carbocyclic may be present on any substitutable position and, include, e.g., the position at which the lkyl is attached.
The term “aryl” used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refers to an aromatic carbocyclic ring system , unless otherwise specified, a total of 6 to 10 ring members. The term “aryl” may be used interchangeably with the term “aryl ring”, “aryl group”, “aryl ,” or “aryl l”. In n embodiments of the present disclosure, “aryl” refers to an aromatic ring system which includes, but is not limited to, phenyl (abbreviated as “Ph”), naphthyl and the like. It will be understood that when specified, optional substituents on an aryl group may be present on any substitutable position and, include, e.g., the position at which the aryl is attached.
The term “heteroaryl” used alone or as part of a larger moiety as in “heteroarylalkyl”, “heteroarylalkoxy”, or "heteroarylaminoalkyl”, refers to a 5—10 —membered aromatic radical 2015/013699 containing 1—4 heteroatoms ed from N, O, and S and includes, for example, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The term oaryl” may be used hangeably with the terms “heteroaryl ring”, “heteroaryl group”, or “heteroaromatic”. The terms “heteroaryl” and “heteroar—”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl rings, where the radical or point of attachment is on the heteroaromatic ring.
Nonlimiting es e indolyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, quinazolinyl, and quinoxalinyl. A heteroaryl group may be mono— or bicyclic. It will be understood that when specified, optional substituents on a heteroaryl group may be present on any tutable position and, e, e.g., the position at which the heteroaryl is attached.
The term “heterocyclyl” means a 4—, 5—, 6— and 7—membered saturated or partially unsaturated cyclic ring containing 1 to 4 heteroatoms independently selected from N, O, and S. The terms “heterocycle”, “heterocyclyl”, “heterocyclyl ring”, “heterocyclic group”, “heterocyclic moiety”, and “heterocyclic radical”, are used interchangeably herein. A heterocyclyl ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. Examples of such saturated or partially unsaturated heterocyclic radicals include, without tion, tetrahydrofuranyl, tetrahydrothienyl, terahydropyranyl, pyrrolidinyl, idonyl, piperidinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, morpholinyl, dihydrofuranyl, dihydropyranyl, dihydropyridinyl, tetrahydropyridinyl, dihydropyrimidinyl, and tetrahydropyrimidinyl. A heterocyclyl group may be mono— or bicyclic.
Unless otherwise specified, bicyclic heterocyclyl groups include, e.g., unsaturated heterocyclic radicals fused to another unsaturated heterocyclic radical or aromatic or heteroaryl ring, such as for e, tetrahydronaphthyridine, indolinone, opyrrolotriazole, imidazopyrimidine, inone, dioxaspirodecane. It will also be understood that when specified, optional substituents on a heterocyclyl group may be present on any tutable position and, include, e.g., the position at which the heterocyclyl is attached.
As used herein the terms “subject” and “patient” may be used interchangeably, and means a mammal in need of treatment, e.g., companion s (e.g., dogs, cats, and the like), farm animals (e.g., cows, pigs, horses, sheep, goats and the like) and laboratory animals (e.g., rats, mice, guinea pigs and the like). Typically, the subject is a human in need of treatment.
Certain of the disclosed compounds may exist in various stereoisomeric forms.
Stereoisomers are compounds that differ only in their spatial ement. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they 2015/013699 contain an trically substituted carbon atom that acts as a chiral . “Enantiomer” means one of a pair of les that are mirror images of each other and are not superimposable. reomers are stereoisomers that contain two or more asymmetrically substituted carbon atoms. The symbol “*” in a structural formula represents the presence of a chiral carbon center. “R” and “S” represent the configuration of substituents around one or more chiral carbon atoms. Thus, “R*” and “8*” denote the relative configurations of substituents around one or more chiral carbon atoms.
“Racemate” or “racemic mixture” means a compound of equimolar quantities of two enantiomers, wherein such mixtures exhibit no optical activity, i.e., they do not rotate the plane of polarized light.
“Geometric isomer” means isomers that differ in the orientation of substituent atoms in relationship to a carbon—carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon—carbon double bond may be in an E (substituents are on opposite sides of the carbon—carbon double bond) or Z (substituents are oriented on the same side) configuration. “R,” “S,” “S*,” “R*,” “E,” “Z,” “cis,” and “trans,” indicate configurations relative to the core molecule.
The nds of the invention may be prepared as individual enantiomers by either enantio—specific synthesis or resolved from an enantiomerically enriched mixture. Conventional resolution techniques include forming the salt of a free base of each isomer of an enantiomeric pair using an lly active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each enantiomer of an enantiomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide of each of the enantiomers of an omeric pair using an optically pure acid, amine or alcohol (followed by chromatographic tion and removal of the chiral auxiliary), or resolving an enantiomeric mixture of either a starting material or a final product using various well known chromatographic methods.
When the stereochemistry of a disclosed compound is named or ed by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight optically pure. Percent optical purity by weight is the ratio of the weight of the enantiomer over the weight of the enantiomer plus the weight of its l isomer.
When a sed nd is named or depicted by structure without indicating the chemistry, and the compound has at least one chiral center, it is to be understood that the name or ure encompasses one enantiomer of compound free from the corresponding optical isomer, a racemic mixture of the compound and mixtures enriched in one enantiomer relative to its corresponding optical isomer.
When a sed compound is named or depicted by structure without indicating the stereochemistry and has at least two chiral centers, it is to be understood that the name or structure encompasses a reomer free of other diastereomers, a pair of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other reomer(s) and mixtures of diastereomeric pairs in which one diastereomeric pair is enriched relative to the other diastereomeric pair(s).
The compounds of the invention may be present in the form of pharmaceutically able salts. For use in medicines, the salts of the nds of the ion refer to non— toxic “pharmaceutically acceptable salts.” Pharmaceutically acceptable salt forms include pharmaceutically able acidic/anionic or basic/cationic salts.
Pharmaceutically acceptable basic/cationic salts e, the sodium, potassium, calcium, magnesium, diethanolamine, n—methyl—D—glucamine, L—lysine, L—arginine, ammonium, ethanolamine, piperazine and anolamine salts.
Pharmaceutically acceptable acidic/anionic salts include, e.g., the acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, carbonate, citrate, dihydrochloride, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, malate, e, malonate, te, nitrate, salicylate, stearate, succinate, e, tartrate, and tosylate. 3. Description of Exemglary Comgounds In a first embodiment, the present invention provides a compound of Formula (I), c 2 x L1 y \ M \Cy1 (\—)—N | n 1T1 N/ R3 (1); or a pharmaceutically acceptable salt thereof, wherein the variables are as described above.
In a second embodiment, the compound of Formula (I) is of Formula (II): R4 O c2y AxCW \ N n L1 (\—)—N | H m / R3 (H); or a pharmaceutically acceptable salt thereof, wherein the variables in structural Formula (II) are as described for Formula (I).
In a third embodiment, the compound of Formula (I) is of Formula (III): R4 O Cy2 \ chw (\—)—N | H m / R3 (III); or a pharmaceutically acceptable salt f, wherein the variables in structural Formula (III) are as described for Formula (I).
In a fourth embodiment, the compound of Formula (I) is of Formula (IV): R4 O Cy? A R2 (IV); or a ceutically acceptable salt thereof, n the variables in structural Formula (IV) are as described for Formula (I).
In a fifth embodiment, the compound of a (I) is of Formula (V): (V); or a pharmaceutically able salt thereof, wherein the variables in structural Formula (V) are as described for Formula (I).
In a sixth embodiment, the compound of Formula (I) is of Formula (VI): 2015/013699 (VI); or a pharmaceutically acceptable salt thereof, wherein the variables in ural Formula (VI) are as bed for Formula (I).
In a seventh embodiment, the compound of Formula (I) is of Formula (VII): S2 (VII); or a pharmaceutically acceptable salt thereof, wherein the variables in structural Formula (VII) are as described for a (I).
In an eighth embodiment, R2 and R3 in Formulas (I) to (VII) are each independently hydrogen, hydroxy, or (C1—C3)alkyl, wherein the remainder of the varables are as described in Formula (I).
In a ninth ment, Cy2 in Formulas (I) to (VII) is present and is selected from aryl, heteroaryl, monocyclic cycloalkyl, and monocyclic heterocyclyl, each of which is optionally substituted with l to 2 groups independently selected from R6, wherein the remainder of the varables are as described in a (I) or the eighth embodiment.
In a tenth embodiment, Cy2 in Formulas (I) to (VII) is phenyl, pyrimidinyl, cyclohexyl, or pyridinyl, each of which are optionally substituted with l to 2 groups independently ed from R6, wherein the remainder of the varables are as described in Formula (I) and the eighth or ninth embodiment.
In an eleventh embodiment, Cy1 in Formulas (I) to (VII) is phenyl, piperidinyl, tetrahydro—2H—thiopyranyl l,l—dioxide, pyridinyl, zinyl, azetidinyl, imidazolyl, tetrahydropyranyl, l,4—dioxanyl, pyridazinyl, pyrazolyl, idinyl, cyclohexyl, morpholinyl, 6,7—dihydro—5H—pyrrolo[2,l—c][l,2,4]triazolyl, l,2,3,4—tetrahydro—l,8—naphthyridinyl, 2,3— dihydro—lH—indenyl, or imidazo[l,2—a]pyrimidinyl, each of which is optionally substituted with l to 2 groups independently selected from R5, wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, or tenth embodiment.
In a twelfth embodiment, Cy1 in Formulas (I) to (VII) is phenyl, piperidinyl, tetrahydro—2H—thiopyranyl l,l—dioxide, pyridinyl, piperazinyl, azetidinyl, imidazolyl, tetrahydropyranyl, l,4—dioxanyl, pyridazinyl, pyrazolyl, pyrrolidinyl, cyclohexyl, linyl, 6,7—dihydro—5H—pyrrolo[2,l—c][l,2,4]triazolyl, l,2,3,4—tetrahydro—l,8—naphthyridinyl, 2,3— dihydro—lH—indenyl, or imidazo[l,2—a]pyrimidinyl, each of which is optionally substituted with l to 2 groups independently selected from R5, wherein at least one R5 is (C1—C3)alkylsulfonyl or (C1—C3)alkylaminosulfonyl, and wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, or th embodiment.
In a thirteenth embodiment, R2 in as (I) to (VII) is (C1—C3)alkyl; n is l or 2; and Cy1 is , pyridinyl, or piperidinyl, each of which is optionally substituted with l to 2 groups independently selected from R5, wherein at least one R5 is (C1—C3)all<ylsulfonyl or (C1— C3)alkylaminosulfonyl, and n the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, or twelfth embodiment.
In a fourteenth embodiment, Cy2 in Formulas (I) to (VII) is cyclohexyl optionally substituted with l to 2 groups independently selected from R6, wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, twelfth, or thirteenth embodiment.
In a fifteenth embodiment, R5 is selected from halo, (C1—C3)alkyl, 1—C3)alkyl, cyano, hydroxy(C1—C3)alkyl, (C1—C3)alkoxycarbonyl, (C1—C3)alkylsulfonyl, (C1—C3)alkoxy, halo(C1—C3)alkoxy, oxo, hydroxy, (C1—C3)alkylcarbonyl, hydroxy(C1—C3)alkylcarbonyl, (C1— C3)all<ylhydroxycarbonyl, (C1—C3)alkylaminosulfonyl, (C1—C3)alkylaminocarbonyl, di(C1— C3)alkylamino(C2—C6)alkoxy, (C1—C6)alkoxycarbonyl, [(C1—C3)alkyl(C4—C6)heterocyclyl] (C1— <yl, and (C1—C3)alkylhydroxy(C1—C3)alkyl; and R6 is selected from halo, (C1—C3)alkyl, halo(C1—C3)alkyl, cyano, hydroxy(C1—C3)alkyl, (C1—C3)alkoxycarbonyl, )alkylsulfonyl, (C1—C3)alkoxy, halo(C1—C3)alkoxy, oxo, y, aryl(C1—C3)alkoxycarbonyl, (C1— C3)all<ylhydroxy(C1—C3)alkyl, heteroaryl, and (C1—C3)alkoxycarbonyl, wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, twelfth, enth, or fourteenth embodiment.
Alternatively, R5 is ed from halo, (C1—C3)alkoxy, hydroxy, )alkyl, hydroxy(C1—C3)alkyl, halo(C1—C3)alkyl, (C1—C6)alkoxycarbonyl, di(C1—C3)alkylamino(C2— C6)alkoxy, 3)alkyl(C4—C6)heterocyclyl](C1—C3)alkyl, oxo, (C1—C3)alkylcarbonyl, (C1— C3)alkylaminosulfonyl, (C1—C3)alkylsulfonyl, and cyano; and R6 is selected from halo(C1—C3)alkyl, (C1—C3)alkoxy, halo, cyano, (C1—C3)alkoxycarbonyl, (C1— C3)all<ylhydroxy(C1—C3)alkyl, 2—methyl—2H—tetrazolyl, hydroxy(C1—C3)alkyl, and halo(C1— C3)alkoxy, wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, twelfth, thirteenth, or fourteenth ment. In another alternative, R5 is selected from halo, cyano, (C1—C3)alkyl, (C1—C3)alkylaminosulfonyl, and (C1—C3)alkylsulfonyl; and R6 is selected from 1—C3)alkyl, (C1—C3)alkoxy, halo, cyano, (C1—C3)alkoxycarbonyl, 2— methyl—2H—tetrazolyl, and 1—C3)alkoxy, wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, th, twelfth, thirteenth, or fourteenth embodiment.
In a sixteenth embodiment, Cy1 in Formulas (I) to (VII) is Z / S//O // \R10 0 ;R10 is (C1—C3)alkyl or (C1—C3)alkylamino; and z is CH or N, n the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, twelfth, enth, fourteenth, or fifteenth embodiment. [005 1] In an seventeenth embodiment, Cy2 in Formulas (I) to (VII) is H}:- N / \ 2— RKM M )2- ; — ; —N ; N_ ; or Rio:-; R12 is (C1-C3)all<oxycarbonyl, halo, dihalo, (C1—C3)alkoxy, or halo(C1— C3)alkyl; R13 is halo or halo(C1—C3)alkyl; and R14 is halo, cyano, halo(C1—C3)alkyl, halo(C1— C3)alkoxy, or 2—methyl—2H—tetrazolyl, wherein the remainder of the varables are as described in a (I) and the eighth, ninth, tenth, eleventh, twelfth, thirteenth, fifteenth, or sixteenth embodiment.
In an eighteenth embodiment, R12 to R14 are each CF3, wherein the remainder of the varables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, twelfth, thirteenth, fifteenth, sixteenth, or seventeenth embodiment.
In a nineteenth embodiment, R2 in as (I) to (VII) is isopropyl and the remainder of the variables are as described in Formula (I) and the eighth, ninth, tenth, eleventh, twelfth, thirteenth, enth, fifteenth, sixteenth, seventeenth, or eighteenth embodiment.
In a eth ment, the compound of Formula (I) is of Formula (X): WO 16904 2 H CY1 Cy \ WU/ (\—)—N m | N/ o R3 (X); or a pharmaceutically acceptable salt thereof, wherein L1 is absent; Cy1 is phenyl optionally substituted with SOZ(C1—C3)alkyl or SOZ(C1—C3)alkylamino; n is 0 or 1; R4 is en or (C1— C3)all<yl; R2 and R3 are each independently hydrogen or (C1—C6)alkyl; m is 0 or 1; and Cy2 is phenyl or cyclohexyl, each optionally substituted with halo(C1—C3)alkyl, (C1—C3)alkyl, halo, or Specific examples of compounds of the invention are provided in the EXEMPLIFICATION. Pharmaceutically acceptable salts as well as the neutral forms of these nds are ed in the invention.
In certain embodiments, the present invention provides a method of treating a patient (e.g., a human) with a disorder mediated by RORY comprising the step of stering to the patient an effective amount of the compound with any compound described herein, or a pharmaceutically acceptable salt or composition thereof. 4. Uses, Formulation and Administration Pharmaceutically able compositions According to r embodiment, the present invention provides a method of treating a subject (e.g., a human) with a disorder mediated by RORY using a composition sing a compound of Formula (I) and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In n embodiments, the amount of compound of Formula (I) in a ed composition is such that it is effective as an inverse agonist or antagonist to RORY in a biological sample or in a subject. In n embodiments, a provided composition is formulated for administration to a subject in need of such composition. In some embodiments, a provided composition is formulated for oral administration to a subject. [005 8] The term “pharmaceutically acceptable r, adjuvant, or vehicle” refers to a non— toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the itions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, l glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium en phosphate, potassium en phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose—based substances, polyethylene , sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene—polyoxypropylene—block rs, polyethylene glycol and wool fat.
Compositions described herein may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
The term teral" as used herein es subcutaneous, intravenous, intramuscular, intra— articular, intra—synovial, intrasternal, intrathecal, epatic, intralesional and intracranial injection or infusion ques.
Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable ons, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents ly used in the art such as, for e, water or other ts, solubilizing agents and fiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, l,3—butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and es thereof. Besides inert diluents, the oral itions can also e adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. [006l] able preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or g agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in l,3—butanediol. Among the acceptable vehicles and solvents that may be ed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono— or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
Injectable formulations can be sterilized, for example, by filtration through a bacterial—retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
In order to prolong the effect of a provided compound, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid sion of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. able depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as ctide— ycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable rs include rthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or mulsions that are compatible with body tissues.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or ium phosphate and/or a) fillers or extenders such as starches, lactose, e, glucose, mannitol, and c acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as aga ——agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as in, f) absorption accelerators such as nary um compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, m stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
Solid compositions of a r type may also be employed as fillers in soft and hard— filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, es, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
Examples of embedding compositions that can be used include polymeric substances and waxes.
Solid compositions of a similar type may also be employed as fillers in soft and hard—filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like. ed nds can also be in micro—encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal ce, additional nces other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or entially, in a certain part of the intestinal tract, ally, in a delayed manner.
Examples of embedding compositions that can be used include polymeric substances and waxes.
Dosage forms for topical or transdermal stration of a compound of this invention include ointments, pastes, creams, s, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed vatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of ermal patches, which have the added advantage of providing controlled ry of a compound to the body.
Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. tion enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
Pharmaceutically acceptable compositions provided herein may be formulated for oral administration. Such formulations may be administered with or t food. In some embodiments, pharmaceutically acceptable itions of this disclosure are administered without food. In other embodiments, pharmaceutically acceptable compositions of this disclosure are administered with food.
The amount of provided compounds that may be combined with carrier materials to produce a composition in a single dosage form will vary depending upon the patient to be treated and the particular mode of administration.
It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of s, including age, body weight, general , sex, diet, time of administration, rate of excretion, drug combination, the judgment of the treating physician, and the severity of the particular disease being treated. The amount of a provided compound in the composition will also depend upon the particular nd in the composition.
Uses of Compounds and Pharmaceutically Acceptable Compositions [007 1] Compounds and compositions described herein are generally useful for the inhibition of RORy. Thus, in some embodiments, the present invention provides a method of treating inflammatory, metabolic and autoimmune diseases or disorders mediated by RORy, comprising administering a ed compound or composition. More ularly, the compounds and compositions described herein act as inverse agonists or nists of RORy.
As used herein, the terms “treatment, 79 ‘6treat,” and “treating” refer to ing, alleviating, delaying the onset of, or inhibiting the progress of a disease or er, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed, i.e., eutic treatment. In other ments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors), i.e., lactic treatment. Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence. es and conditions treatable according to the methods of the ion include, but are not limited to, inflammatory, metabolic and autoimmune diseases or ers mediated by RORy. These diseases and conditions include, for example, asthma, chronic obstructive pulmonary disease (COPD), bronchitis, allergic rhinitis, atopic dermatitis, contact dermatitis, acne, cystic fibrosis, allograft rejection, multiple sis, scleroderma, arthritis, rheumatoid arthritis, juvenile toid arthritis, osteoarthritis, ankylosing spondylitis, systemic lupus erythematosus (SLE), psoriasis, Hashimoto's disease, pancreatitis, autoimmune diabetes, type I diabetes, autoimmune ocular disease, ulcerative colitis, Crohn's disease, regional enteritis, inflammatory bowel disease (IBD), inflammatory bowel syndrome (IBS), Sjogren's syndrome, optic neuritis, y, hepatosteatosis, adipose tissue—associated ation, insulin resistance, type II diabetes, neuromyelitis optica, myasthenia gravis, age related r degeneration, dry eye, uveitis, Guillain—Barré syndrome, psoriasis, psoriatic arthritis (PsA), steroid resistant asthma, Graves' disease, scleritis, endometriosis, obstructive sleep apnea syndrome (OSAS), Behcet's disease, dermatomyositis, polymyocitis, graft versus host disease, primary biliary cirrhosis, liver fibrosis, non—alcoholic fatty liver disease (NAFLD), sarcoidosis, y sclerosing cholangitis, autoimmune thyroid disease, autoimmune polyendocrine syndrome type I, autoimmune polyendocrine syndrome type II, celiac disease, neuromyelitis, juvenile idiopathic arthritis, systemic sclerosis, myocardial infarction, pulmonary hypertension, osteoarthritis, cutaneous leishmaniasis, sinonasal polyposis, and cancer, including but not limited to lung cancer, gastric cancer, breast cancer and colon cancer.
Also included are es or disorders which are implicated by the tion of the circadian rhythm of individuals and include, e.g., major depression, al ive disorder, post—traumatic stress disorder (PTSD), bipolar disorder, autism, epilepsy, Alzheimer’s disease and other central nervous system (CNS) disorders associated with altered sleep and/or circadian rhythms.
In one embodiment, a human patient is treated with a compound of Formula (I) and a pharmaceutically acceptable carrier, nt, or vehicle, wherein said compound is t in an amount to treat or rate one or more of the diseases and conditions d above. In an alternative embodiment, the diseases and conditions d or ameliorated by a compound of Formula (I) include, e.g., asthma, atopic dermatitis, acne, Crohn’s disease, regional enteritis, ulcerative colitis, n's syndrome, uveitis, Behcet's disease, dermatomyositis, multiple sclerosis, ankylosing spondylitis, systemic lupus erythematosus (SLE), scleroderma, psoriasis, psoriatic arthritis (PsA), steroid resistant asthma and rheumatoid arthritis in the t.
The invention further relates to a combination therapy for treating or ameliorating a disease or a disorder described herein. In some embodiments, the combination therapy comprises administering at least one compound represented by ural Formula I in combination with one or more agents for treating or ameliorating inflammatory, metabolic and autoimmune diseases or disorders mediated by RORy. In some embodiments, the combination y comprises administering at least one compound represented by Structural Formula I in combination with one or more agents for the treatment of es including asthma, chronic obstructive pulmonary disease (COPD), itis, ic rhinitis, atopic dermatitis, contact dermatitis, acne, cystic fibrosis, allograft rejection, le sclerosis, scleroderma, arthritis, rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, systemic lupus erythematosus (SLE), psoriasis, Hashimoto's disease, pancreatitis, autoimmune diabetes, type I diabetes, autoimmune ocular disease, ulcerative colitis, Crohn's disease, regional tis, inflammatory bowel disease (IBD), inflammatory bowel me (IBS), Sjogren's syndrome, optic neuritis, y, hepatosteatosis, adipose tissue—associated inflammation, n resistance, type II diabetes, neuromyelitis optica, myasthenia gravis, age related macular degeneration, dry eye, uveitis, Guillain—Barré syndrome, psoriasis, psoriatic arthritis (PsA), steroid resistant asthma, Graves' disease, scleritis, major depression, seasonal affective disorder, PTSD, bipolar disorder, autism, epilepsy, Alzheimer’s, CNS disorders ated with altered sleep and/or circadian rhythms, triosis, obstructive sleep apnea syndrome , Behcet's disease, omyositis, polymyocitis, graft versus host disease, primary y cirrhosis, liver fibrosis, non—alcoholic fatty liver disease (NAFLD), dosis, primary sclerosing cholangitis, autoimmune thyroid disease, autoimmune polyendocrine syndrome type I, autoimmune polyendocrine syndrome type II, celiac disease, neuromyelitis, juvenile idiopathic arthritis, systemic sclerosis, dial infarction, pulmonary hypertension, osteoarthritis, cutaneous leishmaniasis, sinonasal polyposis, and cancer, including but not limited to, lung cancer, gastric cancer, breast cancer and colon cancer.
