OA17657A - Heteroaromatic compounds and their use as Dopamine DI ligands - Google Patents

Heteroaromatic compounds and their use as Dopamine DI ligands Download PDF

Info

Publication number
OA17657A
OA17657A OA1201500511 OA17657A OA 17657 A OA17657 A OA 17657A OA 1201500511 OA1201500511 OA 1201500511 OA 17657 A OA17657 A OA 17657A
Authority
OA
OAPI
Prior art keywords
alkyl
group
compound
cycloalkyl
independently selected
Prior art date
Application number
OA1201500511
Inventor
Michael Aaron Brodney
Jennifer Elizabeth Davoren
Amy Beth Dounay
Ivan Viktorovich Efremov
David Lawrence Firman Gray
Michael Eric GREEN
Jaclyn Louise HENDERSON
Chewah LEE
Scot Richard MENTE
Steven Victor O'neil
Bruce Nelsen Rogers
Lei Zhang
Original Assignee
Pfizer Inc.
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 Pfizer Inc. filed Critical Pfizer Inc.
Publication of OA17657A publication Critical patent/OA17657A/en

Links

Abstract

The present invention provides, in part, compounds of formula l : and pharmaceutically acceptable salts thereof; processes for the preparation of; intermediates used in the preparation of; and compositions containing such compounds or salts, and their uses for treating D1-mediated (or D1-associated) disorders including, e.g., schizophrenia (e.g., its cognitive and negative symptoms), cognitive impairment (e.g., cognitive impairment associated with schizophrenia, AD, PD, or pharmacotherapy therapy), age-related cognitive decline, dementia, and Parkinson's disease.

Description

HETEROAROMATIC COMPOUNDS AND THEIR USE AS DOPAMINE D1 LIGANDS
FIELD OF THE INVENTION
The présent invention generally relates to heteroaromatic compounds, which are dopamine D1 ligands, for example dopamine D1 agonists or partial agonists.
BACKGROUND OF THE INVENTION
Dopamine acts upon neurons through two families of dopamine receptors, D1 -like receptors (D1Rs) and D2-like receptors (D2Rs). The D1 -like receptor family consists of D1 and D5 receptors which are expressed in many régions of the brain. D1 mRNA has been found, for example, in the striatum and nucléus accumbens. See e.g., Missale C, Nash SR, Robinson
SW, Jaber M, Caron MG “Dopamine receptors: from structure to function”, Physiological Reviews 78:189-225 (1998). Pharmacological studies hâve reported that D1 and D5 receptors (D1/D5), namely D1 -like receptors, are linked to stimulation of adenylyl cyclase, whereas D2, D3, and D4 receptors, namely D2-like receptors, are linked to inhibition of cAMP production.
Dopamine D1 receptors are implicated in numerous neuropharmacological and neurobiological functions. For example, D1 receptors are involved in different types of memory function and synaptic plasticity. See e.g., Goldman-Rakic PS et al., “Targeting the dopamine D1 receptor in schizophrenia: insights for cognitive dysfunction”, Psychopharmacology 174(1):316 (2004). Moreover, D1 receptors hâve been implicated in a variety of psychiatrie, neurological, neurodevelopmental, neurodegenerative, mood, motivational, metabolic, cardiovascular, rénal, ophthalmic, endocrine, and/or other disorders described herein including schizophrenia (e.g., cognitive and négative symptoms in schizophrenia), cognitive impairment associated with D2 antagonist therapy, ADHD, impulsivity, autism spectrum disorder, mild cognitive impairment (MCI), age-related cognitive décliné, Alzheimer’s dementia, Parkinson’s disease (PD), Huntington’s chorea, dépréssion, anxiety, treatment-resistant dépréssion (TRD), bipolar disorder, chronic apathy, anhedonia, chronic fatigue, post-traumatic stress disorder, seasonal affective disorder, social anxiety disorder, post-partum dépréssion, serotonin syndrome, substance abuse and drug dependence, Tourette’s syndrome, tardive dyskinesia, drowsiness, sexual dysfunction, migraine, systemic lupus erythematosus (SLE), hyperglycemia, dislipidemia, obesity, diabètes, sepsis, post-ischemic tubular necrosis, rénal failure, résistant edema, narcolepsy, hypertension, congestive heart failure, postoperative ocular hypotonia, sleep disorders, pain, and other disorders in a mammal. See e.g., Goulet M, Madras BK “D(1) dopamine receptor agonists are more effective in alleviating advanced than mild parkinsonism in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated monkeys”, Journal of Pharmacology and Experimental Therapy 292(2):714-24 (2000); Surmeier DJ et al., “The rôle of dopamine in modulating the structure and function of striatal circuits”, Prog. Brain Res. 183:149-67 (2010).