The compounds according to the invention may also be used in combination with immunotherapies for the treatment of a disease or disorder disclosed herein.
Combination therapy includes, e.g., co—administration of a compound of the invention and one or more other agents, tial administration of a compound of the invention and one or more other , administration of a composition containing a compound of the invention and one or more other , or simultaneous administration of separate compositions containing a compound of the invention and one or more other agents.
The ion further provides a method of treating a t, such as a human, suffering from one of the abovementioned ers or diseases.
The invention further relates to the use of provided compounds for the production of pharmaceutical compositions which are employed for the treatment and/or prophylaxis and/or amelioration of the diseases and disorders mentioned . [008l] Compounds or compositions bed herein may be administered using any amount and any route of stration effective for treating or lessening the severity of one or more of the diseases and conditions described herein. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Provided compounds are preferably formulated in unit dosage form for ease of administration and uniformity of dosage.
The expression "unit dosage form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of s including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific ition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
Pharmaceutically able compositions of this disclosure can be administered to humans and other animals orally, rectally, parenterally, isternally, aginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being d. In certain embodiments, provided compounds may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic .
The term “biological sample”, as used , includes, t limitation, cell cultures or extracts thereof, biopsied material obtained from a mammal or ts thereof, and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
The amount of both, a provided compound and onal therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the provided compound may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent.
The amount of additional therapeutic agent present in the compositions of this disclosure will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
EXEMPLIFICATION As depicted in the Examples below, in n exemplary embodiments, compounds are ed according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the t invention, the following l methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
PCT/U82015/013699 GENERAL DESCRIPTION OF SYNTHESIS The compounds of the present invention can be readily ed according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. Many of the reactions can also be carried out under microwave (MW) conditions or using tional heating or utilizing other technologies such as solid phase ts/scavengers or flow chemistry. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in the art, but are not mentioned in greater detail. Furthermore, other methods for preparing compounds of the invention will be readily apparent to a person of ordinary skill in the art in light of the following reaction schemes and examples. In cases where synthetic intermediates and final products contain potentially reactive functional groups, for e amino, hydroxy, thiol and carboxylic acid groups, that may ere with the d reaction, it may be advantageous to employ protected forms of the intermediate. Methods for the selection, introduction and subsequent removal of protecting groups are well known to those skilled in the art. In the discussion below variables have the meanings indicated above unless otherwise indicated. The abbreviations used in these experimental details are listed below and additional ones should be known to a person skilled in the art of synthesis. In addition, one can refer to the following references for suitable methods of synthesis as described in March, Advanced Organic try, 3rd n, John Wiley & Sons, 1985, Greene and Wuts, Protective Groups in Organic Synthesis, 2nd edition, John Wiley & Sons, 1991, and Richard Larock, Comprehensive Organic Transformations, 4th edition, VCH publishers Inc., 1989. lly, reagents in the reaction schemes are used in equimolar s; however, in certain cases it may be desirable to use an excess of one reagent to drive a reaction to completion. This is especially the case when the excess reagent can be readily removed by evaporation or extraction. Bases ed to neutralize HCl in reaction mixtures are generally used in slight to substantial excess (1.05 — 5 lents).
Where NMR data are presented, spectra were obtained on a Varian 400 (400 MHz) or 300 (300 MHz) and are reported as ppm downfield from tetramethylsilane with number of , multiplicities and ng constants ted parenthetically along with reference to deuterated solvent.
The invention is illustrated by way of the following es, in which the following abbreviations may be employed.
Abbreviation Meaning ACN, MeCN, CH3CN acetonitrile AIBN azobisisobutyronitrile aq aqueous Boc utoxycarbonyl or t—butoxycarbonyl brine saturated aqueous NaCl Cbz oxy carbonyl CeC13 ceric de C52CO3 cesium carbonate Cul cuprous iodide DCM or CH2C12 methylene chloride DIEA diisopropyl ethyl amine DMF dimethyl formamide DMS/Megs dimethyl sulfide DMSO dimethyl ide EDCI l—(3—dimethylaminopropyl)—3—ethylcarbodiiimide hydrochloride Etl ethyl iodide Et ethyl EtzO ethyl ether EtgsiH triethylsilane Et3N ylamine EtOAc AcOEt , EA, ethyl acetate EtOH ethanol FeC13 ferric chloride h, hr hour(s) HATU 0—(7—azabenzotriazol— l —yl)—N,N,N’,N’— tetramethyluronium—heXafluorophosphate HBTU O—benzotriazole— l —yl—N,N,N’,N ’—tetramethyluronium— hexafluorophosphate HCl hydrochloric acid H20 water H202 hydrogen peroxide HPLC high performance liquid chromatography i—BuOCOCl iso—butoxycarbonyl chloride ICl iodochloride K2C03 potassium carbonate K3PO4 tripotassium phosphate LC—MS liquid chromatography—mass spectrometry LDA lithium diiisopropylamide LiCl lithium chloride LiOH lithium hydroxide MCPBA, m-CPBA meta—chloroperoxybenzoic acid MeOH methanol Mel methyl iodide Me methyl mg milligram MgSO4 magnesium sulfate (anhydrous) min minute(s) mL iters mmol millimoles melting point mass spectrometry microwave sodium borohydride sodium cyanoborohydride sodium hydride sodium bicarbonate sodium hydroxide sodium methoxide sodium thiosulfate sodium dithionate sodium sulfate um hydroxide um ate ammonium chloride sodium carbonate sodium bicarbonate sodium hydride N—bromosuccinimide n—butyllithium N—methyl—morpholine N—methyl—pyrrolidin—2—one OTf trifluoromethanesulfonate 2015/013699 OTs tosylate PdClzdppf [l, l —bis(diphenylphosphino)ferrocene] dichloropalladium(ii) Pd2(dba)3 tris(dibenzylideneacetone)dipalladium(0) PE eum ether rt room temperature sat. saturated SFC supercritical fluid chromatography t—BuOK ium tert butoxide t-BuLi tert butyl lithium t-BuOOH tert butyl peroxide TBAF tetrabutylammonium fluoride TFA trifluoroacetic acid THF tetrahydrofuran TLC thin layer chromatography Ti(OEt)4 titanium tetra de Zn zinc Zn(CN)2 zinc cyanide Compounds ing to Formula (I), can be prepared by reacting an intermediate compound of Formula (500) with an alkyl or aryl halide, according to reaction Scheme 1, a reaction that is med in a polar aprotic t, such as, for example, acetonitrile, in the presence of a suitable base, such as, for example, isopropylethylamine or potassium carbonate. Alternatively, the final compounds according to Formula (I), can be prepared by reacting an intermediate compound of Formula (500) with an aldehyde or ketone, according to reaction Scheme 1, following art—known reductive amination procedure, in the typical solvent, such as, for example, dichloroethane, dichloromethane, or methanol; in the presence of suitable reducing reagent, such as sodium cyanoborohydride or sodium triacetoxyborohydride. In reaction Scheme 1, all variables are defined as in Formula (I) and G1 is a leaving group, such as for example, bromide, chloride, mesylate (methanesulfonate), tosylate (p—toluenesulfonate), or iodide.
Scheme 1.
R4 R4 A L\1 X L1 C 2 G1 Cy2 X \ 1 \ \ 1 y m M C y HN | M; Cy MmN | n N/ or ive N R2 R3 ammination R2 R3 ediate compound of Formula (500) can be can be prepared by deprotecting an intermediate compound of Formula (501), wherein Pg is a suitable nitrogen ting group (Greene and Wuts, Protective Groups in Organic Synthesis, 2Ild edition, John Wiley & Sons, 1991), e.g., Pg 2 tert—butoxycarbonyl, removed with trifluoroacetic acid according to Scheme 2.
In reaction Scheme 2, all variables are defined as in Formula (1).
Scheme 2.
X‘M’nL1 L1 de rotection X‘M’n R2 R3N R2 R3N Intermediate compound of a (502), wherein X is C(=O)NH, can be prepared from a carboxylic acid (504) and an amine (503), according to Scheme 3. The reaction is conveniently carried out in the presence of an activating reagent, for e, N— (3— dimethylaminopropyl)—N'-ethylcarbodiimide hydrochloride (EDCI) or 0—(7—azabenzotriazol—l— yl)—N,N,N’,N’— tetramethyluronium hexafluorophosphate (HATU), in an organic solvent, for example, N,N—dimethylformamide or dichloromethane, optionally in the presence of a base, e.g., N,N—diisopropylethylamine or triethylamine, at a temperature, for example in the range from 0 to 60 °C.
Scheme 3.
H2N 0 O R4 n Cyl 503 \ \ Ni’i‘u’ I P _Ng | H n Pg—N / / N N Base, 3 R2 R3 R2 R ting reagents Intermediate compound of Formula (505), wherein X is NHC=O, can be prepared from an intermediate compound of Formula (506) and an amide (507), according to Scheme 4.
The reaction is carried out in the ce of a catalyst, for example, tris(dibenzylideneacetone)dipalladium (0) (Pd2(dba)3), in an organic solvent, for e, dioxane or tert—butanol, in the presence of an ve, e.g., potassium phosphate, at a temperature, for example, in the range from 80 to 150 0C.
PCT/U82015/013699 Scheme 4.
H N / 2 WU CI 507 \ 0 Pg—N | —>P9N :HWL/Cy N catalyst, ligand, additive R2 R3 R2 R3N 506 505 PREPARATION OF INTERMEDIATES As a representative example, intermediate compound of a (504) wherein R4 is H, R2 is isopropyl, R3 is H and Pg is tert—butoxycarbonyl, can be ed by following the reaction steps shown in Scheme 5. An intermediate compound of Formula (504) with variables R4, R2 and R3 can be prepared readily according to Scheme 5, or modifications f, using readily available starting materials and ts.
Scheme 5.
PF6' \N \ \iti/ 0' o I CI I Kok/UMEI O t—BuOK,DABCO, o o o BocHN\)J\ MgCI2 CDI ’ BocHNM NH4OAC E0t \ THF THF /\ /\ é OH OMS CI NaBH4,CaC|2 CI \ MsCI,Et3N CI CI \ \ —> | —> | + | BocHN N/ BocHN BocHN EtOH ; CH2C|2 N/ N/ ; : /\ /\ /\ \ n-butanol, Pd(OAc)2, Na“ BocN | dcpp HBF4, CO (1 atm)- \ OH / BocN I THF :' N ‘ N/ /\ DMF 100 C : Mo(CO)5, Hermann's cat, Fu's salt, DBU BocN MeOH, 160 °C, uwave MeOH/HZO To a stirred solution of compound Boc—Val—OH (3.11 g, 14.3 mmol) in THF (40 mL) at rt was added 1,1’—carbonyldiimidazole (3.48 g, 21.5 mmol). The mixture was stirred at rt for 1 h, then ium chloride (1.36 g, 14.3 mmol) and ethyl potassium malonate (2.44 g, 14.3 2015/013699 mmol) were added successively. The mixture was then heated to 50 °C and stirred for 15 h. The mixture was cooled to rt and quenched with 1 N HCl (100 mL). The aqueous phase was extracted with EtOAc (3 x 100 mL), then the combined organic layer was washed with brine (50 mL). The organic layer was dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was ed by silica gel chromatography (eluting with 5% EtOAc in hexanes) to afford ethyl (S)—4—((tert—butoxycarbonyl)amino)—5—methyl—3—oxohexanoate (3.53 g, 86% yield) as a yellow oil. LC—MS tR = 0.91 min in 1 min chromatography, MS (ESI) m/z 288.3 [M + H]+. 1H NMR (CDC13, 400 MHZ): 5 5.08 (d, J: 8.4 HZ, 1H), 4.33 (dd, J: 4.4 HZ, 8.8 HZ, 1H), 4.20 (q, J: 7.2 HZ, 2H), 3.54 (s, 2H), 2.27-2.17 (m, 1H), 1.44 (s, 9H), 1.27 (t, J = 7.2 HZ, 3H), 1.01 (d, J: 6.8 HZ, 3H), 0.82 (d, J: 6.8 HZ, 3H).
To a e of ethyl (S)—4—((tert—butoxycarbonyl)amino)—5—methyl—3—oxohexanoate (9.68 g, 33.7 mmol) from above in THF (100 mL) at 0 °C was added potassium tert—butoxide (3.78 g, 35.4 mmol). The mixture was warmed to rt and stirred for 45 min, at which point 1,4— diazabicyclo[2.2.2]octane (3.78 g, 33.7 mmol) and 2—chloro—1,3—bis(dimethylamino)trimethinium rophosphate (15.5 g, 50.5 mmol) were added successively. The mixture was heated to 45 °C and stirred for 3 h, at which point ammonium acetate (5.19 g, 67.4 mmol) was added. The mixture was then heated to reflux and stirred for 15 h. It was then cooled to rt and concentrated under reduced pressure. The residue was dry—loaded onto a silica gel column and purified (eluting with 5% EtOAc in hexanes, gradient to 15%) to yield 6.09 g of ethyl (S)—2—(1—((tert— butoxycarbonyl)amino)—2—methylpropyl)—5—chloronicotinate (51%). LC—MS tR = 1.14 min in 1 min chromatography, MS (ESI) m/Z 357.3 [M + H]+. 1H NMR (CDClg, 400 MHZ): 5 8.61 (d, J: 2.4 HZ, 1H), 8.18 (d, J: 2.8 HZ, 1H), 5.71 (d, J: 9.6 HZ, 1H), 5.62 (dd, J: 5.2 HZ, 9.6 HZ, 1H), 4.42 (q, J: 7.2 HZ, 2H), 2.08-2.00 (m, 1H), 1.42 (s, 9H), 1.42 (t, J: 7.2 HZ, 3H), 0.93 (d, J: 6.4 HZ, 3H), 0.83 (d, J: 6.4 HZ, 3H).
To a stirred on of ethyl (1—((tert—butoxycarbonyl)amino)—2—methylpropyl)— —chloronicotinate (6.09 g, 17.1 mmol) at 0 0C in EtOH (70 mL) was added sodium borohydride (1.30 g, 34.1 mmol). Calcium chloride (1.89 g, 17.1 mmol) was added portionwise while maintaining the temperature between 0 °C and 5 OC. The ing mixture was stirred at 0 °C for 90 min, then quenched slowly at 0 °C with saturated aqueous um chloride solution (100 mL). The s phase was extracted with EtOAc (3 x 100 mL), then the combined organic layer was washed with brine (50 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. Crude tert—butyl (S)—(1—(5—chloro—3—(hydroxymethyl)pyridin—2—yl)—2— methylpropyl)carbamate was carried forward without any purification. LC—MS tR = 0.94 min in 1 min chromatography, MS (ESI) m/z 315.3 [M + H]+. 1H NMR (CDCl3, 400 MHZ): 5 8.46 (d, J = 2.4 Hz, 1H), 7.67 (d, J: 2.8 Hz, 1H), 5.34 (d, J: 9.2 Hz, 1H), 4.99 (dd, J: 2.0 Hz, 8.4 Hz, 1H), 4.54 (t, J: 9.2 Hz, 1H), 4.41 (dd, J: 10.0 Hz, 12.4 Hz, 1H), 4.33 (d, J: 10.0 Hz, 1H), 2.18-2.12 (m, 1H), 1.36 (s, 9H), 1.10 (d, J: 6.4 Hz, 3H), 0.69 (d, J: 6.8 Hz, 3H).
To a solution of tert—butyl (S)—(1—(5—chloro—3—(hydroxymethyl)pyridin—2—yl)—2— methylpropyl)carbamate (5.33 g, 16.9 mmol) in CHzClz (70 mL) at 0 °C was added triethylamine (3.54 mL, 25.4 mmol) and methanesulfonyl chloride (1.44 mL, 18.6 mmol). The e was warmed to rt and stirred for 3 h, at which point it was quenched with saturated aqueous sodium bicarbonate solution (100 mL). The aqueous phase was extracted with ethyl acetate (3 x 100 mL). The combined organic layer was washed with brine (50 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The crude residue (a 3:1 mixture of (S)—(2—(1— ((tert—butoxycarbonyl)amino)—2—methylpropyl)—5—chloropyridin—3—yl)methyl methanesulfonate and tert—butyl (S)—(1—(5—chloro—3—(chloromethyl)pyridin—2—yl)—2—methylpropyl)carbamate) was carried forward t any purification. LC—MS tR = 1.01 min in 1 min chromatography, MS (ESI) m/z 393.3 [M + H]+. 1H NMR (CDC13, 400 MHz): 5 8.53 (d, J: 2.4 Hz, 1H), 7.74 (d, J: 2.8 Hz, 1H), 5.44 (d, J: 12.4 Hz, 1H), 5.37 (d, J: 12.8 Hz, 1H), 5.31 (d, J: 8.4 Hz, 1H), 4.59 (t, J: 9.2 Hz, 1H), 3.13 (s, 3H), 2.13-2.04 (m, 1H), 1.36 (s, 9H), 1.03 (d, J: 6.8 Hz, 3H), 0.77 (d, J = 6.8 Hz, 3H). Characterization data from a purified sample of (S)—(2—(1—((tert— butoxycarbonyl)amino)—2—methylpropyl)—5—chloropyridin—3—yl)methyl esulfonate.
To a on of (S)—(2—(1—((tert—butoxycarbonyl)amino)—2—methylpropyl)—5— chloropyridin—3—yl)methyl methanesulfonate and tert—butyl —(5—chloro—3— omethyl)pyridin—2—yl)—2—methylpropyl)carbamate (3:1 mixture, 6.39 g, 16.9 mmol) in THF (75 mL) at 0 °C was added sodium hydride (60% dispersion in mineral oil, 811 mg, 20.3 mmol).
The mixture was warmed to rt and stirred for 15 h, at which point it was quenched with saturated aqueous ammonium chloride solution (100 mL). The aqueous phase was extracted with ethyl acetate (3 x 100 mL). The combined organic layer was washed with brine (50 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel tography (eluting with 5% EtOAc in hexanes, nt to 10%) to give tert— butyl (S)—3—chloro—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (4.31 g, 85% yield over 3 steps) as a yellow oil. LC—MS tR = 1.12 min in 1 min chromatography, MS (ESI) m/z 297.3 [M + H]+. 1H NMR , 400 MHz, mixture of rotamers): 5 8.43 (s, 1H), 7.56 (s, 0.6H), 7.50 (s, 0.4H), 4.96 (s, 0.4H), 4.87 (s, 0.6H), 4.86 (d, J: 16.0 Hz, 0.6H), 4.74 (d, J: .6 Hz, 0.4H), 4.52 (d, J: 12.0 Hz, 0.4H), 4.49 (d, J: 15.2 Hz, 0.6H), 2.60—2.51 (m, 0.4H), 2.40—2.36 (m, 0.6H), 1.49 (s, 9H), 1.08 (d, J: 7.2 Hz, 1.2H), 0.99 (d, J: 7.2 Hz, 1.8H), 0.78 (d, J: 6.8 Hz, 1.8H), 0.72 (d, J: 6.8 Hz, 1.2H).
Potassium carbonate (758 mg, 5.49 mmol) and 4A lar sieves (250 mg) were placed in a 50 mL round—bottom flask which was then flame dried. Palladium (II) acetate (32.8 mg, 146 umol) and 1,3—bis(dicyclohexylphosphonium)propane bis (tetrafluoroborate) (179 mg, 292 umol) were added to the flask, which was then sealed with a septum. Tert—butyl (S)—3— chloro—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (1.09 g, 3.66 mmol) was ved in DMF (12 mL) and added to the flask, followed by 1—butanol (3.34 mL, 36.6 mmol). The flask was then evacuated and backfilled with CO three times, with the final time under a balloon of 1 atm of CO. The flask was heated to 100 °C and stirred for 6 h. The mixture was then cooled to rt and quenched with 1 N NaOH (25 mL). The mixture was stirred for 30 min, at which point isopropyl acetate (50 mL) was added. The phases were separated, then the organic phase was extracted with 1 N NaOH (2 x 50 mL), then the combined aqueous layer was ied to pH = 2 with concentrated HCl. The aqueous layer was then extracted with EtOAc (3 x 25 mL), then the combined organic layer was dried over anhydrous MgSO4, filtered and concentrated under reduced re. The crude e (S)—6—(tert—butoxycarbonyl)—7—isopropyl— 6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxylic acid was carried forward without any purification.
Alternative ep procedure for (S)—6—(tert—butoxycarbonyl)—7—isopropyl—6,7— dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxylic acid: To a solution of tert—butyl (S)—3—chloro—7— isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (158 mg, 532 umol) in MeOH (2.5 mL) in a MW vial was added molybdenum hexacarbonyl (155 mg, 587 umol) and 1,8— diazabicyclo[5.4.0]undec—7—ene (279 uL, 1.86 mmol). The mixture was degassed with N2 for 15 min, at which point trans—bis(acetate)bis[o—(di—o—tolylphosphino)benzyl]dipalladium (II) (25.0 mg, 26.6 umol) and tri—tert—butylphosphonium tetrafluoroborate (30.9 mg, 107 umol) were added. The vial was sealed and heated in the MW at 160 °C for 20 min. After cooling to rt, the mixture was filtered through Celite with MeOH and concentrated under reduced re. The residue was purified by silica gel chromatography ng with 10% EtOAc in hexanes, gradient to 25%) to afford 70.7 mg of t—butyl) 3—methyl (S)—7—isopropyl—5,7—dihydro—6H— pyrrolo[3,4—b]pyridine—3,6—dicarboxylate (41% yield). LC—MS tR = 1.04 min in 1 min chromatography, MS (ESI) m/z 321.4 [M + H]+. 1H NMR (CDClg, 400 MHz, mixture of rotamers): 5 9.10 (s, 1H), 8.17 (s, 0.6H), 8.13 (s, 0.4H), 5.05 (s, 0.4H), 4.95 (s, 0.6H), 4.90 (d, J = 15.6 Hz, 0.6H), 4.79 (d, J: 15.6 Hz, 0.4H), 4.58 (d, J: 11.2 Hz, 0.4H), 4.54 (d, J: 15.6 Hz, 0.6H), 3.96 (s, 3H), 2.62—2.53 (m, 0.4H), 2.45—2.38 (m, 0.6H), 1.52 (s, 9H), 1.09 (d, J: 6.8 Hz, 1.2H), 0.99 (d, J: 7.2 Hz, 1.8H), 0.79 (d, J: 6.8 Hz, 1.8H), 0.72 (d, J: 6.8 Hz, 1.2H).
To a solution of 6—(tert—butyl) 3—methyl (S)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4— b]pyridine—3,6—dicarboxylate (70.7 mg, 221 umol) in MeOH was added potassium ide (5.9 M solution in H20, 187 uL, 1.10 mmoL). The mixture was stirred at 40 °C for 1 h, at which point it was cooled to rt and partitioned between diethyl ether (25 mL) and 1 N NaOH (25 mL).
The organic phase was extracted with 1 N NaOH (2 x 25 mL), then the ed aqueous layer was acidified to pH = 2 with concentrated HCl. The aqueous layer was then extracted with EtOAc (3 x 25 mL), then the combined c layer was dried over anhydrous MgSO4, filtered and concentrated under reduced re. The crude residue (S)—6—(tert—butoxycarbonyl)—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxylic acid was carried forward without any purification. LC—MS tR = 0.93 min in 1 min chromatography, MS (ESI) m/z 307.4 [M + H]+. 1H NMR (CDC13, 400 MHz, mixture of rotamers): 5 9.19 (s, 1H), 8.23 (s, 0.6H), 8.19 (s, 0.4H), .09 (s, 0.4H), 4.99 (s, 0.6H), 4.94 (d, J: 15.6 Hz, 0.6H), 4.82 (d, J: 14.4 Hz, 0.4H), 4.60 (d, J = 8.8 Hz, 0.4H), 4.57 (d, J: 16.0 Hz, 0.6H), 2.65—2.57 (m, 0.4H), 2.49—2.41 (m, 0.6H), 1.53 (s, 9H), 1.10 (d, J: 6.4 Hz, 1.2H), 1.00 (d, J: 6.8 Hz, 1.8H), 0.82 (d, J: 6.8 Hz, 1.8H), 0.75 (d, J = 6.8 Hz, 1.2H). (4—(ethylsulfonyl)phenyl)methanamine was prepared following the synthetic route shown in Scheme 6.
Scheme 6.
Etl, cho3 m-CPBA Us NBS, AIBN —< >~SH —> —> /\ —> acetone S/\ CH2C|2 O”Sx‘o CCI4 Br/\©\S/\ HZNS/\/\©\ O MeOH Ol/S\O ] A mixture of ylbenzenethiol (100 g, 0.8 mol) in acetone (1 L) was added iodoethane (190 g, 1.2 mol) and potassium carbonate (220 g, 1.6 mol). The mixture was stirred at 60 °C overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure to afford crude ethyl(p—tolyl)sulfane (120 g, 99%) as a yellow solid, which was used for the next step without further purification.
To a solution of crude p—tolyl)sulfane (35 g, 0.23 mol) in CHZClz (1.5 L) was added m—chloroperoxybenzoic acid (101 g, 0.59 mol) at 0 OC. The mixture was stirred at rt overnight. The mixture was filtered. The filtrate was added to saturated aqueous N32803 (500 mL) slowly and then stirred for 0.5 h. After partitioning, the organic layer was washed with saturated s NaHC03 (500 mL), dried over anhydrous NaZSO4, filtered and concentrated under reduced pressure to afford crude 1—(ethylsulfonyl)—4—methylbenzene (42.3 g, 100%) as a pale yellow solid, which was used for the next step without further cation.
To a solution of 1—(ethylsulfonyl)—4—methylbenzene (5 g, 25.7 mmol) in CCl4 (30 mL) was added N—bromosuccinimide (5.54 g, 30.8 mmol) and azobisisobutyronitrile (0.46 g, 2.57 mmol). The mixture was d at 80 °C overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was added to water (50 mL) and extracted with EtOAc (3 x 30 mL). The ed organic layers were washed with water (2 x 40 mL) and brine (50 mL), dried over anhydrous NaZSO4, filtered and trated under reduced pressure to afford crude l—(bromomethyl)—4—(ethylsulfonyl)benzene (6.62 g, 98%) as a yellow solid, which was used for the next step without further purification.
To a solution of 1—(bromomethyl)—4—(ethylsulfonyl)benzene (6.62 g, 25.2 mmol) in MeOH (30 mL) was added 28% aqueous ammonium hydroxide solution (30 mL). The mixture was stirred at rt overnight. The mixture was then concentrated under reduced pressure. The residue was purified by basic preparative HPLC separation to afford (4— sulfonyl)phenyl)methanamine (1.5 g, 30%) as a yellow solid. LC—MS tR = 1.747 min in 0— 30CD_3 min chromatography (Durashell C18, 2.1*30 mm, 3 um), MS (ESI) m/z 200.0 [M + H]+ and 399.0 [2M + H]+. 1H NMR (CDC13, 400 MHz): 5 7.85 (d, J: 8.0 Hz, 2H), 7.53 (d, J: 8.0 Hz, 2H), 3.98 (s, 2H), 3.10 (q, J: 7.6 Hz, 2H), 1.26 (t, J: 7.6 Hz, 3H). Preparative Basic HPLC Method mobile phase A: water with 0.05% NH3H20 on; mobile phase B: MeCN; flow rate: mL/min; detection: UV 220 nm / 254 nm; column: Synergi 200 mm x 25 mm x 5 um; column temperature: 30 0C Time in min %A %B 0.0 63 37 8.0 33 67 8.10 0 100 .0 0 100 .1 70 30 12 70 30 (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine— 3—carboxamide was prepared ing the synthetic route shown in Scheme 7.
Scheme 7.
HATU i—PrZNEt BocN 802Et DMF \ ”/0 BocN ON/\©\ HN I )lllh,l / 55 8023 (15) CHZCIZ N 802Et To a solution of (S)—6—(tert—butoxycarbonyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxylic acid (83.3 mg, 281 umol) and (4—(ethylsulfonyl)phenyl)methanamine (67.1 mg, 337 umol) in DMF (2 mL) at rt was added HATU (160 mg, 421 umol) and diisopropylethylamine (97.8 uL, 561 umol). The mixture was stirred at rt for 15 h, at which point it was quenched with saturated s ammonium chloride solution (15 mL). EtOAc (25 mL) was added, then the phases were separated. The organic layer was washed with brine (15 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with 50% EtOAc in hexanes, gradient to 100%) to afford 98.0 mg tert—butyl ((4—(ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl—5,7—dihydro— 6H—pyrrolo[3,4—b]pyridine—6—carboxylate, (15), (77%). LC—MS tR = 0.92 min in 1 min chromatography, MS (ESI) m/z 488.5 [M + H]+. 1H NMR (CDgOD, 400 MHz, mixture of rotamers): 5 8.94 (d, J: 2.0 Hz, 1H), 8.16 (d, J: 12.0 Hz, 1H), 7.89 (dd, J: 2.0 Hz, 8.4 Hz, 2H), 7.64 (d, J: 8.8 Hz, 2H), 4.96 (m, 1H), 4.83 (m, 1H), 4.70 (s, 2H), 4.59 (m, 1H), 3.20 (q, J: 7.2 Hz, 2H), .42 (m, 1H), 1.48 (s, 9H), 1.20 (t, J: 7.2 Hz, 3H), 1.06 (d, J: 7.2 Hz, 1.5H), 1.02 (d, J: 6.8 Hz, 1.5H), 0.80 (d, J: 6.8 Hz, 1.5H), 0.74 (d, J: 6.8 Hz, 1.5H).
To a solution of utyl (S)—3—((4—(ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl— hydro—6H—pyrrolo[3,4—[9]pyridine—6—carboxylate (98.0 mg, 201 umol) in CH2C12 (4 mL) at rt was added trifluoroacetic acid (1.5 mL). The solution was stirred for 30 min, then it was cooled to 0 °C and quenched carefully with saturated sodium bicarbonate solution (20 mL). The e was warmed to rt, then 1 N NaOH (10 mL) and brine (10 mL) were added. The s phase was extracted with CH2C12 (5 x 25 mL), then the combined organic layer was dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The crude (S)—N—(4— (ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide was carried forward without any purification. LC—MS tR = 0.49 min in 1 min chromatography, MS (ESI) m/z 388.4 [M + H]+. 1H NMR (CDC13, 400 MHZ): 5 9.06 (t, J: 0.8 Hz, 1H), 8.23 (t, J: 0.8 HZ, 1H), 7.89 (dd, J: 1.6 Hz, 8.4 Hz, 2H), 7.65 (d, J: 8.8 Hz, 2H), 4.85 (d, J: 4.0 Hz, 1H), 4.73 (s, 2H), 4.71 (s, 2H), 3.20 (q, J: 7.6 Hz, 2H), .54 (m, 1H), 1.21 (t, J: 7.6 Hz, 3H), 1.21 (d, J: 6.8 Hz, 3H), 1.07 (d, J: 7.2 Hz, 3H). (5—(ethylsulfonyl)pyridin—2—yl)methanamine was prepared following the synthetic route shown in Scheme 8.
Scheme 8.
EtSOZNa, L-proline, BocHN \ CuI.NaOH BocHN \ HCI | | N / N / N / .HzN/fi Br DMSO 802Et ZHC' MeOH 802Et To a flame dried flask equipped with a stir bar was added tert—butyl ((5— bromopyridin—2—yl)methyl)carbamate (2.92 g, 10.2 mmol), ethane sulfinic acid sodium salt (2.36 g, 20.3 mmol), L—proline (234 mg, 2.03 mmol), copper (I) iodide (194 mg, 1.02 mmol) and sodium hydroxide (81.3 mg, 2.03 mmol). The flask was purged with N2, then DMSO (35 mL) was added. The on mixture was heated to 110 °C and stirred for 15 h. The flask was then cooled to rt and the mixture was partitioned between EtOAc (150 mL) and saturated aqueous ammonium chloride (150 mL). The c phase was separated, washed with brine (50 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was ed by silica gel chromatography (eluting with 35% EtOAc in hexanes, gradient to 60%) to afford 1.81 g tert—butyl ((5—bromopyridin—2—yl)methyl)carbamate (59%). LC—MS tR = 0.74 min in 1 min chromatography, MS (ESI) m/z 301.4 [M + H]+. 1H NMR , 400 MHz): 5 9.02 (dd, J: 0.8 Hz, 2.0 Hz, 1H), 8.15 (dd, J: 2.4 Hz, 8.4 Hz, 1H), 7.49 (dd, J: 0.8 Hz, 8.4 Hz, 1H), .49 (broad s, 1H), 4.55 (d, J: 7.0 Hz, 2H), 3.15 (q, J: 7.2 Hz, 2H), 1.47 (s, 9H), 1.31 (t, J: 7.2 Hz, 3H).
To a solution of tert—butyl ((5—bromopyridin—2—yl)methyl)carbamate (1.81 g, 6.03 mmol) in MeOH (40 mL) at 0 °C was added acetyl chloride (4.30 mL, 60.3 mmol) dropwise over 5 min. The solution was allowed to warm to rt and was stirred for 3 h. The mixture was concentrated under reduced pressure to yield 1.64 g (5—(ethylsulfonyl)pyridin—2—yl)methanamine rochloride (~100%). LC—MS tR = 0.25 min in 1 min chromatography, MS (ESI) m/z 201.2 [M + H]+. 1H NMR (CD3OD, 400 MHz): 5 9.09 (d, J: 1.2 Hz, 1H), 8.35 (dd, J: 2.4 Hz, 8.4 Hz, 1H), 7.49 (d, J: 8.4 Hz, 1H), 4.45 (s, 2H), 3.31 (q, J: 7.2 Hz, 2H), 1.26 (t, J: 7.2 Hz, 3H).
\ H \ BocN I I N/ N / )IIHI, 802Et Tert—butyl (S)—3—(((5—(ethylsulfonyl)pyridin—2—yl)methyl)carbamoyl)—7—isopropyl—5,7— dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (46): Procedure same as that for tert—butyl (S)— 3—((4—(ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6— carboxylate, using (5—(ethylsulfonyl)pyridin—2—yl)methanamine as a starting material. LC—MS tR = 0.86 min in 1 min chromatography, MS (ESI) m/z 489.4 [M + H]+. 1H NMR (CDClg, 400 MHz, mixture of rotamers): 5 9.06 (s, 1H), 8.96 (s, 1H), 8.20 (dd, J: 2.0 Hz, 8.4 Hz, 1H), 8.05 (d, J: 7.0 Hz, 1H), 7.56 (d, J: 8.0 Hz, 1H), 7.52 (s, 1H), 4.99 (m, 2H), 4.90 (m, 2H), 4.80 (d, J = 15.2 Hz, 1H), 4.51 (m, 1H), 3.17 (q, J: 7.6 Hz, 2H), .54 (m, 0.5H), 2.48—2.39 (m, 0.5H), 1.52 (s, 9H), 1.33 (t, J: 7.6 Hz, 3H), 1.10 (d, J: 7.2 Hz, 1.5H), 1.01 (d, J: 6.8 Hz, 1.5H), 0.78 (d, J: 6.8 Hz, 1.5H), 0.73 (d, J: 7.2 Hz, 1.5H).
\ H \ HN- IN/ I : 802Et ((5—(ethylsulfonyl)pyridin—2—yl)methyl)—7—isopropyl—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxamide: Procedure same as that for (S)—N—(4— (ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide, using tert—butyl (S)—3—(((5—(ethylsulfonyl)pyridin—2—yl)methyl)carbamoyl)—7—isopropyl—5,7—dihydro—6H— pyrrolo[3,4—b]pyridine—6—carboxylate as a starting material. LC—MS tR = 0.48 min in 1 min chromatography, MS (ESI) m/z 389.3 [M + H]+. 1H NMR (CDClg, 400 MHZ): 5 9.06 (dd, J: 0.8 Hz, 2.0 Hz, 1H), 8.93 (t, J: 0.8 Hz, 1H), 8.19 (dd, J: 2.0 Hz, 8.4 Hz, 1H), 8.01 (t, J: 0.8 Hz, 1H), 7.56 (dd, J: 0.8 Hz, 8.0 Hz, 1H), 7.52 (s, 1H), 4.89 (d, J: 5.2 Hz, 2H), 4.35 (dd, J: 1.2 Hz, 4.0 Hz, 1H), 4.32 (d, J: 0.8 Hz, 2H), 3.17 (q, J: 7.6 Hz, 2H), 2.34-2.26 (m, 1H), 1.33 (t, J = 7.6 Hz, 3H), 1.09 (d, J: 7.2 Hz, 3H), 0.79 (d, J: 6.8 Hz, 3H). 2—(4—(ethylsulfonyl)phenyl)acetamide was prepared following the synthetic route shown in Scheme 9.
Scheme 9.
HOWNH2 /\0SS"K+ O S\n/OEt 1)aq.KOH,EtOH NaNo2 HCI 3 HO 2) HCI l K CO m--CPBA \\// 3 Eon/OSE—> 0 8“ CHZCIZ 00/9 o o aq. NaOH NH4CI, HATU, Et3N M —> wsv—» 0 EtOH CH2CI2 HO H2N A solution of sodium nitrite (18.4 g, 0.267 mol) in water (133 mL) was added dropwise to a suspension of 2—(4—aminophenyl)acetic acid (40.3 g, 0.267 mol) in water (133 mL) and conc. HCl (54 mL, 0.65 mol) at 0 0C. After addition, the reaction mixture was stirred at the same temperature for 45 min. The solution of the cold diazonium salt was then added dropwise to a mixture of potassium ethyl xanthate (49.3 g, 0.31 mol), water (80 mL) and aqueous sodium carbonate solution (200 mL, 2 M) at rt. After on, the mixture was d to warm to 45 °C and stirred at this temperature until gas evolution ceased (about 3 h to overnight). The mixture was cooled to rt and adjusted to pH = 1 with conc. HCl. The aqueous layer was extracted with ethyl acetate (3 x 300 mL). The combined organic layers were dried over anhydrous NaZSO4, filtered and concentrated under reduced re to afford crude 2— (4— ((ethoxycarbonothioyl)thio)phenyl)acetic acid (50 g, 73%) as a dark red liquid, which was used for next step directly without further cation. 1H NMR ied by pre—TLC, CDC13 300 MHz): 5 7.40 (d, J: 7.5 Hz, 2H), 7.28 (d, J: 7.8 Hz, 2H), 4.54 (q, J: 6.9 Hz, 2H), 3.63 (s, 2H), 1.26 (t, J: 6.9 Hz, 3H).
To a solution of 2—(4—((ethoxycarbonothioyl)thio)phenyl)acetic acid (50.0 g, crude, 0.195 mol) in EtOH (180 mL) was added a solution of KOH (40.5 g, 0.724 mol) in water (180 mL). The mixture was stirred at reflux overnight. The mixture was concentrated under reduced pressure to remove EtOH. The aqueous phase was adjusted to pH = 1~2 with conc. HCl. The aqueous phase was then extracted with ethyl e (3 x 200 mL). The combined organic layers were dried over anhydrous Na2S04, filtered and concentrated under d pressure to afford crude 2—(4—mercaptophenyl)acetic acid (32.0 g, 98%) as a gray solid, which was used for next step directly t r purification. 1H NMR (purified by pre—TLC, CD3OD, 400 MHz): 5 7.23 (d, J: 8.4 Hz, 2H), 7.15 (d, J: 8.0 Hz, 2H), 3.54 (s, 2H).
To a solution of 2—(4—mercaptophenyl)acetic acid (32 g, crude, 0.19 mol) in dry DMF (300 mL) was added potassium carbonate (105 g, 0.76 mol) and iodoethane (118 g, 0.76 mol).
The reaction mixture was stirred at rt overnight. Ethyl acetate (800 mL) and water (600 mL) were added to the mixture. After partitioning, the aqueous layer was ted with ethyl acetate (3 x 500 mL). The combined organic layers were washed with brine (2 x 800 mL), dried over anhydrous NaZSO4, filtered and concentrated under reduced re. The residue was purified by column chromatography on silica gel (eluting with 30:1 petroleum ether:ethyl acetate) to give ethyl 2—(4—(ethylthio)phenyl)acetate (15.3 g, 36%) as a yellow oil. LC—MS tR = 0.881 min in 5— 95AB_1.5min chromatography (Welch Xtimate C18, 2.1*30 mm, 3 um), MS (ESI) m/z 224.8 [M+H]+. 1H NMR (CDC13 300 MHz): 5 7.02 (d, J: 8.1 Hz, 2H), 6.94 (d, J: 8.1 Hz, 2H), 3.89 (q, J: 7.2 Hz, 2H), 3.31 (s, 2H), 2.67 (q, J: 7.5 Hz, 2H), 1.07—0.97 (m, 6H).
To a solution of ethyl 2—(4—(ethylthio)phenyl)acetate (7.8 g, 35 mmol) in CHzClz (100 mL) was added m—chloroperoxybenzoic acid (21 g, 123 mmol) in ns at 0 OC. The reaction mixture was stirred for 16 h at rt. The reaction mixture was filtered. CH2C12 (200 mL) was added to the filtrate and then the e was ed with saturated aqueous N32803 solution (200 mL). After partitioning, the organic layer was washed with saturated aqueous N32803 solution (200 mL) and then saturated s N32C03 solution (300 mL). The combined aqueous layers were extracted with CH2C12 (3 x 400 mL). The combined organic layers were washed with brine (500 mL), dried over anhydrous N32804, filtered and concentrated under reduced pressure. The residue was purified by column on silica gel (eluting with 15% EtOAc in petroleum ether, gradient to 25%) to afford ethyl 2—(4—(ethylsulfonyl)phenyl)acetate (7.0 g, 78%) as a white solid.
LC—MS tR = 0.807 min in 5—95AB_2min chromatography (Welch e C18, 2.1*30 mm, 3 um), MS (ESI) m/z 256.8 [M+H]+. 1H NMR (CDC13 400 MHz): 5 7.87 (d, J: 8.4 Hz, 2H), 7.50 (d, J: 8.0 Hz, 2H), 4.18 (q, J: 6.8 Hz, 2H), 3.72 (s, 2H), 3.11 (q, J: 7.6 Hz, 2H), 1.30-1.25 (m, 6H).
To a on of ethyl 2—(4—(ethylsulfonyl)phenyl)acetate (10.0 g, 39 mmol) in EtOH (100 mL) was added a solution of NaOH (5.7 g, 142.5 mmol) in water (100 mL). The reaction mixture was stirred at rt for 16 h. EtOH was removed under reduced pressure. The aqueous layer was adjusted to pH = 1 with 6 N aq. HCl and ted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine (2 X 100 mL), dried over anhydrous NazSO4, filtered and trated under reduced pressure to give the desired product 2—(4— (ethylsulfonyl)phenyl)acetic acid (7.3 g, 82%) as a white solid. LC—MS tR = 0.573 min in 5— 95AB_1.5min chromatography (Welch Xtimate C18, 2.1*30 mm, 3 um), MS (ESI) m/z 228.8 [M+H]+. 1H NMR (CDC13 400 MHz): 5 7.88 (d, J: 8.0 Hz, 2H), 7.50 (d, J: 8.4 Hz, 2H), 3.77 (s, 2H), 3.12 (q, J: 7.6 Hz, 2H), 1.28 (t, J: 7.6 Hz, 3H).
To a mixture of 2—(4—(ethylsulfonyl)phenyl)acetic acid (3 g, 13.2 mmol), Et3N (4.0 g, 39.6 mmol) and HATU (5.93 g, 15.6 mmol) in anhydrous CHzClz (100 mL) was added NH4Cl (1.54 g, 26.4 mmol). The resulting mixture was d at rt overnight. The mixture was diluted with CH2C12 (100 mL) and washed with water (3 x 80 mL). The organic layer was dried over ous NaZSO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (eluting with 50% EtOAc in petroleum ether, gradient to 100%) to afford crude 2—(4—(ethylsulfonyl)phenyl)acetamide. The crude product was added to petroleum ether/ethyl e (20 mL, 1/ 1), then the mixture was stirred for 0.5 h. The mixture was ed and the filter cake was dried under vacuum to give 2— (4— (ethylsulfonyl)phenyl)acetamide (1.5 g, 50%) as a white solid. LC—MS tR = 0.900 min in 0— 30AB_2 min chromatography (Welch Xtimate C18, 2.1*30 mm, 3 um), MS (ESI) m/z 269.0 [M + H + CH3CN]+. 1H NMR (DMSO-dg, 400 MHz): 5 7.80 (d, J = 8.0 Hz, 2H), 7.58 (broad s, 1H), 7.51 (d, J: 8.0 Hz, 2H), 7.00 (broad s, 1H), 3.50 (s, 2H), 3.25 (q, J: 7.2 Hz, 2H), 1.08 (t, J: 7.2 Hz, 3H).
BocNS I N/ msoza Tert—butyl (S)—3—(2—(4—(ethylsulfonyl)phenyl)acetamido)—7—isopropyl—5,7—dihydro—6H— pyrrolo[3,4—b]pyridine—6—carboxylate: To a flame—dried Vial equipped with a stir bar was added tert—butyl (S)—3—chloro—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (74.7 mg, 252 nmol), ethylsulfonyl)phenyl)acetamide (68.6 mg, 302 nmol), and potassium phosphate (64.1 mg, 302 nmol). Tert—butanol (1.5 mL) was added, then the mixture was degassed with N2 for 15 min. Tris(dibenzylideneacetone)dipalladium (0) (2.3 mg, 252 nmol) and 2—(di—t—butylphosphino)—3,6—dimethoxy—2’,4’,6’—tri—i—propyl—1,1’—biphenyl (4.9 mg, 10.1 umol) were added to the e, then the Vial was sealed and heated at 110 °C for 15 h. The mixture was cooled to rt, then partitioned between EtOAc (15 mL) and brine (15 mL). The organic phase was separated, dried over anhydrous MgSO4, filtered and concentrated under reduced pressure.