New or improved agents that modulate (such as agonize or partially agonize) D1 are needed for developing new and more effective pharmaceuticals to treat diseases or conditions associated with dysregulated activation of D1, such as those described herein.
SUMMARY OF THE INVENTION
The présent invention provides, in part, a compound of Formula I:
or a pharmaceutically acceptable sait thereof, wherein:
each of T1, T2, T3, andT4 is independently selected from the group consisting of H, halogen, -CN, -SF5, -OH, -N(Ra)(Rb), -C(=O)-N(Ra)(Rb), -C(=O)-ORC, -C(=O)-Rd, Ον6 alkyl, Ον6 haloalkyl, C2.6 alkenyl, C2.6 alkynyl, alkoxy, haloalkoxy, -S-fC^ alkyl), C3.7 cycloalkyl, 4to 7-membered heterocycloalkyl, C3.7 cycloalkoxy, 5- or 6- membered heteroaryl, cyclopropylmethyl, and cyclobutylmethyl, wherein each of the Cvs alkyl, C2.6 alkenyl, C2.6 alkynyl, -S-fC^ alkyl), and 0ν6 alkoxy is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, -N(Ra)(Rb), 0^4 alkoxy, Cm haloalkoxy, and -S-(Cm alkyl); and wherein each of the C3.7 cycloalkyl, 4- to 7membered heterocycloalkyl, C3.7 cycloalkoxy, 5- or 6-membered heteroaryl, cyclopropylmethyl, and cyclobutylmethyl of T1, T2, and T3 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, oxo, -N(Ra)(Rb), C(=O)OH, -C(=O)-CM alkyl, -C(=O)-O-CM alkyl, -C(=O)-N(Ra)(Rb), Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, Cm haloalkoxy, and -S-(CV4 alkyl);
L1 is selected from the group consisting of O, S, NH, N(Cm alkyl), N(-Cv2 alkyl-C3.4 cycloalkyl), and N(C3.6 cycloalkyl);
each of Ra and Rb is independently selected from the group consisting of H, Cm alkyl, C3.7 cycloalkyl (e.g., cyclopropyl, cyclobutyl, bicyclo[1.1.1]pentan-1-yl, or bicyclo[ 1.1.1 ]pentan-2yl), and cyclopropylmethyl;
or Ra and Rb together with the N atom to which they are attached form 4- to 7-membered 25 heterocycloalkyl (e.g., azetidinyl, pyrrolidinyl, or 7-azabicyclo[2.2.1]heptan-7-yl) optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, oxo, -NH2, -NH(Cm alkyl), -N(CH alkyl)2, -C(=O)OH, -C(=O)Cm alkyl, -C(=O)-O-Cm alkyl, -C(=O)-NH2, -C(=O)-NH(CM alkyl), - C(=0)-N(Cm alkyl)2, Cm alkyl, Cm haloalkyl, Ci.4 hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, -S-(Cm alkyl), and Cm haloalkoxy;
each of Rc and Rd is independently Cm alkyl, C3.4 cycloalkyl-CM alkyl-, or C3.4 cycloalkyl;
Q1 is selected from the group consisting of Q1a, Q1b, Q1c Q1d, and Q1e:
provided (a) that a ring carbon atom of the Q1 ring is attached to the benzene ring of
Formula I and (b) that when L1 is NH, then the Q1 ring is substituted with at least one non-H R9, pW p11 p12 p13 p9A p10A pWB p11A p12A θρ ρ13Α.
each of X1 and X2 is independently O or S;
each of R1, R2, R3, and R4 is independently selected from the group consisting of H, halogen, -OH, -NO2, -CN, -SF5, C^e alkyl, haloalkyl, C^e haloalkoxy, C2.6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, a 4- to 10-membered heterocycloalkyl, -N(RS)(R6), -N(R7)(C(=O)R8), C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -N(R7)(S(=O)2R8), -S(=O)2-N(R5)(R6), -SR8, and -OR8, wherein each of the Ον6 alkyl, C3.7 cycloalkyl, and heterocycloalkyl is optionally substituted with
1,2, or 3 substituents each independently selected from the group consisting of halogen, -CN, 15 oxo, -OH, CV4 alkyl, C14 alkoxy, C^ haloalkyl, Ci_4 haloalkoxy, C3.6 cycloalkyl, -N(R5)(R6), N(R7)(C(=O)R8), -C(=O)-OR8, -C(=O)H, -C(=O)R8, -C(=O)N(R5)(R6), -N(R7)(S(=O)2R8), -S(=O)2N(R5)(R6), -SR8, and -OR8;
or R2 and R4 together with the two carbon atoms to which they are attached form a fused
5- or 6-membered heteroaryl, a fused 5- or 6-membered heterocycloalkyl ring, a fused 5- or 620 membered cycloalkyl ring, or a fused benzene ring, wherein each of the fused rings is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halo, -CN, -OH, CM alkyl, C^ alkoxy, Ci_4 haloalkyl, and Ci-4 haloalkoxy, and wherein the fused heterocycloalkyl ring or fused cycloalkyl ring is further optionally substituted with 1,2, or 3 oxo;
R5is H, Ci-4 alkyl, C14 haloalkyl, or C3.7 cycloalkyl;
R6 is H or selected from the group consisting of C^ alkyl, haloalkyl, C3.7 cycloalkyl, a 4- to 10-membered heterocycloalkyl, C6.10 aryl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyO-C^ alkyl-, (4- to 10-membered heterocycloalkyl)-Ci_4 alkyl-, (C6.10 aryl)-CV4 alkyl-, and (5- to 10-membered heteroaryl)-CV4 alkyl-, wherein each of the sélections from the group is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of -OH, -CN, Ον4 alkyl, C3.7 cycloalkyl, Ci_4 hydroxylalkyl, -S-Ci_4 alkyl, -C(=O)H,
-C(=O)-C,, alkyl, -0(=0)-0-0,, alkyl, -C(=0)-NH2, -C(=0)-N(C„ alkyl)2, C„ haloalkyl, C„ alkoxy, and CM haloalkoxy;
or R5 and R6 together with the N atom to which they are attached form a 4- to 10membered heterocycloalkyl or a 5- to 10-membered heteroaryl, each optionally substituted with
1,2, 3, 4, or 5 substituents each independently selected from the group consisting of halogen,
-OH, oxo, -C(=O)H, -0(=0)-0).4 alkyl, -C(=0)0H, -0(=0)-0-0).4 alkyl, -C(=0)-NH2, -C(=0)-N(C).