The residue was purified by silica gel chromatography (eluting with 10% EtOAc in hexanes, gradient to 100%) to afford 70.2 mg of Tert—butyl (S)—3—(2—(4—(ethylsulfonyl)phenyl)acetamido)— 7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (57%). LC—MS tR = 0.90 min in 1 min chromatography, MS (ESI) m/z 488.5 [M + H]+. 1H NMR , 400 MHz, mixture ofrotamers): 5 8.58 (s, 0.5H), 8.54 (s, 0.5H), 8.10 (s, 1H), 7.89 (dd, J: 8.0 Hz, 2H), 7.64 (d, J: 8.4 Hz, 2H), 4.77 (d, J: 15.6 Hz, 1H), 4.51 (m, 2H), 3.87 (s, 2H), 3.21 (q, J: 7.2 Hz, 2H), 2.46- 2.37 (m, 1H), 1.52 (s, 9H), 1.22 (t, J: 7.6 Hz, 3H), 1.02 (d, J: 6.8 Hz, 1.5H), 0.99 (d, J: 6.4 Hz, 1.5H), 0.77 (d, J: 7.2 Hz, 1.5H), 0.73 (d, J: 6.8 Hz, 1.5H).
/ O .- N 802Et (S)—2—(4—(ethylsulfonyl)phenyl)—N—(7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridin—3—yl)acetamide: Procedure same as that for (S)—N—(4—(ethylsulfonyl)benzyl)—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide, using tert—butyl (S)—3—(2—(4— (ethylsulfonyl)phenyl)acetamido)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6— carboxylate as a starting material. LC—MS tR = 0.50 min in 1 min chromatography, MS (ESI) m/z 388.3 [M + H]+. 1H NMR (CD3OD, 400 MHz): 5 9.02 (s, 1H), 8.48 (s, 1H), 8.26 (d, J: 2.0 Hz, 1H), 7.84 (m, 2H), 7.66 (d, J: 8.0 Hz, 1H), 7.56 (d, J: 8.4 Hz, 1H), 4.78 (m, 2H), 3.98 (s, 1H), 3.81 (s, 1H), 3.70 (s, 1H), 3.20 (q, J: 7.6 Hz, 2H), 2.65-2.57 (m, 1H), 1.21 (t, J: 7.6 Hz, 3H), 1.21 (d, J: 6.8 Hz, 3H), 1.03 (d, J: 6.8 Hz, 3H).
PREPARATION OF COMPOUNDS OF FORMULA I Compounds of Formula (I) were prepared according to the general procedures outlined below.
General ure A: (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6—(4— (trifluoromethyl)benzyl)—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (1). woo Lamchro802Et CH3CN 802Et ] (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine— 3—carboxamide (11.3 mg, 29.2 umol), fluoromethyl)benzyl bromide (10.5 mg, 43.9 umol), and ium carbonate (8.1 mg, 58.6 umol) were stirred together in CH3CN (1 mL) at rt for 2 h. The reaction mixture was quenched with saturated aqueous ammonium chloride on (5 mL) and extracted with EtOAc (10 mL). The organic phase was separated, washed with brine (5 mL), dried over anhydrous MgSO4, filtered and concentrated under d pressure. The residue was purified by silica gel chromatography (eluting with 50% EtOAc in hexanes, gradient to 100%), then r purified by reverse—phase HPLC to yield 5.2 mg of the HCl salt of (S)—N— (4—(ethylsulfonyl)benzyl)—7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamide (1, 31%). LC—MS m/z 546.5 [M + H]+. 1H NMR (CD3OD, 400 MHz): 9.08 (d, J = 1.6 Hz, 1H), 8.28 (s, 1H), 7.89 (d, J = 8.8 Hz, 2H), 7.84 (d, J = 8.0 Hz, 2H), 7.64 (d, J = 8.4 Hz, 2H), 5.03 (d, J = 16.0 Hz, 1H), 4.91 (m, 2H), 4.82 (m, 2H), 4.71 (s, 2H), 3.20 (q, J = 7.2 Hz, 2H), 2.42-2.29 (m, 1H), 1.21 (t, J = 7.6 Hz, 3H), 1.17 (d, J = 8.0 Hz, 3H), 0.89 (d, J = 6.8 Hz, 3H).
Trans—4—(trifluoromethyl)cyclohexane—1—carbaldehyde (was prepared following the synthetic route shown in Scheme 10.
Scheme 10.
-, O ID/H THF -,,,/0H CH2CI2, —78 °c "W0 To a solution of trans—4—(trifluoromethyl)cyclohexane carboxylic acid (789 mg, 4.02 mmol) in THF (12 mL) at rt was added lithium aluminum hydride (1.0 M in THF, 4.02 mL). The mixture was heated to reflux and d for 3 h. It was then cooled to 0 °C and quenched successively with water (152 uL), 15% aqueous sodium hydroxide (152 uL), and water (456 uL).
The mixture was then filtered through Celite and concentrated under reduced pressure. The crude liquid (trans—4—(trifluoromethyl)cyclohexyl)methanol was carried forward without any purification and without placing under high vacuum due to its lity.
] To a solution of oxalyl chloride (6.2 mL, 87.4 mmol) in anhydrous CHZClz (300 mL) was added dropwise DMSO (12.5 mL, 0.17 mol) at —78 °C under N2. After the mixture was stirred at —78 °C for 30 min., a solution of (trans—4—(trifluoromethyl)cyclohexyl)methanol (5.3 g, 29.1 mmol) in CH2C12 (40 mL) was added dropwise while keeping the internal temperature below —65 0C. After being stirred for 30 min., a on of Et3N (40.5 mL, 0.29 mol) in CHZClZ (60 mL) was added dropwise slowly, keeping the internal temperature below —65 OC. The reaction e was stirred at —78 °C for 1 h, and warmed to rt overnight. The mixture was washed with water (3 x 300 mL) and brine (300 mL), dried over ous Na2S04, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica (eluting with 15% EtOAc in petroleum ether) to give trans—4— oromethyl)cyclohexane—1—carbaldehyde (4.6 g, 87%) as a yellow oil.
General procedure B: (S)—N—((5—(ethylsulfonyl)pyridin—2—yl)methyl)—7—isopropyl—6—((trans—4— (trifluoromethyl)cyclohexyl)methyl)—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (2).
O O \ N \ O""\\O,NaBH30N,AcoH 3 \ N \ HN | H | ’—N | H | / N / N / _ N/ N SOzEt MeOH 802Et To a solution of (S)—N—((5—(ethylsulfonyl)pyridin—2—yl)methyl)—7—isopropyl—6,7— dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (3.6 g, 9.25 mmol), trans—4— (trifluoromethyl)cyclohexane—1—carbaldehyde (3.3 g, 18.5 mmol) in anhydrous MeOH (100 mL) was added acetic acid dropwise until the pH was between 6 and 7. Sodium cyanoborohydride (1.7 g, 27.75 mmol) was added portionwise at rt. The mixture was heated to 70 °C for 1 h. Upon WO 16904 completion, the reaction mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in ethyl acetate (100 mL), then the organic phase was washed with water (3 X 100 mL) and brine (100 mL). The organic layer was dried over anhydrous Na2S04, ed and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with 50% EtOAc in heXanes, gradient to 100%), then further purified by a chiral column using the following ions: instrument: Berger MultiGramTM SFC, Mettler Toledo Co, Ltd; column: AD 300 mm X 50 mm, 10 um; mobile phase: A: supercritical C02, B: iPrOH (0.05% lamine), A:B = 60:40 at 200mL/min; column temp: 38 OC; nozzle pressure: 100 bar; nozzle temp: 60 OC; evaporator temp: 20 OC; trimmer temp: 25 OC; wavelength: 220 nm. Isomer SFC tR = 2.28 min in 12 min chromatography was isolated as the major isomer, which was then further purified by reverse—phase HPLC to give (S)—N—((5—(ethylsulfonyl)pyridin—2—yl)methyl)—7— isopropyl—6—((trans—4—(trifluoromethyl)cycloheXyl)methyl)—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carb0Xamide (2, HCl salt) as a light yellow solid. LC—MS tR = 0.69 min in 1 min tography, MS (ESI) m/z 553.1 [M + H]+. 1H NMR (CD3OD, 400 MHz): 5 9.12 (s, 1H), 9.01 (d, J: 2.0 Hz, 1H), 8.36—8.32 (m, 2H), 7.75 (d, J: 8.0 Hz, 1H), 5.16 (d, J: 15.6 Hz, 1H), 4.89-4.87 (m, 2H), 4.73 (d, J: 15.2 Hz, 1H), 3.42-3.34 (m, 3H), 3.31—3.28 (m, 2H), 2.58—2.51 (m, 1H), 2.20-1.90 (m, 6H), 1.50-1.39 (m, 2H), 1.33 (d, J: 7.2 Hz, 3H), 1.26 (t, J: 7.2 Hz, 3H), 1.24—1.20 (m, 2H), 1.10 (d, J = 6.8 Hz, 3H). HCl preparative HPLC method mobile phase A: water with 0.05% HCl; mobile phase B: CH3CN; flow rate: 80 mL/min; detection: UV 220 nm/ 254 nm; column: PhenomeneX Gemini C18 (250 mm X 50 mm X 5 um); column temperature: 30 Time in min %A %B 0.00 75 25 .00 50 50 .00 0 100 A sample of 2 was converted to the HBr salt by the ing procedure: The HCl salt of 2 (57.5 mg, 97.7 umol) was dissolved in EtOAc (25 mL) and washed with ted aqueous sodium bicarbonate (20 mL) and brine (10 mL). The organic phase was dried over anhydrous MgSO4, ed and trated under reduced pressure. It was then redissolved in acetonitrile (200 uL), to which was added hydrobromic acid (48 wt% in water, 69 uL) to give a light yellow clear solution. The solvent was removed under reduced pressure, then more acetonitrile (300 uL) was added. This procedure was performed iteratively until most of the water and eXcess HBr was removed, leaving behind a yellow solid. This solid was redissolved in acetonitrile (6 mL), seeded with an HBr salt crystal (<1 mg), and stirred at rt for 30 min to give a white solid. The solid was filtered and dried under high vacuum for 3 h to give the HBr salt (40.2 mg, 86%). Melting point 2 171-173 0C. 1H NMR (CD3OD, 400 MHz): 5 9.12 (s, 1 H), 9.07 (s, 1H), 8.46 (d, J = 8.4 Hz, 1H), 8.34 (s, 1H), 7.85 (d, J = 8.4 Hz, 1H), 5.18 (d, J: 14.2 Hz, 1H), 4.94 (m, 1H), 4.87 (s, 2H), 4.75 (d, J: 14.2 Hz. 1H), 3.41 (m, 2H), 3.38 (q, J: 7.6 Hz, 2H), 2.54 (m, 1H), 2.17 (m, 1H), 2.04 (m, 5H), 1.45 (m, 2H), 1.32 (d, J: 7.2 Hz, 3H), 1.27 (t, J: 7.6 Hz, 3H), 1.23 (m, 2H), 1.10 (d, J: 6.4 Hz, 3H).
HBr seed crystals were formed as follows: 2 (5.6 mg) was converted to the free base, then to the HBr salt, as described above. The resultant yellow solid was dissolved in acetonitrile (200 uL), then left to stand overnight at rt in a capped vial. ess crystals formed, which were identified as plate—shaped under a cope.
General procedure C: (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6—(4— (trifluoromethyl)pyrimidin—2—yl)—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (6).
O N O / F3C \>—CI Moor\ N —N ,i-PerEt N mm\ N i-PrOH, 140 °C, uwave SOzEt /\ /\ To a solution of (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxamide (27.3 mg, 70.4 umol) in panol (1.5 mL) in a MW vial was added 2—chloro—5—(trifluoromethyl)pyrimidine (19.3 mg, 106 umol) and diisopropylethylamine (24.5 uL, 140.7 umol). The vial was sealed and heated in the MW at 140 0C for 2 h. The solvent was then evaporated and the residue was ed by silica gel chromatography (eluting with 60% EtOAc in hexanes, gradient to 100%), then further purified by reverse—phase HPLC to yield 7.2 mg of (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6—(4— (trifluoromethyl)pyrimidin—2—yl)—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (6, 19%). LC-MS tR = 0.97 min in 1 min chromatography, MS (ESI) m/z 534.5 [M + H]+. 1H NMR (CD3OD, 400 MHz): 5 9.34 (t, J: 1.2 Hz, 1H), 8.97 (s, 1H), 8.69 (d, J: 4.8 Hz, 1H), 8.25 (s, 1H), 7.90 (d, J: 8.4 Hz, 2H), 7.65 (d, J: 8.0 Hz, 2H), 7.02 (d, J: 4.8 Hz, 1H), 5.39 (s, 1H), .14 (d, J: 15.6 Hz, 1H), 4.87 (m, 1H), 4.72 (d, J: 6.0 Hz, 2H), 3.20 (q, J: 7.6 Hz, 2H), 2.79- 2.68 (m, 1H), 1.22 (broad s, 3H), 1.21 (t, J: 7.6 Hz, 3H), 0.69 (broad s, 3H).
] General procedure D: (S)—6—((4—cyanophenyl)sulfonyl)—N—(4—(ethylsulfonyl)benzyl)— 7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (13).
\ N NC—Q8020| 1 Eth HN l/ H —» \ N N SOZEt CH2C|2,rt 0438‘“ IN/ H /\ s 80E2 t To a solution of (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxamide (6.9 mg, 17.8 umol) in CHZClZ (600 uL) was added triethylamine (5.0 uL, 36.6 umol) and 4—cyanobenzenesulfonyl chloride (5.4 mg, 26.8 umol).
The mixture was stirred at rt for 15 h, at which point it was ed with saturated aqueous sodium bicarbonate (10 mL). The mixture was extracted with EtOAc (10 mL), then the organic phase was washed with brine (10 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was purified by reverse—phase HPLC to yield 4.9 mg of (S)— 6—((4—cyanophenyl)sulfonyl)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxamide as an HCl salt (13, 46%). LC-MS tR = 0.84 min in 1 min chromatography, MS (ESI) m/z 553.5 [M + H]+. 1H NMR (CD3OD, 400 MHZ): 5 8.85 (d, J = 2.0 Hz, 1H), 8.09 (d, J: 1.6 Hz, 1H), 8.03 (dd, J: 2.0 Hz, 8.8 Hz, 2H), 7.86 (m, 4H), 7.60 (d, J = 8.4 Hz, 2H), 4.93 (m, 2H), 4.73 (dt, J: 1.2 Hz, 16.4 Hz, 1H), 4.66 (s, 2H), 3.19 (q, J: 7.6 Hz, 2H), 2.41-2.32 (m, 1H), 1.20 (t, J: 7.6 Hz, 3H), 1.00 (d, J: 7.2 Hz, 3H), 0.96 (d, J: 6.8 Hz, 3H).
General procedure E: benzyl (S)—3—((4—(ethylsulfonyl)benzyl)carbamoyl)—7— isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (16).
O o \ N BnOCOCI, Et3N, DMAP BnO \ N HN | H / Hm >_N I .- N 802Et O CH2C|2, rt N SOzEt «5 /\ 16 To a solution of (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxamide (11.3 mg, 29.2 umol) in CHZClZ (1 mL) was added benzyl chloroformate (11.7 uL, 3.0 M in e), ylamine (6.1 uL, 43.8 umol) and 4— dimethylaminopyridine (50 ug, 4.09 umol). The mixture was stirred at rt for 15 h, at which point it was quenched with saturated s sodium bicarbonate (10 mL). The mixture was extracted with EtOAc (10 mL), then the organic phase was washed with brine (10 mL), dried over anhydrous MgSO4, ed and concentrated under reduced pressure. The residue was purified by silica gel chromatography ng with 30% EtOAc in hexanes, gradient to 100%), then r purified by reverse—phase HPLC to yield 14.9 mg of benzyl (S)—3—((4— (ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6— ylate as an HCl salt (16, 91%). LC-MS tR = 0.91 min in 1 min chromatography, MS (ESI) m/z 522.5 [M + H]+. 1H NMR (CD3OD, 400 MHz): 5 9.00 (s, 1H), 8.34 (s, 1H), 7.87 (dd, J: 1.6 Hz, 6.8 Hz, 2H), 7.63 (d, J: 8.8 Hz, 2H), 7.42 (dd, J: 1.6 Hz, 8.4 Hz, 2H), 7.33 (m, 3H), 5.20 (m, 3H), 4.96 (m, 1H), 4.71 (m, 3H), 3.20 (q, J: 7.6 Hz, 2H), 2.57-2.36 (m, 1H), 1.19 (t, J: 7.6 Hz, 3H), 1.00 (dd, J: 6.8 Hz, 16.0 Hz, 3H), 0.78 (dd, J: 6.8 Hz, 25.6 Hz, 3H). (7S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—5—methyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamide was prepared following the synthetic route shown in Scheme 11.
Scheme 11. 0 0 0' NaBH4 CI EC \ HNMe(OMe)-HCI,MeMgBr \ | | BOCHN N/ B°CHN THE-20°C N/ EtOH,O°C ; ; /\ /'\ 0' MsCI EtN \ BocHN / BocHN . N N l/N 5 CH2C|2 /\ /\ /\ n—butanol, Pd(OAc)2 “©3028 HATU 1,_'Pr NEt dcpp HBF4, CO (1 atm). 2 BocN DMF 100°C 802Et CH2C|2 Usoza ] To a solution of ethyl (S)—2—(1—((tert—butoxycarbonyl)amino)—2—methylpropyl)—5— chloronicotinate (104 mg, 291 umol) and N,O—dimethylhydroxylamine hydrochloride (45.5 mg, 466 umol) in THF (1 mL) was added dropwise methylmagnesium bromide (1.4 M in 3:1 toluene:THF, 1.04 mL) at —20 OC. The mixture was stirred at —20 °C for 45 min, at which point it was quenched carefully with cold ted aqueous ammonium chloride (10 mL). The mixture was extracted with EtOAc (20 mL), then the organic phase was washed with brine (10 mL), dried over anhydrous MgSO4, ed and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with 10% EtOAc in hexanes, gradient to 60%) to afford 62.3 mg of tert—butyl (S)—(1—(3—acetyl—5—chloropyridin—2—yl)—2—methylpropyl)carbamate (56% yield). LC-MS tR = 1.08 min in 1 min chromatography, MS (ESI) m/z 327.4 [M + HT". 2015/013699 To a solution of tert—butyl (S)—(1—(3—acetyl—5—chloropyridin—2—yl)—2— methylpropyl)carbamate (62.3 mg, 191 umol) in EtOH (1 mL) at 0 °C was added sodium borohydride (7.2 mg, 191 umol) as a solid. The mixture was stirred at 0 °C for 90 min, then it was quenched with saturated s ammonium chloride (10 mL). The mixture was extracted with EtOAc (20 mL), then the organic phase was washed with brine (10 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The e was purified by silica gel chromatography (eluting with 10% EtOAc in hexanes, gradient to 100%) to afford 53.4 mg of tert—butyl ((lS)—1—(5—chloro—3—(1—hydroxyethyl)pyridin—2—yl)—2— methylpropyl)carbamate (85% yield). LC-MS tR = 1.01 min in 1 min chromatography, MS (ESI) m/z 329.4 [M + H]+. 1H NMR , 400 MHz, mixture of diastereomers): 5 8.44 (d, J = 2.4 Hz, 0.3H), 8.41 (d, J: 2.4 Hz, 0.7H), 7.86 (d, J: 2.4 Hz, 0.7H), 7.77 (d, J: 2.4 Hz, 0.3H), 5.44 (d, J: 9.6 Hz, 0.7H), 5.31 (m, 0.7H), 5.23 (m, 0.7H), 4.73 (t, J: 8.8 Hz, 0.3H), 4.63 (t, J: 9.6 Hz, 0.3H), 4.56 (s, 0.3H), 2.18—2.12 (m, 0.3H), 2.08—2.00 (m, 0.7H), 1.56 (d, J: 6.8 Hz, 0.9H), 1.52 (d, J: 6.8 Hz, 2.1H), 1.40 (s, 9H), 1.11 (d, J: 6.8 Hz, 0.9H), 1.00 (d, J: 6.8 Hz, 2,1H), 0.80 (d, J: 6.8 Hz, 2.1H), 0.69 (d, J: 6.8 Hz, 0.9H). 1—(2—((S)—1—((tert—butoxycarbonyl)amino)—2—methylpropyl)—5—chloropyridin—3—yl)ethyl esulfonate. Procedure same as that for (S)—(2—(1—((tert—butoxycarbonyl)amino)—2— methylpropyl)—5—chloropyridin—3—yl)methyl esulfonate, using tert—butyl ((1S)—1—(5—chloro— 3—(1—hydroxyethyl)pyridin—2—yl)—2—methylpropyl)carbamate as a starting material. LC-MS tR = 1.03 min in 1 min chromatography, MS (ESI) m/z 407.4 [M + H]+.
Tert—butyl (7S)—3—chloro—7—isopropyl—5—methyl—5,7—dihydro—6H—pyrrolo[3,4— b]pyridine—6—carboxylate. Procedure same as that for tert—butyl (S)—3—chloro—7—isopropyl—5,7— dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate, using 1—(2—((S)—1—((tert— butoxycarbonyl)amino)—2—methylpropyl)—5—chloropyridin—3—yl)ethyl methanesulfonate as a starting material. LC-MS tR = 1.12 min in 1 min chromatography, MS (ESI) m/z 311.4 [M + H]+. 1H NMR , 400 MHz, mixture of diastereomers): 5 8.43 (d, J = 1.6 Hz, 1H), 7.52 (s, 0.7H), 7.46 (s, 0.3H), 5.05—5.00 (m, 1H), 4.94 (s, 0.3H), 4.81 (s, 0.7H), 2.88—2.39 (m, 0.3H), 2.32—2.08 (m, 0.7H), 1.55 (m, 3H), 1.44 (s, 9H), 1.26 (d, J: 7.2 Hz, 0.9H), 1.09 (broad s, 2.1H), 0.81 (broad s, 2.1H), 0.48 (d, J: 7.2 Hz, 0.9H). (7S)—6—(tert—butoxycarbonyl)—7—isopropyl—5—methyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxylic acid. Procedure same as that for (S)—6—(tert—butoxycarbonyl)—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxylic acid, using utyl (7S)—3— chloro—7—isopropyl—5—methyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate as a starting material. LC-MS tR = 0.99 min in 1 min chromatography, MS (ESI) m/z 321.5 [M + H]+. utyl (7S)—3—((4—(ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl—5—methyl—5,7— dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate. Procedure same as that for utyl (S)—3— ((4—(ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6— ylate, using (7S)—6—(tert—butoxycarbonyl)—7—isopropyl—5—methyl—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxylic acid as a starting material. LC-MS tR = 0.95 min in 1 min chromatography, MS (ESI) m/z 502.6 [M + H]+. 1H NMR (CD3OD, 400 MHz, mixture of diastereomers): 5 8.93 (s, 1H), 7.79 (d, J: 8.4 Hz, 2H), 7.50 (d, J: 8.4 Hz, 2H), 7.24 (s, 1H), .06—4.88 (m, 2H), 4.82—4.70 (m, 2H), 3.06 (q, J: 7.2 Hz, 2H), 2.48-2.26 (m, 1H), 1.59 (d, J: 7.2 Hz, 0.9H), 1.56 (d, J: 7.2 Hz, 2.1H), 1.53 (s, 9H), 1.25 (t, J: 7.2 Hz, 3H), 1.09 (d, J: 7.2 Hz, 0.9H), 0.97 (broad s, 2.1H), 0.83 (broad s, 2.1H), 0.48 (d, J: 7.2 Hz, 0.9H). (7S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—5—methyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamide. Procedure same as that for (S)—N—(4—(ethylsulfonyl)benzyl)—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide, using tert—butyl (7S)—3—((4— (ethylsulfonyl)benzyl)carbamoyl)—7—isopropyl—5—methyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine— 6—carboxylate as a starting material. LC-MS tR = 0.49 min in 1 min chromatography, MS (ESI) m/z 402.3 [M + H]+. 1H NMR (CDgOD, 400 MHz, mixture of diastereomers): 5 8.91 (dd, J = 1.2 Hz, 2.0 Hz, 1H), 8.11 (dd, J: 0.8 Hz, 2.0 Hz, 0.4H), 8.07 (dd, J: 1.2 Hz, 1.6 Hz, 0.6H), 7.89 (d, J: 8.4 Hz, 2H), 7.63 (d, J: 8.4 Hz, 2H), 4.70 (s, 2H), 4.57 (m, 0.4H), 4.48 (m, 0.6H), 4.30 (m, 1H), 3.20 (q, J: 7.2 Hz, 2H), 2.40-2.33 (m, 0.6H), 2.27-2.21 (m, 0.4H), 1.50 (d, J: 6.8 Hz, 1.8H), 1.47 (d, J: 7.2 Hz, 1.2H), 1.20 (t, J: 7.6 Hz, 3H), 1.14 (d, J: 7.2 Hz, 1.8H), 1.08 (d, J: 7.2 Hz, 1.2H), 0.81 (d, J: 6.8 Hz, 1.2H), 0.80 (d, J: 6.8 Hz, 1.8H).