4 alkyl)2, -CN, 0).4 alkyl, C).4 alkoxy, Cm hydroxylalkyl, C).4 haloalkyl, and C).4 haloalkoxy; R7 is selected from the group consisting of H, C).4 alkyl, and C3.7 cycloalkyl; R8 is selected from the group consisting of alkyl, C3-7 cycloalkyl, a 4- to 10- membered heterocycloalkyl, C6.)0aryl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-C).4 alkyl-, (4- to 10-membered heterocycloalkyl)-C)_4 alkyl-, (C6.10 aryl)-C).4 alkyl-, and (5- to 10membered heteroaryl)-C).4 alkyl-, wherein each of the sélections from the group is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -CF3, -CN, -OH, oxo, -S-C).4 alkyl, C^ alkyl, C)_4 haloalkyl, C2.6 alkenyl, C2.6 alkynyl,
C3.7 cycloalkyl, C).4 alkoxy, and C)_4 haloalkoxy;
each R9 and R12 is independently selected from the group consisting of halogen, -OH, CN, -SF5, -N02, C)-6 alkyl, C).6 haloalkyl, C).6 hydroxylalkyl, C).6 alkoxy, C).6 haloalkoxy, C3.7 cycloalkyl, C2.6 alkenyl, C2.6 alkynyl, a 4- to 10-membered heterocycloalkyl, a 5- to 10membered heteroaryl, (C3.7 cycloalkyl)-C).4 alkyl-, (4- to 10-membered heterocycloalkyl)-C)_4 alkyl-, (C6.10 aryl)-C).4 alkyl-, (5- to 10-membered heteroaryl)-C).4 alkyl-, -N(R5)(R6), N(R7)(C(=O)R8), -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=0)-R8, -C(=O)-OR8, -SR8, and -OR8, wherein each of the C^ alkyl, C3.7 cycloalkyl, 4- to 10-membered heterocycloalkyl, 5- to 10membered heteroaryl, (C3.7 cycloalkyl)-C).4 alkyl-, (4- to 10-membered heterocycloalkyl)-C)_4 alkyl-, (C6.10 aryl)-CV4 alkyl-, and (5- to 10-membered heteroaryl)-C).4 alkyl- is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -N02, C)_4 alkyl, C).4 hydroxylalkyl, C).4 alkoxy, -N(R5)(R6), -S-(CM alkyl), -S(=O)2-(C).4 alkyl), C6-i0 aryloxy, [(C6.10 aryl)-C).4 alkyloxy- optionally substituted with 1 or 2 C).4 alkyl], oxo, -C(=O)H, -C(=O)-CM alkyl, -C(=O)O-CM alkyl, -C(=O)NH2, -NHC(=0)H, -NHC(=O)- (C).4 alkyl), C3.7 cycloalkyl, a 5- or 6-membered heteroaryl, C).4 haloalkyl, and C)_4 haloalkoxy;
each of R10, R11 and R13 is independently selected from the group consisting of halogen,
-OH, -CN, -SF5, -N02j C)-6 alkyl, C).6 haloalkyl, C).6 hydroxylalkyl, C).6 alkoxy, C).6 haloalkoxy, C3.7 cycloalkyl, C2.6 alkenyl, C2.6 alkynyl, C6.)o aryl, a 4- to 10-membered heterocycloalkyl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-C).4 alkyl-, (4- to 10-membered heterocycloalkyl)-C).4 alkyl-, (C6.10 aryl)-C).4 alkyl-, (5- to 10-membered heteroaryl)-C)_4 alkyl-, -N(R5)(R6), 35 N(R7)(C(=O)R8), -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=0)-R8, -C(=O)-OR8, -SR8, and -OR8, wherein each of the C).6 alkyl, C3.7 cycloalkyl, C6.10 aryl, 4- to 10-membered heterocycloalkyl, 5to 10-membered heteroaryl, (C3.7 cycloalkyl)-C)_4 alkyl-, (4- to 10-membered heterocycloalkyl)-
C)-4 alkyl-, (C6.10 aryl)-C)_4 alkyl-, and (5- to 10-membered‘heteroaryl)-C).4 alkyl- is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -NO2, CM alkyl, hydroxylalkyl, CV4 alkoxy, -N(R5)(R6), -3-(0,.4 alkyl),
-3(=0)2-(0,-4 alkyl), C6-10 aryloxy, [(C6-io aryl)-C,.4 alkyloxy- optionally substituted with 1 or 2 0,.4 alkyl], oxo, -C(=O)H, -0(=0)-0,.4 alkyl, -0(=0)0-0,.4 alkyl, -C(=O)NH2, -NHC(=0)H, -NHC(=O)(0,.4 alkyl), C3.7 cycloalkyl, a 5- or 6-membered heteroaryl, haloalkyl, and C,.4 haloalkoxy;
each of R9A and R10A is independently selected from the group consisting of H, C,.6 alkyl, C,.6 hydroxylalkyl, C2.6 alkenyl, -S(=0)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=0)-R8, -C(=O)-OR8, SR15, -C(R14)2-OH, -C(R14)2-OS(=O)2H, -C(R14)2-OP(=O)(OH)2j -C(R14)2-0R15, -C(R14)2-OC(=O)R15, -C(R14)2-N(R5)(R6), each of R10B, R11A, R12A, and R13A is independently selected from the group consisting of H, C,.6 alkyl, C,.6 haloalkyl, 0,-6 hydroxylalkyl, C3.7 cycloalkyl, C3.6 alkenyl, C3.6 alkynyl, C6.10 aryl, a 4- to 10-membered heterocycloalkyl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-C,.4 alkyl-, (4- to 10-membered heterocycloalkylj-C,^ alkyl-, (C6.10 aryl)-Ci.4 alkyl-, (5- to 10membered heteroaryl)-CM alkyl-, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, and -C(=O)OR8, wherein each of the 0,.6 alkyl, C3.7 cycloalkyl, C6.10 aryl, 4- to 10-membered heterocycloalkyl, 5- to 10-membered heteroaryl, (C3.7 cycloalkylj-C,^ alkyl-, (4- to 10-membered heterocycloalkyl)-Ci-4 alkyl-, (C6.10 arylj-C,^ alkyl-, and (5- to 10-membered heteroarylj-C,^ alkyl-, is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -N02, 0,.4 alkyl, 0,.4 hydroxylalkyl, CM alkoxy, N(RS)(R6), -3-(0,.4 alkyl), -S(=O)2-(CM alkyl), C6.,0 aryloxy, [(C6-10 aryl)-C,.4 alkyloxy- optionally substituted with 1 or 2 C,.4 alkyl], oxo, -C(=0)H, -C(=0)-C,.4 alkyl, -0(=0)0-0,.4 alkyl, C(=O)NH2, -NHC(=0)H, -NHC(=O)-(C,.4 alkyl), -00(=0)-0,.4 alkyl, C3.7 cycloalkyl, a 5- or 6membered heteroaryl, C1.4 haloalkyl, and C1.4 haloalkoxy;
each R14 is independently H or selected from the group consisting of C1-10 alkyl, C3.,4 cycloalkyl, Ο2.,0 alkenyl, C2.10 alkynyl, C6.,o aryl, 4- to 10-membered heterocycloalkyl, 5- to 10membered heteroaryl, (03.14 cycloalkyl)-C,.,o alkyl-, (4- to 14-membered heterocycloalkyl)-C,.,o alkyl-, (C6.,0 aryl)-C,.,o alkyl-, (5- to 10-membered heteroaryl)-C,.10 alkyl-, wherein each of the sélections of the group is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -N02, 0,.4 alkyl, C-1.4 hydroxylalkyl, C^ alkoxy, -N(R5)(R6), -N(R7)C(=O)R8, -N(R7)C(=0)0R8, -N(R7)S(=0)2R8, S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -SR8, -OR8, -S(=0)2-R8, C6.10 aryloxy, [(C6.10 aryl)-Ci.4 alkyloxy- optionally substituted with 1 or 2 C1.4 alkyl], oxo, -C(=O)H, NHC(=O)H, C3.7 cycloalkyl, a 5- or 6-membered heteroaryl, 0-|.4 haloalkyl, and C14 haloalkoxy;
R15 is selected from the group consisting of C,.^ alkyl, C3.14 cycloalkyl, C2.20 alkenyl, C2.20 alkynyl, C6-io aryl, 4- to 14-membered heterocycloalkyl, 5- to 10-membered heteroaryl, (C3.14 cycloalkyl)-Ci_20 alkyl-, (4- to 10-membered heterocycloalkyl)-Ci.20 alkyl-, (C6.10 arylj-C,^ alkyl-, (5- to 10-membered heteroarylj-C^o alkyl-, wherein each of the sélections of the group is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -NO2, Cm alkyl, CM hydroxylalkyl, Cm alkoxy, -N(RS)(R6), N(R7)C(=O)R8, -N(R7)C(=O)OR8, -N(R7)S(=O)2Rs, -S(=O)2N(Rs)(R6), -C(=O)-N(R5)(R6), -C(=O)R8, -C(=O)-OR8, -SR8, -OR8, -S(=O)2-R8, C6.10 aryloxy, [(C6-io aryl)-CM alkyloxy- optionally substituted with 1 or 2 Cm alkyl], oxo, -C(=O)H, -NHC(=O)H, C3.7 cycloalkyl, a 5- or 65 membered heteroaryl, Cm haloalkyl, and Cm haloalkoxy;
t1 is 0, 1, or 2;
t2 is 0 or 1 ; and t3 is 0, 1, or 2.