] N—(4—(ethylsulfonyl)benzyl)—7—(tetrahydrofuran—3—yl)—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamide was prepared following the synthetic route shown in Scheme 12.
Scheme 12.
KOK/lkOEt EtOOC HZN COOH BocHN COOH Boc20 NaOH MgCI2 CDI BocHN THF/HZO HF \N \ \N I OH 0' EtOOC t—BuOK DABCO CI NH4OAc BocHN NaBH4. CaClz | TsCI, Et3N , BocHN N/ _> EtOH THF CHZCIZ H2N/\©\/\©\802Et 6,H'ermann5 cat.
BOON CFu6's salt DBU BocHN dioxane 160 °C uwave \ N \ N BocN | TFA N/ HO/\©\ ,s\/\ —’ N/ H/\©\,s\/\ O’ \O CH2C|2 O, \O O 0 To a solution of 2—amino—2—(tetrahydrofuran—3—yl)acetic acid hydrochloride (1 g, 5.52 mmol) in a mixture of THF (15 mL) and water (1.5 mL) was added t—butyl dicarbonate (1.2 g, 5.52 mmol) and sodium hydroxide (0.9 g, 22.1 mmol). The mixture was stirred at rt overnight.
Water (50 mL) was added to the mixture, followed by acidification with 2N aq. HCl solution to pH: 2. The mixture was ted with ethyl acetate (3 x 20 mL). The ed organic layers were dried over anhydrous sodium sulfate, filtered, concentrated under reduce pressure to afford 2—((tert—butoxycarbonyl)amino)—2—(tetrahydrofuran—3—yl)acetic acid (1.11 g, 80%) as a colorless oil, which was used for the next step without further purification. MS: MS (ESI) m/z 268.1180 [M+Na]+.
Ethyl 4—((tert—butoxycarbonyl)amino)—3—oxo—4—(tetrahydrofuran—3—yl)butanoate.
Procedure same as that for ethyl (S)—4—((tert—butoxycarbonyl)amino)—5—methyl—3—oxohexanoate, using 2—((tert—butoxycarbonyl)amino)—2—(tetrahydrofuran—3—yl)acetic acid as a ng material. 1H NMR (CD3OD, 400 MHz): 5 .13 (m, 3H), 4.00-3.45 (m, 4H), 2.75-2.65 (m, 1H), 2.12- 1.98 (m, 1H), 1.77-1.62 (m, 1H), 1.49 (s 9H), 1.30 (t, J: 7.2 Hz, 3H).
Ethyl 2—(((tert—butoxycarbonyl)amino)(tetrahydrofuran—3—yl)methyl)—5— chloronicotinate. Procedure same as that for ethyl (1—((tert—butoxycarbonyl)amino)—2— methylpropyl)—5—chloronicotinate, using ethyl 4—((tert—butoxycarbonyl)amino)—3—oxo—4— (tetrahydrofuran—3—yl)butanoate as a starting material. 1H NMR (CDgOD, 400 MHz): 5 8.73 (s, 1H), 8.24 (s, 1H), 5.66 (d, J: 8.0 Hz, 1H), 4.61 (s, 1H), 4.45 (q, J: 6.8 Hz, 2H), .86 (m, 1H), 3.82—3.56 (m, 3H), 2.82—2.86 (m, 1H), 1.95 (q, J: 7.2 Hz, 1H) 1.74 (q, J: 6.8 Hz, 1H), 1.44 (t, J: 7.2 Hz, 3H), 1.40 (d, J: 6.8 Hz, 9H).
Tert—butyl ((5 —chloro—3 —(hydroxymethyl)pyridin—2—yl) (tetrahydrofuran—3— yl)methyl)carbamate. Procedure same as that for tert—butyl (S)—(1—(5—chloro—3— (hydroxymethyl)pyridin—2—yl)—2—methylpropyl)carbamate, using ethyl 2—(((tert— 2015/013699 butoxycarbonyl)amino)(tetrahydrofuran—3—yl)methyl)—5—chloronicotinate as a starting material.
LCMS: tR = 0.753 min in 5—95AB_1.5min chromatography (MK RP18e 25—2mm), MS (ESI) m/z 342.9 [M+H]+.
To a solution of tert—butyl ((5—chloro—3—(hydroxymethyl)pyridin—2— yl)(tetrahydrofuran—3—yl)methyl)carbamate (600 mg, 1.74 mmol) in anhydrous CHzClz (25 mL) was added Et3N (0.5 mL, 2.62 mmol) and p—toluenesulfonyl chloride (400 mg, 2.1 mmol) at 0 OC. The mixture was stirred at rt for 2 h. The e was then washed with water (3 x 20 mL).
The organic layer was dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure and purified by chromatography column on silica gel (eluting with 10% EtOAc in petroleum ether, gradient to 20%) to afford tert—butoxycarbonyl)amino)(tetrahydrofuran—3— yl)methyl)—5—chloropyridin—3—yl)methyl 4—methylbenzenesulfonate (470 mg, 54%) as a colorless oil and tert—butyl ((5 —chloro—3—(chloromethyl)pyridin—2—yl)(tetrahydrofuran—3— yl)methyl)carbamate (200 mg, 32%) as a white solid. LCMS: tR = 0.947 min in 5—95AB_1.5min chromatography (MK RP18e 25—2mm), MS (ESI) m/z 519.1 [M+Na]+.
To a solution of tert—butyl ((5—chloro—3—(chloromethyl)pyridin—2—yl)(tetrahydrofuran— 3—yl)methyl)carbamate (470 mg, 0.95 mmol) in anhydrous DMF (5 mL) was added with sodium hydride (115 mg, 2.84 mmol, 60% in mineral oil) in portions at 0 OC. The mixture was stirred at rt for 2 h. The mixture was quenched with water (40 mL) and extracted with ethyl e (3 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered, concentrated under d pressure and purified by chromatography column on silica gel (eluting with 10% EtOAc in petroleum ether) to afford tert—butyl 3—chloro—7—(tetrahydrofuran—3— yl)—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (285 mg, 91%) as a colorless oil.
LCMS: tR = 0.862 min in 5—95AB_1.5min tography (MK RP—18e 25—2mm), MS (ESI) m/z 324.9 [M+H]+.1H NMR (CD3OD, 400 MHz): 5 8.47 (s, 1H), 7.83 (d, J: 13.6 Hz, 1H), 5.10 (s, 1H), 4.65—4.50 (m, 1.5H), 3.95 (t, J: 8.0 Hz, 0.5H), 3.85—3.67 (m, 4H), 3.03—2.87 (m, 1H), .65 (m, 2H), 1.54 (s, 9H).
To a solution of tert—butyl 3—chloro—7—(tetrahydrofuran—3—yl)—5,7—dihydro—6H— pyrrolo[3,4—b]pyridine—6—carboxylate (60 mg, 0.19 mmol) in a MW Vial in anhydrous dioxane (0.5 mL) was added molybdenum hexacarbonyl (6 mg, 0.02 mmol), (4— (ethylsulfonyl)phenyl)methanamine (56 mg, 0.28 mmol) and 1,8—diazabicyclo[5.4.0]undec—7—ene (90 mg, 0.57 mmol). The mixture was degassed with N2 for 15 min, at which point rt— butylphosphonium tetrafluoroborate (58 mg, 0.19 mmol) and trans—bis(acetate)bis[o—(di—o— tolylphosphino)benzyl]dipalladium (II) (10 mg, 0.01 mmol) were added. The Vial was sealed and heated in the MW at 160 °C for 20 min. The reaction mixture was filtered, then the filtrate was concentrated under reduced pressure and purified by preparative TLC (eluting with 12% EtOAc in petroleum ether) to afford tert—butyl (ethylsulfonyl)benzyl)carbamoyl)—7— (tetrahydrofuran—3—yl)—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (25 mg, 26%) as a colorless oil. LCMS: tR = 0.761 min in 5—95AB_1.5min chromatography (MK RPl8e 25—2mm), MS (ESI) m/z 516.2 .1H NMR (CD3OD, 400 MHz): 5 8.96 (s, 1H), 8.20 (d, J: 10.0 Hz, 1H), 8.15-8.00 (m, 1H), 7.91 (d, J: 8.4 Hz, 2H), 7.65 (d, J: 8.0 Hz, 2H), 5.19 (s, 1H), 5.00— 4.90 (m, 2H), 4.75—4.60 (m, 3H), 4.00—3.65 (m, 5H), 3.25 (q, J: 7.2 Hz, 2H), .95 (m, 1H), .75 (m, 2H), 1.56 (s, 9H), 1.23 (t, J: 7.2 Hz, 3H).
N—(4—(ethylsulfonyl)benzyl)—7—(tetrahydrofuran—3—yl)—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamide. Procedure same as that for (S)—N—(4—(ethylsulfonyl)benzyl)—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide, using tert—butyl 3—((4— (ethylsulfonyl)benzyl)carbamoyl)—7—(tetrahydrofuran—3—yl)—5,7—dihydro—6H—pyrrolo[3,4— b]pyridine—6—carboxylate as a starting material.
] (S)—6—(4—chlorobenzyl)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H— pyrrolo[3,4—[9]pyridine—3—carboxamide (9) was prepared following the synthetic route shown in Scheme 13.
Scheme 13. o O O EtO \ TFA BocHN HNbL/j/CI N/ HZNEtOfij/CI CH2C|2 N/ W N/ ; 5 /\ A /\ C| CI 0 o r ,NaH CI CI —. Q \ + Q \ THF,O°C N | N | / / N OHN H2N/\©\ CI 3023 Mo(CO)6, Hermann's cat, 0 O Fu 3 salt, DBU \ N —> N | H dioxane, 160 °C, uwave N :. 802Et /\ 9 Ethyl (S)—2—(1—amino—2—methylpropyl)—5—chloronicotinate. Procedure same as that for (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3— carboxamide, using ethyl (S)—2—(1—((tert—butoxycarbonyl)amino)—2—methylpropyl)—5— 2015/013699 chloronicotinate as a starting material. LCMS: tR = 0.59 min in in 1 min chromatography, MS (ESI) m/z 257.3 [M + H]+.
A solution of ethyl (1—amino—2—methylpropyl)—5—chloronicotinate (282 mg, 1.10 mmol) was heated in toluene (5 mL) at 100 °C for 15 h. The solvent was removed in vacuo to yield crude (S)—3—chloro—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridin—5—one, which was carried forward without further purification. LCMS: tR = 0.73 min in in 1 min chromatography, MS (ESI) m/z 211.2 [M + H]+.1H NMR (CD3OD, 400 MHz): 5 8.70 (d, J: 2.8 Hz, 1H), 8.07 (d, J: 2.0 Hz, 1H), 6.75 (broad s, 1H), 4.55 (dd, J: 1.2 Hz, 3.6 Hz, 1H), 2.49-2.41 (m, 1H), 1.23 (d, J: 7.2 Hz, 3H), 0.74 (d, J: 6.8 Hz, 3H).
To a degassed solution of (S)—3—chloro—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridin—5—one (42.2 mg, 200 umol) in THF (2 mL) at 0 °C was added sodium hydride (60% dispersion in mineral oil, 9.6 mg, 240 umol). The mixture was d at 0 °C for 30 min, at which point 4—chlorobenzyl bromide (49.3 mg, 240 umol) was added. The mixture was allowed to warm to rt and was stirred for 15 h. The mixture was ed with saturated aqueous ammonium chloride (10 mL) and extracted with EtOAc (10 mL). The organic phase was washed with brine (10 mL), dried over anhydrous MgSO4, filtered and concentrated under d pressure. The e was purified by silica gel chromatography (eluting with 5% EtOAc in hexanes, gradient to 50%) to afford (S)—3—chloro—6—(4—chlorobenzyl)—7—isopropyl—6,7—dihydro— 5H—pyrrolo[3,4—b]pyridin—5—one (19.6 mg, 29%) and 3—chloro—6—(4—chlorobenzyl)—7—hydroxy—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridin—5—one (20.1 mg, 29%). LCMS: tR = 1.09 min in in 1 min chromatography, MS (ESI) m/z 335.3 [M + H]+. 1H NMR (CDgOD, 400 MHz): 5 8.74 (d, J: 2.0 Hz, 1H), 8.17 (d, J: 2.4 Hz, 1H), 7.37-7.32 (m, 4H), 5.16 (d, J: 15.6 Hz, 1H), 4.47 (d, J: 15.2 Hz, 1H), 4.40 (d, J: 3.2 Hz, 1H), 2.56-2.48 (m, 1H), 1.27 (d, J: 7.2 Hz, 3H), 0.48 (d, J: 7.2 Hz, 3H).
General procedure F: (S)—6—(4—chlorobenzyl)—N—(4—(ethylsulfonyl)benzyl)—7— isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide (9).
SOZEt CFu'5 salt DBU6,H'ermann5 cat. wI H dioxane 160 °C uwave N 802Et ] Procedure same as that for tert—butyl 3—((4—(ethylsulfonyl)benzyl)carbamoyl)—7— (tetrahydrofuran—3—yl)—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate (Scheme 12), using (S)—3—chloro—6—(4—chlorobenzyl)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridin—5—one as a starting material. LCMS: tR = 0.92 min in in 1 min chromatography, MS (ESI) m/z 526.4 [M + H]+.1H NMR (CD3OD, 400 MHz): 5 9.22 (d, J=2.0 Hz, 1H), 8.57 (d, J: 2.0 Hz, 1H), 7.90 (dd, J: 2.0 Hz, 6.8 Hz, 2H), 7.66 (d, J: 8.8 Hz, 2H), 7.36 (m, 4H), 5.19 (d, J: 15.6 Hz, 1H), 4.73 (s, 2H), 4.48 (m, 2H), 3.20 (q, J: 7.2 Hz, 2H), 2.60-2.52 (m, 1H), 1.29 (d, J: 7.2 Hz, 3H), 1.21 (t, J: 7.2 Hz, 3H), 0.49 (d, J: 6.8 Hz, 3H).
(S)—7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxylic acid was prepared following the tic route shown in Scheme 14.
Scheme 14.
O O \ HC| FC4<3 >—\ OMe \ OMe Bra K2003 BocN I HN I / / N dioxane/MeOH N ~ :- CH3CN /\ /\ F3C F3C O O LIOH, N328203 \ OMe \ OH N | N / HZO/MeOH I N 5 N /\ /\ To a solution of 6—(tert—butyl) 3—methyl (S)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4— b]pyridine—3,6—dicarboxylate (296 mg, 0.92 mmol) in MeOH (3 mL) was added HCl (4 mL, 4.0 M in dioxane). The mixture was stirred for 30 min at rt for 30 min. The reaction mixture was concentrated to dryness to give methyl (S)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3— ylate as the HCl salt. LC-MS tR = 0.56 min in 2 min chromatography, MS (ESI) m/z 221 [M + H]+. t—butyl) 3—methyl (S)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—3,6— dicarboxylate. ure same as that for (S)—N—(4—(ethylsulfonyl)benzyl)—7—isopropyl—6—(4— (trifluoromethyl)benzyl)—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxamide, using methyl (S)—7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine—3—carboxylate as a starting material. LC- MS tR = 1.45 min in 2 min chromatography, MS (ESI) m/z 379 [M + H]+.
To a solution of 6—(tert—butyl) 3—methyl (S)—7—isopropyl—5,7—dihydro—6H—pyrrolo[3,4— b]pyridine—3,6—dicarboxylate (216 mg, 674 umol) in MeOH (4 mL) was added 10% s NaZSZOg (3 drops) and aqueous lithium hydroxide (1.2 mL, 2.0 M). The mixture was stirred for 3 h at rt. The reaction mixture was then concentrated, and the residue was ed by preparative HPLC to give 230 mg of (S)—7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxylic acid as the TFA salt. LC-MS tR = 1.22 min in 2 min chromatography, MS (ESI) m/z 365 [M + H]+. 1H NMR (CD3OD, 400 MHz): δ 9.14, (s, 1H), 8.35 (s 1H), 7.84-7.81 (m, 4H), 4.81-4.58 (m, 5H), 2.38 (m, 1H), 1.13 (d, J = 6.8 Hz, 3H), 0.91 (d, J = 6.8 Hz, 3H).
General procedure G: tert-butyl (S)isopropyl((4- (methoxycarbonyl)benzyl)carbamoyl)-5,7-dihydro-6H-pyrrolo[3,4-b]pyridinecarboxylate.
O O H2N HATU, i-Pr2NEt OH N BocN + OMe BocN H N DMF N Tert-butyl isopropyl((4-(methoxycarbonyl)benzyl)carbamoyl)-5,7- dihydro-6H-pyrrolo[3,4-b]pyridinecarboxylate. Procedure same as that for tert-butyl (S) ((4-(ethylsulfonyl)benzyl)carbamoyl)isopropyl-5,7-dihydro-6H-pyrrolo[3,4-b]pyridine carboxylate, using methyl 4-(aminomethyl)benzoate as a starting material. LC-MS tR = 1.61 min in 2 min chromatography, MS (ESI) m/z 454. ((7-isopropyl(4-(trifluoromethyl)benzyl)-6,7-dihydro-5H-pyrrolo[3,4- b]pyridinecarboxamido)methyl)benzoic acid was prepared ing the synthetic route shown in Scheme 15.
Scheme 15.
O O N HCl N BocN H HN H O O N dioxane/MeOH N O O N H O 65 N H N 147 Methyl (S)((7-isopropyl-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine carboxamido)methyl)benzoate. Procedure same as that for methyl (S)isopropyl-6,7- dihydro-5H-pyrrolo[3,4-b]pyridinecarboxylate, using tert-butyl (S)isopropyl((4- (methoxycarbonyl)benzyl)carbamoyl)—5,7—dihydro—6H—pyrrolo[3,4—b]pyridine—6—carboxylate as a starting material. LC-MS tR = 0.73 min in 2 min chromatography, MS (ESI) m/z 354.
Methyl (S)—4—((7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro—5H— pyrrolo[3,4—b]pyridine—3—carboxamido)methyl)benzoate (65). Procedure same as that for (S)—N— (4—(ethylsulfonyl)benzyl)—7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamide, using methyl (S)—4—((7—isopropyl—6,7—dihydro—5H—pyrrolo[3,4— b]pyridine—3—carboxamido)methyl)benzoate as a starting al. LC-MS tR = 1.34 min in 2 min chromatography, MS (ESI) m/z 512 [M + H]+.
General procedure H: (S)—4—((7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7— dihydro—SH—pyrrolo[3,4—b]pyridine—3—carboxamido)methyl)benzoic acid (147). Procedure same as that for (S)—7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro—5H—pyrrolo[3,4—b]pyridine— 3—carboxylic acid, using methyl (S)—4—((7—isopropyl—6—(4—(trifluoromethyl)benzyl)—6,7—dihydro— SH—pyrrolo[3,4—b]pyridine—3—carboxamido)methyl)benzoate as a starting material. LC-MS tR = 1.20 min in 2 min chromatography, MS (ESI) m/z 498 [M + H]+.
The following compounds in Table 1 were prepared ing to the methods described .
Table 1.
Cmpd 1 Structure LCMS H-NMR Starting Material (CD30D) 6 9.36 (t, J = 1.2 Hz, 1H), 9.05 (s, 1H), 8.23 (s, 1H), 7.89 (d, J = 8.4 Hz, 2H), 7.63 Q (d, J = 8.4 Hz, 2H), o 7.59 (d, J = 8.4 Hz, 2H), 7.52 (d, J = 3 \ ”U N l 8.4 Hz, 2H), 4.90 , H)+ ,s'\/ (m, 1H), 4.74 (m, /7\ 0' 4H), 4.57 (s, 2H), 3.20 (q, J = 7.6 Hz, 2H), 2.42-2.29 (m, 1H), 1.21 (t, J = 7.6 Hz, 3H), 1.13 (d, 6.8 Hz, 3H), 0.90 d, J = 6.8 Hz, 3H. (00300) 6 9.36 (t, J = 1.2 Hz, 1H), 9.03 (s, 1H), 8.21 (s, 1H), 7.88 (d, J = 8.8 Hz, 2H), 7.86 o (d, J = 8.4 Hz, 2H), 4 503.5 7.78 (d, J = 8.8 Hz, N (M+H)+ 2H), 7.63 (d,J= N’ ”D7 o., 8.4 Hz, 2H), 4.84 /\ 0" (d, J = 15.6 Hz, 1H), 4.71 (m, 3H), 4.59 (m, 3H), 3.20 (q, J = 7.6 Hz, 2H), 2.43-2.32 m, 1H , 1.21 (t, J = 7.2 Hz, 3H), 1.12 (d, 6.8 Hz, 3H), 0.92 (d, J = 6.8 Hz, 3H (CD30D) 6 9.36 (t, J = 1.2 Hz,1H), 9.04 (s, 1H), 8.23 (s, 1H), 7.89 (d, J = 8.8 Hz, 2H), 7.64 (d, J = 8.4 Hz, 4H), 7.27 (d, J = 8.4 Hz, 544.4 2H), 6.92 (t, J = 73.6 Hz, 1H), 4.90 (M+H)+ (m, 2H), 4.72 (m, 3H), 4.56 (m, 2H), 3.20 (q, J = 7.6 Hz, 2H), 2.40-2.29 (m, 1H),1.21 (t, J = 7.6 ,1.12(d, 6.8 Hz, 3H), 0.90 d, J = 6.8 Hz, 3H.