The présent invention also provides a composition (e.g., a pharmaceutical composition) comprising a compound of Formula I or a pharmaceutically acceptable sait thereof.
Compounds of Formula I and pharmaceutically acceptable salts thereof are D1 modulators (e.g., D1 agonists or partial agonists). According, the présent invention further provides a method for treating a D1-mediated (or D1-associated) disorder (e.g., cognitive impairment such as cognitive impairment associated with schizophrenia or cognitive impairment 15 associated with Alzheimer’s disease; schizophrenia; Alzheimer’s disease; or Parkinson’s disease), comprising administering to a mammal (e.g., a human) in need thereof an amount of a compound of Formula I or a pharmaceutically acceptable sait thereof effective in modulating (e.g., agonizing or partially agonizing) D1.
As used herein, the term “n-membered” where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n. For example, pyridine is an example of a 6-membered heteroaryl ring and thiophene is an example of a 5-membered heteroaryl group.
At various places in the présent spécification, substituents of compounds of the invention are disclosed in groups or in ranges. It is specifically intended that the invention include each 25 and every individual subcombination of the members of such groups and ranges. For example, the term “Cm alkyl” is specifically intended to include Ci alkyl (methyl), C2 alkyl (ethyl), C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl. For another example, the term “a 5- to 10-membered heteroaryl group” is specifically intended to include any 5-, 6-, 7-, 8-, 9- or 10-membered heteroaryl group.
As used herein, the term alkyl is defined to include saturated aliphatic hydrocarbons including straight chains and branched chains. In some embodiments, the alkyl group has 1 to carbon atoms, 1 to 10 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. For example, the term Cmo alkyl” refers to linear or branched aliphatic hydrocarbon chains of 1 to 20 carbon atoms; the term ’Ci.w alkyl” refers to linear or branched aliphatic hydrocarbon chains of 1 to 10 carbon atoms. For another example, as used herein, the term Cm alkyl,” as well as the alkyl moieties of other groups referred to herein (e.g., Cm alkoxy) refers to linear or branched radicale of 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyi, isopropyl, n-butyl, isobutyl, secbutyl, tert-butyl, n-pentyl, or n-hexyl). For yet another example, the term Cm alkyl” refers to linear or branched aliphatic hydrocarbon chains of 1 to 4 carbon atoms; the term Cm alkyl”
refers to linear or branched aliphatic hydrocarbon chains of 1 to 3 carbon atoms; the term C^ alkyl” refers to linear or branched aliphatic hydrocarbon chains of 1 to 2 carbon atoms; and the term Ci alkyl” refers to methyl. An alkyl group optionally can be substituted by one or more (e.g.
to 5) suitable substituents.
As used herein, the term alkenyl refers to aliphatic hydrocarbons having at least one carbon-carbon double bond, including straight chains and branched chains having at least one carbon-carbon double bond. In some embodiments, the alkenyl group has 2 to 20 carbon atoms, 2 to 10 carbon atoms, 2 to 6 carbon atoms, 3 to 6 carbon atoms, or 2 to 4 carbon atoms.