(CD30D) 6 9.06 (s, 1H), 8.26 (s, 1H), 7.89 (d, J = 8.4 Hz, 2H), 7.66 (m, 4H), 7.26 (d, J = 8.4 Hz, 2H), 4.96 (d, J = .6 Hz, 1H), 4.82 (m, 2H), 4.73 (d, J 496.5 = 6.0 Hz, 2H), 4.64 (M+H)+ (d, J = 13.2 Hz, 1H), 4.59 (d, J = 13.2 Hz, 1H), 3.20 (q, J = 7.6 Hz, 2H), 2.42-2.33 (m, 1H), 1.21 (t, J = 7.2 Hz, 3H), 1.12 (d, 6.8 Hz, 3H), 0.87 (d, J = 6.8 Hz, 3H (CD30D) 6 8.97 (d, J = 2.0 Hz,1H), 8.68 (s, 2H), 8.25 (t, J = 0.8 Hz, 1H), 7.89 (dd, J = 1.6 Hz, 8.4 Hz, 2H), 7.65 (d, J = 8.4 Hz, 2H), 5.39 (dd, J = 2.0 Hz, 3.2 Hz, 1H), 534.4 .15 (d, J = 16.4 (M+H)+ Hz, 1H), 4.88 (d, J =16.4 Hz, 1H), 4.72 (s, 2H), 3.20 (q, J = 7.2 Hz, 2H), 2.80-2.71 (m, 1H), 1.22 (d, J = 6.8 Hz, 3H), 1.21 (t, J = 7.2 Hz, 3H), 0.64 (d, J = 6.8 Hz, 3H W0 2015/116904 (CD30D) 6 9.02 (d, J = 7.6 Hz,1H), 8.99 (s, 1H), 8.26 (s, 1H), 7.90 (dd, J = 2.0 Hz, 6.8 Hz, 2H), 7.65 (d, J = 8.8 Hz, 2H), 5.44 (d, J = 9.2 Hz,1H), F F 5.22 (dd, J = 10.0 F o Hz, 16.8 Hz, 1H), o 4.93 (d, J = 16.4 mom”N 606.6 Hz, 1H), 4.72 (s, /‘0 ‘N .. N/ (M+H)+ s 2H), 4.36 (q, J = ’\ 7.2 Hz, 2H), 3.20 (q, J = 7.6 Hz, 2H), 2.81-2.70 (m, 1H), 1.37 (t, J = 7.2 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H), 0.88 (dd, J = 7.2 Hz, 13.2 Hz, 3H), 0.69 (dd, J = 7.2 Hz, 17.6 Hz, (CD30D) 6 9.23 (d, J :24 Hz, 1H), 8.53 (d, J = 2.4 Hz, 1H), 7.90 (dd, J = 2.0 Hz, 6.8 Hz, 2H), 7.65 (d, J = 8.4 Hz, CI 2H), 7.49 (d, J = 8.4 Hz, 2H), 7.32 0 O (dd, J = 2.0 Hz, 6.8 11 \ 542.5 Hz, 2H), 4.80 (d, J N | (M+H)+ = 14.0 Hz, 1H), 4.73 (m, 2H), 4.61 (d, J = 15.2 Hz, 1H), 3.20 (q, J = 7.2 Hz, 2H), 2.48- 2.41 (m,1H),1.27 (d, 6.8 Hz, 3H), 1.20 (t, J = 7.2 Hz, 3H), 0.39 (d, J = 7.2 Hz, 3H .
(CD30D) 6 9.02 (s, 1H), 8.75 (s, 1H), 8.40 (s, 1H), 7.90 (dd, J = 1.6 Hz, 6.8 Hz, 2H), 7.66 (d, J F F = 8.4 Hz, 2H), 5.51 F o (t, J = 2.8 Hz, 1H), N (d, J =16.4 5.21 12 woo” 564.5 Hz, 1H), 4.93 (m, Reduction of 10 HO —N N’ (M+H)+ 1H), 4.73 (s, 2H), ’\ 4.67 (s, 2H), 3.20 (q, J = 7.2 Hz, 2H), 2.78-2.66 (m, 1H), 1.21 (t, J = 7.2 Hz, 3H), 1.17 (d, J = .6 Hz, 3H), 0.78 d, J = 5.6 Hz, 3H.
(CD30D) 6 9.09 (d, F F o J: 2.0 Hz, 1H), F Oxidation of 8.93 N (s, 1H), 8.59 12 followed by 14 / \FNCEYL 578.5 (s, 1H), 7.90 (dd, J HO —N N’ (M+H)+ :16 Hz, 6.8 Hz, methyl 2H), 7.67 (d, J = rd /\ 8.4 Hz, 2H), 5.64 addition dd, J = 3.2 Hz, 7.0 2015/013699 Hz, 1H), 5.28 (d, J = 16.4 Hz, 1H), .13 (q, J = 6.4 Hz, 1H), 4.95 (d, J = .6 Hz, 1H), 4.74 (s, 2H), 3.20 (q, J = 7.6 Hz, 2H), 2.76- 2.66 (m, 1H),1.47 (d, J = 6.4 Hz, 3H), 1.21 (t, J = 7.2 Hz, 3H), 1.13 (d, 6.8 Hz, 3H), 0.81 (dd, J = 6.8 Hz, 3H (CD30D) 6 9.39 (brs, 1H), 9.09 (s, 1H), 8.28 (s, 1H), 7.96 CI (d, J: 8.4 Hz, 2H), 0 7.70-7.60 (m, 4H), 7.56 (d, J: 8.0 Hz, 17 \ 498.0 N I, 00 2H), 5.05-4.95 (m, . (M+H)+ N 2H), 4.75-4.60 (m, s 0&0 /\ 5H), 3.13 (s, 3H), 2.39-2.35 (m, 1H), 1.16 (d, J: 6.8 Hz, 3H), 0.90 (d, J= 6.8 Hz, 3H .
(CD30D) 6 9.08 (d, J = 1.6 Hz, 1H), 8.27 (s, 1H), 7.96 (dd, J CI =1.6,6.8 Hz, 2H), 7.67-7.63 (m, 4H), 7.55 (d, J: 8.4 Hz, 18 498.0 2H), 5.10-4.95 (m, (M+H)+ 2H), 4.75-4.60 (m, 5H), 3.13 (s, 3H), 2.38-2.34 (m, 1H), 1.14 (d, J: 6.8 Hz, 3H), 0.90 (d, J= 7.2 Hz, 3H .
(CD30D) 6 9.07 (d, J :16 Hz, 1H), 8.27 (s, 1H), 7.96 (dd, J = 2.0, 6.8 Hz, 2H), 7.88 (dd, J: 8.8, 22.4 Hz, 4H), 7.65 19 489.1 (d, J: 8.4 Hz, 2H), (M+H)+ 4.85-4.65 (m, 7H), N I:”\ 3.13 (s, 3H), 2.42- O 2.37(m,1H),1.16 (d, J: 6.8 Hz, 3H), 0.91 (d, J: 6.8 Hz, 3H .
(CD30D) 6 9.08 (s, 1H), 8.30 (s, 1H), F F 7.95 (d, J: 8.4 Hz, 2H), 7.91 (s, 4H), 7.66 (d, J: 8.4 Hz, 2H), 5.07 (d, J= 489.0 \ 15.6 Hz, 1H), 4.90- , N —N | (M+H)+ / H/\©\ p 4.70 (m, 3H), 4.72 g N .,3\/ (s, 3H), 3.13 (s, /\ 0 3H), 2.42-2.37 (m, 1H),1.16(d,J= 6.8 Hz, 3H), 0.90 (d, J: 6.8 Hz, 3H).
(CD30D) 6 9.08 (s, 1H), 8.30 (s, 1H), 7.95 (d, J= 8.4 Hz, 2H), 7.91 (s, 4H), 7.66 (d, J= 8.4 Hz, 2H), 5.07 (d, J= 21 489.0 .6 Hz, 1H), 4.90- (M+H)+ 4.70 (m, 3H), 4.72 (s, 3H), 3.13 (s, 3H), 2.42-2.37 (m, 1H), 1.16 (d, J: 6.8 Hz, 3H), 0.90 d, J= 6.8 Hz, 3H.
(CD3OD) 5 9.09 (d, J = 2.0 Hz, 1H), 8.30 (s, 1H), 7.95 (d, J: 8.0 Hz, 2H), 8.88 (q, J= 8.4 Hz, 4H), 7.66 (d, J= 8.4 Hz, 22 532.1 2H), 5.04 (d, J= (M+H)+ .2 Hz, 2H), 4.85- N as: 4.70 (m, 5H), 3.13 o 0 (s, 3H), 2.41-2.37 (m, 1H), 1.17 (d, J = 6.8 Hz, 3H), 0.90 d, J= 6.8 Hz, 3H .
(CD30D) 5 9.08 (s, 1H), 8.71 (cl, J: 2.4 Hz, 1H), 8.32 (s, 1H), 8.00 (dd, J: 2.4, 8.4 Hz, 1H), 7.95 (d, J= 8.4 Hz, O 2H), 7.66 (d, J: \ / 8.4 Hz, 2H), 7.62 23 \ 499.1 N I (d, J= 8.8 , / H / (M+H)+ .15 N d’S‘b (d, J= 16.0 /\ Hz, 2H), 4.85-4.75 (m, 3H), 4.73 (s, 2H), 3.13 (s, 3H), 2.62-2.54 (m, 1H), 1.22 (d, J= 7.2 Hz, 3H), 0.96 (d, J= 6.8 Hz, 3H .
(CD3OD) 5 9.08 (d, J = 1.6 Hz, 1H), 8.31 (s, 1H), 7.95 (d, J: 8.4 Hz, 2H), 7.75 F (dd, J= 4.8, 8.4 Hz, 2H), 7.66 (d, J= o 8.4 Hz, 2H), 7.28 (t, 24 \ 482.1 N J= 8.4 Hz, 2H), N I, 90, (M+H)+ 5.10-5.03 (m, 2H), i 093.0 4.90-4.70 (m, 4H), /\ 4.59 (d, J= 12.8 Hz, 1H), 3.13 (s, 3H), 2.37-2.30 (m, 1H), 1.11 (d, J: 6.4 Hz, 3H), 0.87 d, J= 6.8 Hz, 3H .
CI (CD30D) 6 8.96 (s, 1H), 8.22 (d, J : 11.6 Hz, 1H), 7.89 o (d, J = 8.8 Hz, 2H), 556.6 o \ N 7.64 (d, J = 8.8 Hz, I HUS) H (M+H)+ , 2H), 7.42 (d, J : O N s ..S\/ 8.8 Hz, 2H), 7.38 /\ 0 (d, J = 8.8 Hz, 2H), 2015/013699 (m, 1H), 4.85 (m, 2H), 4.71 (s, 2H), 3.20 (q, J = 7.2 Hz, 2H), 2.58-2.39 (m, 1H),1.21 (t, J = 7.2 Hz, 3H), 1.03 (dd, 6.8 Hz, 16.8 Hz, 3H), 0.75 (dd, J = 6.8 Hz, 18.8 Hz, (CD30D) 6 9.30 (s, 2H), 9.09 (s, 1H), 8.30 (s, 2H), 7.89 F F (dd, J = 2.0 Hz, 8.4 F”H Hz, 2H), 7.65 (d, J = 8.4 Hz, 2H), 4.95 26 NL<N (m, 3H), 4.82 (m, 548.5 2H), 4.71 (s, 2H), l (M+H)+ 3.20 (q, J = 7.2 Hz, 2H), 2.62-2.53 (m, «1 1H), 1.25 (d, J = 6.8 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H), 0.98 (d, J = 6.8 Hz, 3H .
(CD30D) 6 9.08 (d, J = 1.6 Hz,1H), 9.03 (s, 1H), 8.32 (d,J=1.6 Hz,1H), 8.26 (dd, J = 2.0 Hz, 8.0 Hz, 1H), 7.89 (d, J = 8.4 Hz, 2H), 7.77 (d, J = 8.0 Hz, 1H), 7.65 27 N/ 547.5 (d, J = 8.4 Hz, 2H), (M+H)+ 5.18 (d, J = 15.6 Hz, 1H), 4.97 (s, 2H), 4.92 (m, 2H), 4.72 (s, 2H), 3.20 (q, J = 7.6 Hz, 2H), 2.68-2.59 (m, 1H), 1.24 (d, J = 7.2 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H), 0.96 (d, J = 6.8 Hz, 3H (CD30D) 6 9.11 (s, 1H), 8.32 (s, 1H), 7.90 (d, J = 8.8 Hz, 2H), 7.65 (d, J = CI 8.4 Hz, 2H), 7.59 (m, 4H), 5.25-4.83 (m, 2H), 4.73 (s, 28 526.3 2H), 4.62 (m, 1H), (M+H)+ 4.15 (m, 1H), 3.20 (q, J = 7.2 Hz, 2H), 2.21-2.10 (m, 1H), 1.47 (d, J = 6.4 Hz, 3H), 1.21 (t, J = 7.2 Hz, 3H), 0.95 (m, 3H), 0.79 (d, J = 6.8 Hz, 3H .
(CD30D) 6 9.07 (d, J = 2.0 , 8.22 (s, 1H), 7.89 (dd, J = 2.0 Hz, 6.8 Hz, 2H), 7.71 (d, J = 6.8 Hz, 2H), 7.64 CI (d, J = 8.0 Hz, 2H), 7.57 (m, 2H), 5.21 0 (q, J = 6.8 Hz,1H), 29 \ N 526.3 4.99 (d, J = 3.2 Hz, N | , HUD (M+H)+ 1H), 4.82 (d, J = s‘ N ,S\/ 3.6 Hz, 1H), 4.72 /\ O (m, 3H), 3.20 (q, J = 7.2 Hz, 2H), 2.16- 2.09(m,1H),1.75 (d, J = 6.8 Hz, 3H), 1.29 (d, J = 6.8 Hz, 3H), 1.21 (t, J = 7.2 Hz, 3H), 0.86 (d, J = 6.8 Hz, 3H (CD30D) 6 9.11 (s, 1H), 8.32 (s, 1H), 7.87 (m, 6H), 7.66 F F (d, J = 8.0 Hz, 2H), .35 (m, 1H), 4.83 (m, 1H), 4.73 (s, E 2H), 4.63 (m, 1H), . 560.6 \ N 4.28 (m, 1H), 3.21 N | (M+H)+ / H/\©\,Q (q, J = 7.2 Hz, 2H), : N ,,S\/ 2.23-2.11 (m, 1H), /\ o 1.97 (d, J = 6.8 Hz, 3H), 1.21 (t, J = 7.2 Hz, 3H), 0.97 (m, 3H), 0.80 (d, J = 6.8 Hz, 3H .
(CD30D) 6 9.08 (d, J = 1.2 Hz,1H), 8.24 (s, 1H), 7.95 (d, J = 8.4 Hz, 2H), F F 7.89 (m, 4H), 7.64 (d, J = 8.8 Hz, 2H), .24 (m, 1H), 5.01 o (d, J = 2.8 Hz,1H), 31 560.6 \ N 4.88 (m, 2H), 4.71 N ' (M+H)+ , an (s, 2H), 3.20 (q, J = i N 55¢ 7.6 Hz, 2H), 2.18- /\ 0 2.10(m,1H),1.75 (d, J = 6.8 Hz, 3H), 1.28 (d, J = 6.8 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H), 0.87 (d, J = 6.8 Hz, 3H (CD30D) 6 9.23 (d, J = 2.0 , 8.59 (d, J: 2.4 Hz, 1H), 7.90 (d, J = 8.8 Hz, 2H), 7.65 FF (m, 4H), 7.55 (d, J = 8.8 Hz, 2H), 5.26 O o (d, J = 15.6 Hz, 32 560.5 1H), 4.74 (s, 2H), \ N N I/ H (M+H)+ 4.63 (d, J = 16.4 N Hz, 1H), 4.53 (d, J /\ = 3.2 Hz, 1H), 3.20 (q, J = 7.2 Hz, 2H), 2.60-2.52 (m, 1H), 1.30 (d, J = 6.8 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H), 0.50 (d, J = 6.8 Hz, 3H (CD30D) 6 9.12 (s, 1H), 9.10 (d, J: 1.2 Hz, 1H), 8.53 (dd, J = 2.0 Hz, 8.4 Hz, 1H), 8.34 (s, 1H), FF 7.93 (d, J = 8.0 Hz, F 2H), 7.91 (d, J = 6.4 Hz, 1H), 7.84 O (d, J = 8.0 Hz, 2H), 33 547.5 \ N 5.07 (d, J =15.6 N l/ H (M+H)+ Hz, 1H), 4.84 (m, : N 3H), 4.73 (d, J = /\ 12.8 Hz, 1H), 3.35 (q, J = 7.2 Hz, 2H), 2.42-2.29 (m, 1H), 1.28 (t, J = 7.2 Hz, 3H), 1.15 (d, J = 6.8 Hz, 3H), 0.89 d, J = 6.8 Hz, 3H.
(CD30D) 6 9.13 (dd, J: 1.6 Hz, 3.6 Hz, 2H), 8.59 (dd, J = 2.0 Hz, 8.4 Hz, 1H), 8.35 (s, 1H), 7.98 (d, J = 8.0 Hz, 1H), 7.70 (d, J = CI 8.8 Hz, 2H), 7.54 0 (d, J = 8.8 Hz, 2H), .07 (d, J = 15.2 34 \ 513.5 N l/ Hz, 1H), 4.87 (m, H (M+H)+ N 2H), 4.73 (d, J = 13.2 Hz, 1H), 4.60 (d, J =12.8 Hz, 1H), 3.37 (q, J = 7.6 Hz, 2H), 2.41- 2.25 (m, 1H),1.29 (t, J = 7.6 Hz, 3H), 1.12 (d, J = 6.4 Hz, 3H), 0.88 (d, J = 6.8 Hz, 3H .
(CD30D) 6 9.11 (d, J: 2.0 Hz, 2H), 8.56, (dd, J = 2.0 Hz, 8.4 Hz, 1H), N\\ 8.33 (s, 1H), 8.01- 7.79 (m, 5H), 5.07 (d, J = 15.2 Hz, 504.5 1H), 4.91-4.80 (m, NI, (M+H)+ 3H), 4.72 (d, J = 14.4 Hz, 1H), 3.36 : N /\ (q, J = 7.2 Hz, 2H), 2.64-2.53 (m, 1H), 1.28 (t, J = 7.2 Hz, 3H), 1.15 (d, J = 6.8 Hz, 3H), 0.89 d, J = 7.2 Hz, 3H.
(CD30D) 5 9.09 (s, 1H), 9.04 (s, 1H), 8.33 (s, 1H), 8.28 (d, J: 8.0 Hz, 2H), 7.95 FF (d, J: 8.4 Hz, 2H), 7.80 (d, J: 8.0 Hz, 2H), 7.66 (d, J: 36 N/ 533.1 8.4 Hz, 2H), 5.20 NI/ (M+H)+ (d, J: 15.6 Hz, 1H), 5.25-4.93 (m, s N /\ 4H), 4.73 (s, 2H), 3.13 (s, 3H), 2.68- 2.63 (m, 1H), 1.25 (d, J: 6.8 Hz, 3H), 0.98 (d, J: 6.8 Hz, 3H .
(CD3OD) 5 9.43 (t, J = 6.0 Hz, 1H), 9.07 (d, J: 18.8 Hz, FF 2H), 8.33-8.25 (m, F 2H), 7.96 (d, J: 8.4 Hz, 2H), 7.76 \ (d, J: 8.0,1H), 37 N/ 533.0 \ 7.66 (d, J: 8.4 Hz, NI/ (M+H)+ 2H), 5.20-4.92 (m, N 5H), 4.73 (d, J: 6.0, 2H), 3.13 (s, 3H), .61 (m, 1H),1.25(d,J= 7.2 Hz, 3H), 0.98 d, J: 6.8 Hz, 3H.
(CD30D) 5 9.08 (s, 1H), 8.31 (s, 1H), 8.26 (d, J= 8.0 Hz, 2H), 7.90 (d, J= 8.0 Hz, 2H), 7.86 (d, J= \N.N‘.N 8.0 Hz, 2H), 7.65 N‘ (d, J= 8.0 Hz, 2H), .10 (d, J= 15.2 O Hz, 1H), 4.99-4.96 38 560.1 \ (m, 1H), 4.85-4.77 N (M+H)+ N I H (m, 2H), 4.73-4.67 s: N (m, 3H), 4.46 (s, /\ 3H), 3.22 (q, J: 7.6 Hz, 2H), 2.38- 2.32 (m, 1H), 1.22 (t, J= 7.6 Hz, 3H), 1.14 (d, J= 6.4 Hz, 3H), 0.90 (d, J= 6.8 Hz, 3H . (c0300) 5 9.10 (d, J = 2.0 Hz, 1H), 9.05 (s, 1H), 8.46 (dd, J = 2.0, 8.0 Hz, 1H), F F 8.31 (s, 1H), 7.99 (d, J= 8.0 Hz,1H), O 7.91 (d, J= 8.4 Hz, \ / 39 547.1 2H), 7.66 (d, J: \ N N I (M+H)+ 8.0 Hz, 2H), 5.06- / 4.98 (m, 2H), 4.86- :5 N 4.81 /\ (m, 3H), 4.73 (s, 2H), 3.22 (q, J: 7.6 Hz, 2H), 2.52- 2.48 (m, 1H),1.23- 1.19 (m, 6H), 0.94 (d, J= 6.8 Hz, 3H).
(CD30D) 5 9.08 (s, 1H), 8.99 (s, 1H), 8.36 (d, J= 7.6 Hz, 1H), F F 8.26 (s, 1H), 7.98 F (d, J= 8.0 , —N 7.91 (d, J= 8.0 Hz, 40 \ / 547.1 2H), 7.66 (d, J= \ N 8.4 Hz, 2H), 5.01- N I (M+H)+ I H 4.96 (m, 1H), 4.82- N 4.68 (m, 6H), 3.22 (q, J: 7.2 Hz, 2H), 2.52-2.48 (m, 1H), 1.23-1.19 (m, 6H), 0.47 (d, J= 6.8 Hz, (CD30D) 6 9.09 (d, J= 1.6 Hz,1H), 8.30 (d, J= 1.2 Hz,, 1H), 7.91 (d, J: 8.4 Hz, 2H), 7.67 FF (d, J= 8.4 Hz, 2H), F 5.14 (d, J= 14.4 O Hz, 1H), 4.74 (s, 41 552.1 3H), 3.52 (d, J: \ N N I, 90 (M+H)+ 5.6 Hz, 2H), 3.22 s N 55 (q, J: 7.6 Hz, 2H), O 2.54-2.52 (m, 1H), 2.22-2.19 (m, 2H), 1.83-1.60 (m, 8H), 1.33 (d, J= 6.8 Hz, 4H), 1.22 (d, J: 7.2 Hz, 3H), 1.11 d, J= 6.4 Hz, 3H. ) 6 9.08 (s, 1H), 8.29, (s, 1H), 7.90 (d, J = 7.6 Hz, 2H), 7.65 (d, J = 7.6 Hz, 2H), 5.16 (d, J =14.4 Hz, FF 1H), 4.73 (d, J : 14.4 Hz, 1H), 4.72 0 (s, 2H), 3.41 (m, 42 \ N 1H), 3.20 (q, J : 520.6 N | (M+H)+ 7.2 Hz, 2H), 2.58- N H/\©\O : "3 2.47 (m, 1H), 2.20- /\ o 2.06 (m, 3H), 2.05- 1.95 (m, 4H), 1.92- 1.8 (m,2H),1.58- 1.32 , 2H), ( ) (s, 1H), 9.00, (d, J : 2.0 Hz, 1H), 8.32 (s, 1H), 8.30 (d, J = 2.0 Hz, 1H), 7.72 (d, J = 8.4 Hz,1H), .17 (d, J = 15.6 FF Hz, 1H), 4.83 (s, 0 2H), 4.74 (d, J = 14.8 Hz, 1H), 3.46 43 \ \ 521.5 N l, ' (m, 1H), 3.30 (q, J H O N/ (M+H)+ . = 7.6 Hz, 2H), 2.60- s: N 2.49 (m, 1H), 2.20- /\ o 2.06 (m, 3H), 2.05- 1.95 (m, 4H), 1.92- 1.82 (m, 2H), 1.58- 1.41 (m,2H),1.33 (d, J = 6.8 Hz, 3H), 1.27 (t, J = 7.6 Hz, 3H), 1.10 (d, J = 6.8 Hz, 3H . ) 5 9.08 (s, 1H), 8.28 (s, 1H), 7.90 (d, J= 8.4 Hz, 2H), 7.65 (d, J= 8.4 Hz, 2H), 5.16-5.05 (m, 1H), 4.72 (s, 3H), 3.68 (s, 3H), 3.44- 44 542.1 3.29 (m, 2H), 3.21 (M+H)+ (q, J: 7.6 Hz, 2H), 2.52-2.33 (m, 2H), 2.13-1.83 (m, 6H), 1.57-1.44 (m, 2H), 1.31 (d, J= 6.4 Hz, 4H), 1.21 (t, J= 7.6 Hz, 4H), 1.09 (d, J = 6.4 Hz, 3H (CD30D) 6 9.42 (t, J= 6.0 Hz,1H), 9.10 (s, 1H), 8.30 (s, 1H), 7.91 (d, J: 8.4 Hz, 2H), 7.66 (d, J= 8.4 Hz, 2H), .13-5.08 (m, 2H), 4.77-4.62 (m, 4H), 45 \ 514.2 N 3.41-3.37 (m, N I ), / H (M+H)+ 3.26-3.17 (m, N ), 2.56-2.48 (m, ) 2.21-2.13 (m, ), 2.03-1.92 (m, 3H), 1.32 (d, J= 7.2 Hz, 3H), 1.28-1.19 (m, 6H), 1.10 (d, J: 6.8 Hz, 3H . (c0300) 5 9.24 (d, J = 2.0 Hz, 1H), 8.99 (d, J= 1.6 , 8.55 (d, J= 2.0 Hz, 1H), 8.27 (dd, J: 2.4, 8.4 Hz, 1H), F F 7.69 (d, J= 8.4 Hz, F 1H), 4.83 (s, 2H), 4.71 (d, J= 3.2 Hz, O O 1H), 3.88 (dd, J: 47 567.53 "a \ N 9.2, 13.8 Hz, 1H), _N I H (M+H)+ / 3.30 (q, J: 7.6 Hz, s“ 2H), 3.16 (dd, J: ’\ 5.2, 13.8 Hz, 1H), 2.62-2.58 (m, 1H), 2.33-1.71 (m, 6H), 1.38 (d, J= 6.8 Hz, 3H), 1.35-1.04 (m, 4H), 1.25 (t, J= 7.6 Hz, 3H), 0.53 (d, J = 6.8 Hz, 3H (CD30D) 5 9.01 (s, 1H), 8.21 (s, 1H), 7.86 N (dd, J= 8.4, 10.4 \\Q Hz, 4H), 7.76 (d, J = 8.0 Hz, 2H), 7.62 (d, J= 8.4 Hz, 2H), 48 \ N 531.1 4.75-4.50 (m, 6H), N | H (M+H)+ 4.00-3.68 (m, 5H), 3.18 (q, J: 7.6 Hz, 2H), 3.00-2.90 (m, 1H), 2.10-2.04 (m, 1H), 1.98-1.85 (m, 1H), 1.19 (t, J= 7.6 Hz, 3H .
(CD30D) 5 9.08 (s, 1H), 8.30 (s, 1H), 7.89 (d, J: 8.4 Hz, 2H), 7.65 (d, J= 8.4 Hz, 2H), 4.72 (s, 2H), 4.12-4.05 (m, 2H), 3.65-3.60 (m, 2H), 3.50-3.40 49 QN| 567.1 (m, 2H), 3.30-3.25 (M+H)+ (m, 5H), 3.20 (q, J = 7.2 Hz, 2H), 2.60- 2.50 (m, 1H), 2.34- 2.20(m,3H),1.82- 1.73 , 1.32 (d, J: 7.2 Hz, 3H), 1.21 (t, J: 7.6 Hz, 3H),1.15-1.00(m, 3H .
(CD30D) 5 8.98 (s, 1H), 8.17 (s, 1H), 7.85 N (dd, J: 8.4, 14.4 b Hz, 4H), 7.76 (d, J = 8.0 Hz, 2H), 7.62 (d, J: 8.4 Hz, 2H), 50 \ 531.1 4.80-4.40 (m, 6H), N I (M+H)+ 4.00-3.65 (m, 5H), 3.18 (q, J: 7.6 Hz, 2H), 3.00-2.88 (m, 1H), 2.25-2.13 (m, 1H), 2.13-2.00 (m, 1H), 1.19 (t, J: 7.6 Hz, 3H .
(CD30D) 6 9.00 (s, 1H), 8.19 (s, 1H), Nb; 7.84 (t, J: 8.0 Hz, 4H), 7.78 (d, J= 8.0 Hz, 2H), 7.62 (d, J: 8.0 Hz, 2H), 51 \ 531.1 4.80-4.45 (m, 6H), N I (M+H)+ 4.00-3.65 (m, 5H), 3.18 (q, J: 7.6 Hz, 2H), 3.00-2.88 (m, 1H), 2.27-2.13 (m, 1H), 2.10-1.98 (m, 1H),1.19(t,J= 7.6 Hz, 3H .
(CD30D) 5 8.99 (s, 1H), 8.19 (s, 1H), 7.85 (dd, J: 8.0, 17.6 Hz, 4H), .70 (m, 2H), 7.62 (d, J = 8.4 Hz, 2H), 4.70- 52 531.1 4.35 (m, 6H), 3.95- (M+H)+ 3.65 (m, 5H), 3.18 (q, J: 7.6 Hz, 2H), 3.00-2.88 (m, 1H), 2.12-2.06 (m, 1H), 2.00-1.87 (m, 1H), 1.19 (t, J: 7.6 Hz, (CD30D) 5 9.12 (s, 1H), 9.01 (cl, J: 2.0 Hz, 1H), 8.36-8.32 (m, 2H), 7.75 (d, J = 8.0 Hz, 1H), 5.16 FF (d, J= 15.6 Hz, 1H), 4.89-4.87 (m, 2H), 4.73 (d, J: .2 Hz, 1H), 3.42- 53 553.1 a, \ N \ 3.34 (m, 3H), 3.31- ‘N I (M+H)+ , Hmr 3.28 (m, 2H), 2.58- N ”3:0 2.51 (m, 1H), 2.20- 0 1.90 (m, 6H) 1.50- 1.39 , 1.33 (d, J= 7.2 Hz, 3H), 1.26 (t, J= 7.2 Hz, 3H),1.24-1.20 (m, 2H), 1.10 (d, J: 6.8 Hz, 3H .
(CD30D) 5 9.11 (s, 1H), 9.01 (s, 1H), 8.35 (dd, J = 2.0, 8.4 Hz, 1H), 8.31 (s,1H), 7.75 (d, J= 8.4 Hz, 1H), 7.62 (d, J: 1.2 Hz, 1H), 7.50 F (dd, J= 1.6, 8.5 Hz, 1H), 7.37 (d, J: 8.0 Hz, 1H), 5.01 54 559.50 \ N \ (d,J=15.6 Hz, (M+H)+ 1H), 4.84 (s, 2H), 4.68 (dd, J= 12.8, /\ 00 24.0 Hz, 2H), 3.34 (s, 2H), 3.30 (q, J: 7.6 Hz, 2H), 2.38 (broad s, 1H), 1.25 (t, J= 7.6 Hz, 3H), 1.16 (d, J= 6.0 Hz, 3H), 0.90 (d, J: 6.8 Hz, 3H .
(CD30D) 6 8.60 (s, 1H), 8.33 (s, 1H), 7.89 (d, J = 8.4 Hz, 2H), 7.64 (d, J : 8.4 Hz, 2H), 4.95 F F (m, 1H), 4.62 (m, 4H), 3.89 (s, 2H), 3.21 (q, J = 7.2 Hz, 55 552.7 2H), 2.48-2.38 (m, \ N7(\©\ _N | (M+H)+ 1H), 2.24-2.12 (m, / o [p 1H), 2.11-1.86 (m, g N ,,3\/ /\ 0 5H), 1.48-1.38 (m, 2H), 1.37-1.24 (m, 2H), 1.29 (d, J 6.8 Hz, 3H), 1.22 (t, J = 7.6 Hz, 3H), 1.06 (d, J = 6.8 Hz, 3H .
(CD30D) 6 8.69 (s, CI 1H), 8.31 (s,1H), 7.89 (d, J = 8.4 Hz, H 2H), 7.64 (d, J : 56 \ NYU 512.8 8.8 Hz, 4H), 7.52 N | , 0 P (M+H)+ (d, J = 8.8 Hz, 2H), g N 55¢ 4.95 (d, J =15.6 /\ 0 Hz, 1H), 4.72 (m, 2H), 4.66 (d, J : 14.4 Hz, 1H ,4.54 (d, J = 12.8 Hz, 1H), 3.90 (s, 2H), 3.21 (q, J = 7.2 Hz, 2H), 2.38-2.23 (m, 1H), 1.22 (t, J = 7.2 Hz, 3H), 1.08 (d, J = 6.0 Hz, 3H), 0.86 d, J = 6.8 Hz, 3H .
(CD30D) 6 8.66 (s, 1H), 8.30 (s, 1H), 7.90-7.82 (m, 6H), 7.65-7.56 (m, 2H), 4.94 (d, J = 15.6 Hz, 1H), 4.71 (m, 57 N 503.7 2H), 4.64 (m, 2H), I (M+H)+ 3.90 (s, 2H), 3.21 N/ O I". (q, J = 7.2 Hz, 2H), 2.39-2.27 (m, 1H), 1.22 (t, J = 7.2 Hz, 3H), 1.04 (d, J = 6.8 Hz, 3H), 0.87 d, J = 6.8 Hz, 3H.
(CD30D) 6 8.67 (s, 1H), 8.31 (s,1H), 7.90-7.81 (m, 6H), 7.67-7.56 (m, 2H), 4.95 (d, J =15.6 Hz, 1H), 4.73 (m, 58 N 546.5 3H), 4.65 (m, 1H), | (M+H)+ 3.90 (s, 2H), 3.21 N/ o (q, J = 7.2 Hz, 2H), 2.38-2.25 (m, 1H), 1.22 (t, J = 7.2 Hz, 3H), 1.04 (d, J = 7.2 Hz, 3H), 0.87 d, J = 6.8 Hz, 3H . ) 6 9.11 (d, J = 1.6 Hz,1H), 9.03 (d, J = 2.0 Hz, 1H), 8.36-8.25 (m, 2H), 7.89 (dd, J = 8.0, 24.0 Hz, 4H), 7.78 (d, J = 8.4 Hz, 1H), 5.38 (q, J = 59 561.1 7.2 Hz, 1H), 5.08- (M+H)+ 5.03 (m, 1H), 4.85- 4.65 (m, 4H), 3.32- 3.29 (m, 2H), 2.40- 2.35(m,1H),1.68 (d, J = 7.2 Hz, 3H), 1.28 (t, J = 7.2 Hz, 3H), 1.16 (d, J = 6.8 Hz, 3H), 0.90 d, J = 6.8 Hz, 3H .
(CD30D) 6 9.12 (d, J = 1.6 Hz,1H), 9.02 (d, J :16 Hz, 1H), .25 (m, 2H), 7.88 (q, J = 8.0 Hz, 4H), 7.75 (d, J = 8.0 Hz,1H), O 5.38 (q, J = 7.2 Hz, 60 561.1 1H), 5.05-4.95 (m, \ N \ N I I H (M+H)+ 1H), 4.85-4.71 (m, , N / N .,Sf\ 4H), 3.31-3.28 (m, /\ o o 2H), 2.45-2.35 (m, 1H), 1.67 (d, J = 6.8 Hz, 3H), 1.28 (t, J = 7.2 Hz, 3H), 1.17 (d, J = 6.8 Hz, 3H), 0.90 (d, J = 6.8 Hz, 3H .
F F 61 510.5 N I (M+H)+ / H/\OS:ON : N b F F p F 8:0 62 524.5 \ N N I (M+H)+ / H 63 539.5 (M+H)+ 64 479.5 (M+H)+ JLJ<o 547.7 N | H (M+H)+ 2015/013699 67 547.5 \ N N I. MO (M+H)+ : N .3.“ /\ 00 68 \ 533.6 l, H/Y‘NO N N (M+H)+ : N \g /\ K 69 495.6 N {Iv-NN I, (M+H)+ : N— F FF 0TI 1» 7o 552.6 \ N N I/ IN Z H (M+H)+ OH OI g N F OH 71 532.6 \ N N I, H (M+H)+ H2N s CI 72 526.6 N I/ HN’XC‘O (M+H)+ 73 458.5 N I, M \,N (M+H)+ - N WO 16904 74 O 547.7 N |\ H NJ< (M+H)+ / o 75 518.6 \ N N I/ l (M+H)+ : ’n’ /\ O 76 458.53 \ N // N I, H/\C—\NN (M+H)+ o (\o 77 477.55 \ NNNQ N I/ H (M+H)+ 78 N 455.41 \ N \ N I, H |/ (M+H)+ £- N O O 489.55 N I/ H \? (M+H)+ _.- N 80 528.56 \ N N I/ H (M+H)+ j N 0 Nd 81 566.46 \ N N I/ H (M+H)+ WO 16904 82 458.5 \ N (M+H)+ é N ‘N 83 456.49 \ N \ N N I/ H | . (M+H)+ s: N 84 456.49 \ N N I/ H (M+H)+ 85 464.45 \ N N l/ H (M+H)+ : N 86 484.5 \ N/ng/ N | (M+H)+ 87 510.5 N | HE N (M+H)+ 88 554.7 \ N N l/ H (M+H)+ s‘ N 89 524.6 \ N N I, (M+H)+ s‘ N WO 16904 90 476.6 Nl/ (M+H)+ s.-N 91 546.6 Nl/ (M+H)+ 92 512.6 NI/ (M+H)+ s.-N 93 621.6 NI/ (M+H)+ 94 485.5 Nl/ (M+H)+ 95 561.6 NI, (M+H)+ 96 472.51 \ N/\/\N \ NI, H SN (M+H)+ s.-N 0 / 97 469.55 \ N \N NI/ H (M+H)+ 2015/013699 98 N03 541.66 (M+H)+ g- N O /\ R o (\N’ 99 490.59 N I/ (M+H)+ S= N 100 475.57 \ N (M+H)+ N ‘ O /N 101 485.45 \ N OH N l/ H (M+H)+ 102 o/\//:/NS\/ 539.5 \ N N I/ H (M+H)+ Fh o 0 NJLO/ 103 Na/L 539.5 N l/ H (M+H)+ s N 104 525.5 \ N N I, H/TN./\ (M+H)+ : 0"S‘b WO 16904 105 491.5 \ N N I, H/\C\N (M+H)+ : If)" 106 /" 539.5 \ N N l/ , H N,,S:O (M+H)+ N O 107 ’ 505.6 (M+H)+ 108 517.6 /III \ N N l/ H H (M+H)+ N ‘ /\ O 109 553.5 \ N N l/ H (M+H)+ i N 3. 110 519.6 N:(YLNl/ H/U (M+H)+ N o\ g ‘n’ /\ O 111 554.5 \ N N. IN, H/U.S.N\H (M+H)+ 112 533.6 \ N OH N l, H (M+H)+ s 1% /\ O WO 16904 113 519.6 \N OH Nl/H (M+H)+ /\ O 114 503.6 N I, H/U (M+H)+ sN ‘n/ /\ O 115 455.5 \N \ N I, H IN (M+H)+ 116 030 532.5 Nam;\N \ (M+H)+ 117 F 446.4 N| (M+H)+ , ”NF 118 534.5 N l/ Hno (M+H)+ S‘N OJ 119 546.5 waffll\N (M+H)+ s=N Br 120 N 482.5 N HIIJ\ I, (M+H)+ é-N N WO 16904 F F O NH O 121 557.5 HN2 G N I: H (M+H)+ = N CI Cl F F 122 406.4 \ J\ J\ G N l (M+H)+ , N H2N F F 123 562.4 N I (M+H)+ , HLQ N Br F F 124 F 428.4 N | N/Y H (M+H)+ / F 125 496.5 \ N' N I H (M+H)+ l (SH F F 125 544.6 \ 0\ N N | (M+H)+ / H s: N F F 127 532.6 N |\ H O (M+H)+ / "Ir/A / i— O WO 16904 128 604.5 (M+H)+ 129 485.6 \ N N l/ (M+H)+ : N I/\§N 130 496.5 \ N NI H (M+H)+ / OH 131 496.5 \ N NI I (M+H)+ / H OH 132 496.5 NI (M+H)+ (CD30D) 6 9.02 (s, 1H), 8.24 (s, 1H), 8.18 (s, 1H), 7.90 FF (s, 1H), 7.82 (m, 4H),4.96 (d, J = 16.0 Hz, 1H), 4.91 133 536.46 (m, 2H), 4.78 (s, N l/ (M+H)+ 2H), 4.52 (s, 2H), 3.58 (q, J = 7.6 Hz, : N ’\ 2H), 2.38 (m, 1H), 1.21 (t, J = 7.6 Hz, 3H), 1.17 (d, J = 8.0 Hz, 3H), 0.89 d, J = 6.8 Hz, 3H. 134 520.56 NI (M+H)+ WO 16904 I O O O 135 593.61 NH | G (M+H)+ H2N 136 579.47 135 H (M+H)+ 137 532.62 HgN \ (M+H)+ o 138 530.5 (M+H)+ 139 518.5 (M+H)+ 140 590.5 (M+H)+ WO 16904 (CD30D) 5 9.12 (s, 1H), 8.90 (s, 1H), 8.32 (s, 1H), 8.17 (d, J: 8.4 Hz, 1H), 7.90 (d, J : 8.0 Hz, 2H), 7.84 (d, J: 8.0 Hz, 2H), 7.65 (d, J: 8.4 Hz, 1H), 5.05 (d, J: 15.6 Hz, 1H), 4.77 (dd, J: 141 NflN%H Eff): 13.2,30.0Hz,2H), 3.34(s,2H),3.26 , N/ N/,,S_.’\ (q, J: 7.6 Hz, 2H), /\ 00 2.37 (broad s, 1H), 1.84 (dd, J= 4.4, 7.6 Hz, 2H), 1.52 (dd, J= 4.4, 7.6 Hz, 2H), 1.23 (t, J= 7.6 Hz, 3H), 1.16 (d, J = 6.0, 3H), 0.89 (d, J= 6.8 Hz, 3H .
(CD30D) 5 9.11 (s, 1H), 8.87 (s, 1H), 8.33 (d, J= 8.0 Hz, 2H), 7.90 (d, J= 8.0 Hz, 2H), 7.84 (d, J= 8.0 Hz, 2H), 5.04 (d, J= 15.6 Hz, 1H), 4.89-4.82 (m, 142 Nflfifi0 561.55 2H), 4.87 (s, 2H), (M+H)+ 4.76 (dd, J: 13.2, N’ N / ,sf\ 28.8 Hz, 2H),3.33 /\ o o (q, J: 7.6 Hz, 2H), 2.58 (s, 3H), 2.36 (bs, 1H), 1.26 (t, J = 7.6 Hz, 3H), 1.14 (d, J= 6.0 Hz, 3H), 0.88 (d, J: 6.8 Hz, (CD30D) 5 9.12 (5, 1H), 8.72 (d,J 7.6 Hz, 1H), 8.35 (s, 1H), 7.92 (d, J: 7.6 Hz, 1H), 7.92 (d, J: 8.0 Hz, 2H), 7.83 (d, J: 8.0 Hz, 2H), 5.07 (d, J: .2 Hz, 1H),4.91- 143 561 52 \ N \ (M+H)+ 4.71 (m, 4H), 4.77 N I I / H N / (dd, J: 12.4, 35.4 N /\ :~' ISeO Hz, 2H), 3.41 (q, J /\ O : 7.6 Hz, 2H), 3.06 (s, 3H), 2.36 (br,J=d0a 3, 1H), 1.28 (t 7.6 Hz, 3H), 1.14 (d, J: 5.2 Hz, 3H), 0.88 (d J: 6.8 Hz, (CD30D) 5 9.08 (s, 1H), 8.81 (s, 1H), 8.28 (s, 1H), 8.12 (d, J: O F 8.8 Hz, 1H), 7.91 \ N \ (Sn/ISEHE): (d, J : 8.0 Hz, 2H), N I I / N / 7.83 (d, J: 8.0 Hz, 3 N 01.3.1 2H), 5.04 (d, J: /\ O 14.4 Hz, 1H), 4.91- (s, 2H), 4.76 (dd, J = 12.4, 33.4 Hz, 2H), 3.33 (q, J: 7.6 Hz, 2H), 2.35 (broad s, 1H), 1.25 (t, J: 7.6 Hz, 3H), 1.13 (d, J: 5.2 Hz, 3H), 0.87 (d, J: 6.8 Hz, 3H .
HO O 145 518.55 \ N 137 N I H - : N F F O OH 146 506.58 \ N O 134 (M+H)+- = N s O F F 148 504.6 \ N 127 N l, (M+H)+- N Hm. g "”n’ /\ o F F 149 O 509.5 I: OH \ N 10/ (M+H)+ N I / H s: N 150 513.6 \ N \ N I I H (M+H)+ N / O\ g N’ /\ O F F 151 499.6 \ N \ 150 N l I (M+H)+- , H N / OH s: N /\ O WO 16904 F F FAQ O O“s"o N. \ 152 /\/C/ 525-6 N | j ” (M+H)+ ; N F F 153 5115- \ N N I <M+H>+ . HMS/“C g N d, ¢O 154 o 0 525.5 \ 1", -n(0X11 155 480.5 / / 2 12 z_ \ (M+H)+ Z\ // )Ill z F F 156 577.5 \ n CII, N I, (M+H)+ 157 m, 561.6 N |\ (M+H)+ , H N 0 ”11/ /\ A0 0 F F CN 158 0 509.5 (M+H)+ \ N N I / H OH OH : H2N WO 16904 (CD30D) 6 9.13 (d, J: 2.0 Hz, 1H), 8.32 (d, J: 3.2 Hz, 1H), 8.29 (s, 1H), F F 8.07 (d, J: 8.8 Hz, F 1H), 7.92-7.85 (m, 0 4H), .04 (m, 159 N, 548.1 3H), 4.89-4.85 (m, N H | (M+H)+ 1H), 4.80-4.76 (m, 3H), 3.60 (q, J: /‘\ 0"S\/ 7.2 Hz, 2H), 2.42- 2.38 (m, 1H),1.33 (t, J: 7.2 Hz, 3H), 1.17 (d, J: 6.8 Hz, 3H), 0.91 (d, J: 7.2 Hz, 3H .
BIOLOGICAL ASSAYS Radio-Ligand RORy Binding Assay (Assay 1) ] Compounds of the present invention were tested for ability to bind to RORY in a cell— free competition assay with commercially available radio—ligand (RL), 25—hydroxy [26,27—3H]— cholesterol (PerkinElmer, Cat. # NET674250UC), for a ligand binding site on a recombinant RORY Ligand Binding Domain (LBD) protein expressed as a Glutathione—S—Transferase (GST) fusion. The assay was performed in 96—well SPA plates (PerkinElmer, Cat. # 1450—401) in 50 mM HEPES buffer, pH 7.4, ning 150 mM NaCl, 5 mM MgClg, 10% (v/v) glycerol, 2 mM CHAPS, 0.5 mM B—octylglucopyranoside and 5 mM DTT. Tested compounds were dissolved in DMSO, and semi—log (3.162X) serial dilutions of the compounds were prepared in the same solvent. Two uL of the DMSO solutions were mixed with 28 uL of 8.6 nM 25— hydroxy [26,27—3H]— cholesterol and 50 uL of 24 nM RORY LED. The plate was shaken at 700 rpm for 20 min and incubated for 10 min at rt, after which 40 uL of poly—Lys YSi SPA beads (PerkinElmer, Cat. # RPNQ0010) were added to e 50 ug of the beads per well. The plate was incubated on an orbital shaker for 20 min and then for 10 min without agitation at rt. SPA signal for tritium beta radiation was registered on PerkinElmer Microbeta plate reader. Percent inhibition values were calculated based on the high signal obtained with DMSO control and the low signal observed with 10 uM rd RORY inverse agonist T09013l7 (SigmaAldrich, Cat.
# . The percent inhibition vs. concentration data were fit into a four—parameter model, and IC50 values were calculated from the fit as the concentrations corresponding to the inflection points on the dose—response curves. Inhibitory constants (Ki) were calculated using the following equation, where [RL] is the concentration in the assay and KD is a dissociation nt of roxy [26,27—3H]— cholesterol: RORyt SXRORE Assay in Jurkat Cells (Assay 2) ] Compounds of the t invention were tested for RORY inverse t activity in a cell—based, transcriptional activity assay. Secreted c® luciferase was used as a reporter for transcriptional activity of the full—length RORyt in Jurkat cells (ATCC, Cat. # TIE—152). A reporter plasmid was constructed by inserting 5 repeats of the ROR Response Element (RORE) AAAGTAGGTCA (SEQ ID NO: 1) into a commercially available promoterless plasmid pNL1.3[sechuc] (Promega, Cat. # N1021) using KpnI and I restriction sites. The expression plasmid for RORyt was purchased (Geneocopoeia, Cat. # EX—T6988—M02). Jurkat cells (30 million cells) were transfected with 11 u g of EX—T6988—M02 and 26 ug of the reporter plasmid in OptiMEM® media using Lipofectamine® LTX and PlusTM reagents (Life Technologies, Cat. # 15338—100). After 5—6 hrs of incubation at 37 °C/5% C02, the cells were collected, resuspended in phenol—red free RPMI media containing 10% (v/v) delipidated FBS (Hyclone, Cat. # SH30855.03) and dispensed into l clear bottom tissue culture plates (CoStar, Cat. # 3603), at 80,000 cells per well. Tested compounds were added to the cells in the same media (final concentration of DMSO was 0.1% , and the plates were incubated at 37 °C/5% C02 for 16—18 hrs. Luciferase ty in the conditioned supernatants was determined with NanoGlo® assay reagents (Promega, Cat.# N1130). Percent inhibition values were calculated based on the fully inhibited and non—inhibited (DMSO) controls, and the values were regressed against concentrations of the tested compounds to derive IC50 values using a four— parameter near fitting model.
The results of assays 1 and 2 are shown in Table 2.
Table 2: Compound RORy RORytSX Compound RORy RORytSX # Binding IC50 # Binding IC50 Ki Range* Ki Range* Range* (nM) (nM) (nM) 1 +++ +++ 2 +++ +++ 3 +++ +++ 4 +++ +++ +++ +++ 6 +++ + 7 +++ ++ 8 +++ + WO 16904 9 +++ ++ +++ 11 ++ \D ++++++ 12 ++ +++ \D 13 \D 14 ++ +++ \DUI ++ 16 ++ 17 +++ +++ \D\l +++ +++ 100 21 ++ ++ 101 22 +++ +++ 102 ++ 23 ++ 103 24 +++ 104 ++ +++ ++ 105 + 26 ++ 106 ++ 27 +++ ++ 107 ++ 28 +++ +++ 108 29 +++ +++ 109 +++ + +++ +++ 110 + 31 +++ +++ 111 ++ 32 +++ ++ 112 + 33 +++ +++ 113 ++ 34 +++ +++ 114 + +++ ++ 115 ++ 36 ++ 116 ++ 37 117 38 +++ ++ 118 39 +++ +++ 119 40 ++ 120 41 +++ 121 42 +++ 122 43 +++ 123 ++ 44 +++ 124 45 +++ 125 46 +++ 126 47 +++ 127 ++ 48 128 ++ 49 +++ 129 ++ 50 130 51 ++ 131 ++ 52 ++ 132 ++ 53 ++ 133 + + ++ 54 +++ +++ 134 55 +++ +++ 135 56 +++ ++ 136 57 +++ +++ 137 58 +++ +++ 59 +++ +++ 60 ++ 61 + 62 + 63 +++ ++ 64 +++ ++ 65 ++ 66 + 67 +++ +++ 68 + 69 + 70 + 71 ++ 72 ++ 73 + 74 + 75 + 76 ++ 77 + 78 ++ 79 + 80 + >l<+ means > 1000 nM; ++ means 100 nM — 1000 nM; +++ means < 100 nM.
] While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds and s of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.
The contents of all references (including literature references, issued patents, published patent applications, and co—pending patent applications) cited hout this application are hereby expressly incorporated herein in their entireties by reference. Unless ise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art.
Listing of

Claims (25)

Claims:
1. A compound of Formula (I): (I); or a pharmaceutically acceptable salt thereof, wherein R2 is (C1-C3)alkyl; R3 is hydrogen, hydroxy, monocyclic cycloalkyl, monocyclic heterocyclyl, or (C1- C6)alkyl, wherein the (C1-C6)alkyl is optionally tuted with 1 to 2 groups independently selected from hydroxy, halo, and cyano; R4 is hydrogen, (C1-C3)alkyl, or =O; X is –C(O)NH- or –NHC(O)-; m is 0, 1, or 2; n is 0, 1, 2, or 3; L1 is absent or is SO2 or CR7R8; Cy1 is phenyl, dinyl, tetrahydro-2H-thiopyranyl 1,1-dioxide, pyridinyl, zinyl, azetidinyl, imidazolyl, tetrahydropyranyl, 1,4-dioxanyl, pyridazinyl, pyrazolyl, pyrrolidinyl, cyclohexyl, morpholinyl, 6,7-dihydro-5H-pyrrolo[2,1-c][1,2,4]triazolyl, 1,2,3,4-tetrahydro-1,8- naphthyridinyl, 2,3-dihydro-1H-indenyl, or imidazo[1,2-a]pyrimidinyl, each of which is ally substituted with 1 to 2 groups independently selected from R5, wherein at least one R5 is (C1-C3)alkylsulfonyl or (C1-C3)alkylaminosulfonyl when Cy1 is phenyl or pyridiyl; Cy2 is selected from (C1-C6)alkoxycarbonyl, phenyl(C1-C3)alkoxycarbonyl, halophenyl(C1-C3)alkoxycarbonyl, aryl, heteroaryl, monocyclic cycloalkyl, and monocyclic heterocyclyl, n the aryl, heteroaryl, monocyclic lkyl, and moncyclic heterocyclyl are each ally substituted with 1 to 3 groups independently selected from R6; R5 and R6 are each independently selected from halo, cyano, nitro, amino, hydroxy, carboxy, )alkyl, heterocyclyl, hydroxy(C1-C6)alkyl, CO2H, (CH2)1-3COOH, (C1- C3)alkylcarbonyloxy, (C3-C6)cycloalkyl, hydroxy(C3-C6)cycloalkyl, (C4-C7)cycloalkylalkyl, (C2- C6)alkenyl, halo(C2-C6)alkenyl, hydroxy(C2-C6)alkenyl, (C2-C6)alkynyl, )cycloalkyl(C2- C4)alkynyl, halo(C1-C6)alkyl, halo(C3-C6)cycloalkyl, halo(C4-C7)cycloalkylalkyl, (C1-C6)alkoxy, (C3-C6)cycloalkoxy, (C4-C7)cycloalkylalkoxy, halo(C1-C6)alkoxy, 3-C6)cycloalkoxy, halo(C4-C7)cycloalkylalkoxy, )alkylthio, (C3-C6)cycloalkythio, (C4-C7)cycloalkylalkylthio, halo(C1-C6)alkylthio, halo(C3-C6)cycloalkythio, 4-C7)cycloalkylalkylthio, (C1- C6)alkylsulfinyl, (C3-C6)cycloalkylsulfinyl, (C4-C7)cycloalkylalkylsulfinyl, halo(C1- ylsulfonyl, halo(C3-C6)cycloalkylsulfinyl, halo(C4-C7)cycloalkylalkylsulfinyl, (C1- C6)alkylsulfonyl, (C3-C6)cycloalkylsulfonyl, (C4-C7)cycloalkylalkylsulfonyl, halo(C1- C6)alkylsulfonyl, halo(C3-C6)cycloalkylsulfonyl, halo(C4-C7)cycloalkylalkylsulfonyl, (C1- C6)alkylamino, di(C1-C6)alkylamino, (C1-C6)alkoxy(C1-C6)alkoxy, halo(C1-C6)alkoxy(C1- C6)alkoxy, (C1-C6)alkoxycarbonyl, H2NCO, H2NSO2, (C1-C6)alkylaminocarbonyl, di(C1- C6)alkylaminocarbonyl, (C1-C3)alkoxy(C1-C3)alkylaminocarbonyl, heterocyclylcarbonyl, (C1- ylaminosulfonyl, di(C1-C6)alkylaminosulfonyl, heterocyclylsulfonyl, (C1- C6)alkylcarbonylamino, (C1-C6)alkylcarbonylamino(C1-C6)alkyl, (C1-C6)alkylsulfonylamino, (C1-C6)alkylsulfonylamino(C1-C6)alkyl, (C1-C6)alkoxycarbonyl(C1-C6)alkoxy, (C1- C6)alkoxy(C1-C6)alkyl, halo(C1-C6)alkoxy(C1-C6)alkyl, hydroxy(C1-C6)alkoxy, aryl, heteroaryl, oxo, C1-C6)alkyl, (C1-C6)alkylamino(C1-C6)alkyl, di(C1-C6)alkylamino(C1-C6)alkyl amino(C2-C6)alkoxy, (C1-C6)alkylamino(C2-C6)alkoxy, di(C1-C6)alkylamino(C2-C6)alkoxy, (C1- C6)alkylcarbonyl, hydroxy(C1-C6)alkylcarbonyl, (C1-C6)alkylhydroxycarbonyl, (C1- C6)alkylhydroxy(C1-C6)alkyl, (C3-C6)cycloalkylcarbonyl, (C3-C6)cycloalkylaminocarbonyl, {(C3-C6)cycloalkyl}{(C1-C6)alkyl}aminocarbonyl, di(C3-C6)cycloalkylaminocarbonyl, (C3- loalkylaminosulfonyl, {(C3-C6)cycloalkyl}{(C1-C6)alkyl}aminosulfonyl, di(C3- C6)cycloalkylaminosulfonyl, cyano(C1-C6)alkyl, aminocarbonyl(C1-C6)alkyl, (C1- C6)alkylaminocarbonyl(C1-C6)alkyl, di(C1-C6)alkylaminocarbonyl(C1-C6)alkyl, (C3- C6)cycloalkylaminocarbonyl(C1-C6)alkyl, {(C3-C6)cycloalkyl}{(C1-C6)alkyl}aminocarbonyl(C1- C6)alkyl, [(C1-C6)alkyl(C4-C6)heterocyclyl](C1-C6)alkyl, and di(C3- C6)cycloalkylaminocarbonyl(C1-C6)alkyl; and R7 and R8 are each independently hydrogen, hydroxy, )alkyl, hydroxy(C1-C3)alkyl, mono(C1-C3)alkylamino, C3)alkylamino, CO2H, (CH2)1-3COOH, moncyclic heterocyclyl, (C1-C3)alkoxycarbonyl, (C1-C3)alkyl(C1-C3)alkoxycarbonyl, halophenyl, halophenyl(C1- C3)alkyl, or quinolin-2(1H)one-4yl-methyl; or R7 and R8, together with the carbon atom to which they are attached, form a 3- to 6- membered cycloalkyl or heterocyclyl.
2. The compound of Claim 1, wherein the compound is of Formula (II): (II); or a pharmaceutically acceptable salt thereof.
3. The compound of Claim 1 or 2, wherein the compound is of a (III): (III); or a pharmaceutically able salt thereof.
4. The compound of any one of Claims 1 to 3, wherein the compound is of Formula (IV): (IV); or a pharmaceutically able salt thereof.
5. The compound of any one of Claims 1 to 4, wherein the compound is of Formula (V) or (VI): (V); or (VI); or a pharmaceutically acceptable salt thereof.
6. The compound of any one of Claims 1 to 5, wherein the compound is of Formula (VII): (VII); or a pharmaceutically acceptable salt thereof.
7. The compound of any one of Claims 1 to 6, n R3 is hydrogen, hydroxy, or (C1- C3)alkyl.
8. The compound of any one of Claims 1 to 7, wherein Cy2 is selected from aryl, heteroaryl, monocyclic cycloalkyl, and monocyclic heterocyclyl, each of which is optionally tuted with 1 to 2 groups independently selected from R6.
9. The compound of any one of Claims 1 to 8, wherein Cy2 is phenyl, pyrimidinyl, cyclohexyl, or pyridinyl, each of which are optionally substituted with 1 to 2 groups independently selected from R6.
10. The compound of any one of Claims 1 to 9, wherein n is 1 or 2; and Cy1 is phenyl, pyridinyl, or piperidinyl, each of which is optionally substituted with 1 to 2 groups ndently selected from R5, wherein at least one R5 is (C1-C3)alkylsulfonyl or (C1- C3)alkylaminosulfonyl.
11. The compound of any one of Claims 1 to 10, wherein Cy2 is cyclohexyl optionally substituted with 1 to 2 groups independently selected from R6.
12. The compound of any one of Claims 1 to 11, wherein R5 is selected from halo, (C1-C3)alkyl, halo(C1-C3)alkyl, cyano, hydroxy(C1-C3)alkyl, )alkoxycarbonyl, (C1-C3)alkylsulfonyl, (C1-C3)alkoxy, halo(C1-C3)alkoxy, oxo, hydroxy, (C1-C3)alkylcarbonyl, hydroxy(C1-C3)alkylcarbonyl, )alkylhydroxycarbonyl, (C1- C3)alkylaminosulfonyl, (C1-C3)alkylaminocarbonyl, di(C1-C3)alkylamino(C2-C6)alkoxy, (C1- C6)alkoxycarbonyl, [(C1-C3)alkyl(C4-C6)heterocyclyl](C1-C3)alkyl, and (C1-C3)alkylhydroxy(C1- C3)alkyl; and R6 is selected from halo, (C1-C3)alkyl, halo(C1-C3)alkyl, cyano, hydroxy(C1-C3)alkyl, )alkoxycarbonyl, (C1-C3)alkylsulfonyl, (C1-C3)alkoxy, 1-C3)alkoxy, oxo, hydroxy, aryl, (C1-C3)alkylhydroxy(C1-C3)alkyl, heteroaryl, and (C1-C3)alkoxycarbonyl.
13. The compound of any one of Claims 1 to 12, wherein R5 is selected from halo, (C1-C3)alkoxy, hydroxy, (C1-C3)alkyl, hydroxy(C1-C3)alkyl, halo(C1-C3)alkyl, (C1-C6)alkoxycarbonyl, di(C1-C3)alkylamino(C2-C6)alkoxy, [(C1-C3)alkyl(C4- C6)heterocyclyl](C1-C3)alkyl, oxo, (C1-C3)alkylcarbonyl, )alkylaminosulfonyl, (C1- ylsulfonyl, and cyano; and R6 is selected from halo(C1-C3)alkyl, (C1-C3)alkoxy, halo, cyano, (C1-C3)alkoxycarbonyl, (C1-C3)alkylhydroxy(C1-C3)alkyl, hydroxy(C1-C3)alkyl, and halo(C1-C3)alkoxy.
14. The compound of any one of Claims 1 to 13, wherein R5 is selected from halo, cyano, (C1-C3)alkyl, (C1-C3)alkylaminosulfonyl, and (C1-C3)alkylsulfonyl; and R6 is selected from halo(C1-C3)alkyl, (C1-C3)alkoxy, halo, cyano, (C1-C3)alkoxycarbonyl, and 1-C3)alkoxy.
15. The compound of any one of Claims 1 to 14, wherein Cy1 is ; R10 is (C1-C3)alkyl or (C1-C3)alkylamino; and Z is CH or N.
16. The compound of any one of Claims 1 to 15, wherein Cy2 is ; ; ; ; or ; R12 is (C1-C3)alkoxycarbonyl, halo, dihalo, )alkoxy, or halo(C1-C3)alkyl; R13 is halo or halo(C1-C3)alkyl; and R14 is halo, cyano, halo(C1-C3)alkyl, or halo(C1-C3)alkoxy.
17. The nd of Claim 16, wherein R12 to R14 are each CF3.
18. The compound of any one of Claims 1 to 17, wherein R2 is isopropyl.
19. The compound of Claim 1, wherein the compound is of the Formula: or a pharmaceutically acceptable salt thereof.
20. The compound of Claim 1, wherein the compound is selected from any one of the following formulae or a pharmaceutically acceptable salt thereof Structure N H O N S S N H O O N SO2Et N H O N S N H N S N H N S O N N H O O N S N H N O N S N H N S N H N S N H N S O O ’3 \ N \ /\ OQ 100100 O O N S O O N S N H N N S N H S O N O NI/ HM) é~N SI\ /\ 00 NI/ O §N Y : N/ N—\ N l/ N MN N '/ N HEN/{0% 103103 N H N N (11,, 2 IZ \ OH II“, II”, 2 IZ 105105 O N N H \ N N l/ HKCNA N d’S‘b N l/ HKCNON N ‘n/\ \ N N| HrN’§s 00 s N/ NI HEN/NO s N/ \ ///,,, N| H H é N/ /\ O \ N N I/ H N :,sf\ /\ Oo 107107 N H N \ N I;\ N l/ H N N O 0NH \ N N l/ H = N 5 CI \ N"= N l/ H EH l/ H/UN O S: N ”(QAO/ \ Nas S N (”S t N 109109 Nl/HOH Nl/HEH Nl/HOH 110110 N O N H N OH 112112 N H N S
21. A nd selected from any one of the following formulae or a pharmaceutically acceptable salt thereof / OH sN \\ = N F FF NI/ H F OH NI/ H gN CI \ N o/ NI/ H \N \ N l/ H l/ 114114 N|\N 0 Nd \ N N l/ H s‘ N 0 00 N I/ H S: N O /N| \ N N l/ H 5‘ N H [01 115115 'l'l 'l'l “H “H TI TI 'l'l “H “H 116116 117117 N/ O \ N N I/ HO O O \ N OH N l/ H i N \ N/x/ N I/ H 5: N \ N \ NI ' N/ N/ O\ /\ O \ N \ N I/ I N/ OH 5 N /\ O 118118 N H N
22. A pharmaceutical composition comprising a compound of any one of Claims 1 to 21, or a ceutically acceptable salt thereof, and a ceutically acceptable carrier.
23. The use of a therapeutically effective amount of a compound according to any one of Claims 1 to 21, or a ceutically able salt thereof in the manufacture of a medicament for the treatment of one or more diseases or disorders that are responsive to the inhibition of ROR gamma in a subject,
24. The use of Claim 23, wherein the disease or disorder is selected from asthma, chronic obstructive pulmonary disease (COPD), bronchitis, allergic rhinitis, atopic dermatitis, contact dermatitis, acne, cystic fibrosis, allograft rejection, multiple sclerosis, scleroderma, arthritis, rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, sing spondylitis, systemic lupus erythematosus (SLE), psoriasis, Hashimoto's disease, pancreatitis, autoimmune diabetes, type I es, autoimmune ocular disease, ulcerative colitis, Crohn's disease, regional tis, inflammatory bowel disease (IBD), inflammatory bowel syndrome (IBS), Sjögren's me, optic neuritis, obesity, hepatosteatosis, adipose tissue-associated inflammation, insulin ance, type II diabetes, neuromyelitis optica, myasthenia gravis, age related macular degeneration, dry eye, uveitis, Guillain-Barré syndrome, sis, psoriatic arthritis (PsA), steroid resistant asthma, Graves' disease, scleritis, major depression, seasonal affective disorder, PTSD, bipolar disorder, autism, epilepsy, Alzheimer’s, CNS disorders associated with altered sleep and/or circadian s, endometriosis, obstructive sleep apnea syndrome , Behçet's disease, dermatomyositis, polymyocitis, graft versus host disease, primary biliary cirrhosis, liver fibrosis, non-alcoholic fatty liver disease (NAFLD), sarcoidosis, primary sclerosing cholangitis, autoimmune thyroid disease, autoimmune polyendocrine syndrome type I, mune polyendocrine syndrome type II, celiac disease, neuromyelitis, juvenile idiopathic arthritis, systemic sclerosis, myocardial tion, pulmonary hypertension, osteoarthritis, cutaneous leishmaniasis, sal polyposis, and cancer.
25. The use of Claim 24, wherein the disease or er is selected from , atopic dermatitis, acne, s disease, regional tis, ulcerative colitis, Sjögren's syndrome, uveitis, Behçet's disease, dermatomyositis, multiple sclerosis, ankylosing spondylitis, systemic lupus erythematosus (SLE), scleroderma, psoriasis, psoriatic arthritis (PsA), steroid resistant asthma, and rheumatoid arthritis.
NZ722413A 2014-02-03 2015-01-30 Dihydropyrrolopyridine inhibitors of ror-gamma NZ722413B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201461935162P 2014-02-03 2014-02-03
US61/935,162 2014-02-03
US201461970637P 2014-03-26 2014-03-26
US61/970,637 2014-03-26
PCT/US2015/013699 WO2015116904A1 (en) 2014-02-03 2015-01-30 Dihydropyrrolopyridine inhibitors of ror-gamma

Publications (2)

Publication Number Publication Date
NZ722413A NZ722413A (en) 2021-08-27
NZ722413B2 true NZ722413B2 (en) 2021-11-30

Family

ID=

Similar Documents

Publication Publication Date Title
US11535614B2 (en) Dihydropyrrolopyridine inhibitors of ROR-gamma
US9868748B2 (en) Thiazolopyrrolidine inhibitors of ROR- γ
US10829448B2 (en) Substituted benzoimidazoles as modulators of ROR-γ
US11001583B2 (en) Dihydropyrrolopyridine inhibitors of ROR-gamma
US9845308B2 (en) Isoindoline inhibitors of ROR-gamma
CA2963140A1 (en) Dihydropyrrolopyridine inhibitors of ror-gamma
CA3124130A1 (en) Substituted 3-((3-aminophenyl)amino)piperidine-2,6-dione compounds, compositions thereof, and methods of treatment therewith
CA3196061A1 (en) Cftr modulator compounds, compositions, and uses thereof
NZ722413B2 (en) Dihydropyrrolopyridine inhibitors of ror-gamma
JP2023520005A (en) N-heteroarylalkyl-2-(heterocyclyl and heterocyclylmethyl)acetamide derivatives as SSTR4 agonists