For example, as used herein, the term C2.2o alkenyl refers to straight or branched chain unsaturated radicals (having at least one carbon-carbon double bond) of 2 to 20 carbon atoms; the term C2.10 alkenyl refers to straight or branched chain unsaturated radicals (having at least one carbon-carbon double bond) of 2 to 10 carbon atoms; the term C3.6 alkenyl refers to straight or branched chain unsaturated radicals (having at least one carbon-carbon double bond) of 3 to 4 carbon atoms; and the term C2.4 alkenyl refers to straight or branched chain unsaturated radicals (having at least one carbon-carbon double bond) of 2 to 4 carbon atoms. For another example, the term C2.6 alkenyl means straight or branched chain unsaturated radicals (having at least one carbon-carbon double bond) of 2 to 6 carbon atoms, including, but not limited to, ethenyl, 1-propenyl, 2-propenyl (allyl), isopropenyl, 2-methyl-1-propenyl, 1butenyl, 2-butenyl, and the like.. An alkenyl group optionally can be substituted by one or more (e.g. 1 to 5) suitable substituents. When the compounds of Formula I contain an alkenyl group, the alkenyl group may exist as the pure E form, the pure Z form, or any mixture thereof.
As used herein, the term alkynyl refers to aliphatic hydrocarbons having at least one carbon-carbon triple bond, including straight chains and branched chains having at least one carbon-carbon triple bond. In some embodiments, the alkynyl group has 2 to 20, 2 to 10, 2 to 6, 25 or 3 to 6 carbon atoms. For example, as used herein, the term C2.6 alkynyl” refers to straight or branched hydrocarbon chain alkynyl radicals as defined above, having 2 to 6 carbon atoms. For another rexample, the term C2.20 alkynyl” is used herein to mean straight or branched hydrocarbon chain alkynyl radicals as defined above, having 2 to 20 carbon atoms; the term C2. 10 alkynyl” refers to straight or branched hydrocarbon chain alkynyl radicals as defined above, 30 having 2 to 10 carbon atoms; and the term C3.6 alkynyl” refers to straight or branched hydrocarbon chain alkynyl radicals as defined above, having 3 to 6 carbon atoms. An alkynyl group optionally can be substituted by one or more (e.g. 1 to 5) suitable substituents.
As used herein, the term cycloalkyl” refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings (e.g., monocyclics such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, or bicyclics including spiro, fused, or bridged Systems (such as bicyclo[1.1.1]pentanyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl or bicyclo[5.2.0]nonanyl, decahydronaphthalenyl, etc.). The cycloalkyl group has 3 to 15 carbon atoms. In some embodiments the cycloalkyl may optionally contain
one, two or more non-cumulative non-aromatic double or triple bonds and/or one to three oxo groups. In some embodiments, the bicycloalkyl group has 6 to 14 carbon atoms. For example, the term C3.14 cycloalkyl” refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings of 3 to 14 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[1.1 .Ijpentanyl, or cyclodecanyl); and the term C3.7 cycloalkyl” refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings of 3 to 7 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[1.1,1]pentan-1-yl, or bicycio[1.1.1]pentan-2-yl). For another example, the term C3.6 cycloalkyl” refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings of 3 to 6 ring-forming carbon atoms. For yet another example, the term C3.4 cycloalkyl” refers to cyclopropyl or cyclobutyl. Also included in the définition of cycloalkyl are moieties that hâve one or more aromatic rings (including aryl and heteroaryl) fused to the cycloalkyl ring, for example, benzo or thienyl dérivatives of cyclopentane, cyclopentene, cyclohexane, and the like (e.g., 2,3-dihydro-1 H15 indene-1-yl, or 1 H-inden-2(3/4)-one-1-yl). The cycloalkyl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term aryl refers to ali-carbon monocyclic or fused-ring polycyclic aromatic groups having a conjugated pi-electron system. The aryl group has 6 or 10 carbon atoms in the ring(s). Most commonly, the aryl group has 6 carbon atoms in the ring. For example, as used herein, the term “C6-io aryl” means aromatic radicals containing from 6 to 10 carbon atoms such as phenylor naphthyl. The aryl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term “heteroaryl” refers to monocyclic or fused-ring polycyclic aromatic heterocyclic groups with one or more heteroatom ring members (ring-forming atoms) 25 each independently selected from O, S and N in at least one ring. The heteroaryl group has 5 to 14 ring-forming atoms, including 1 to 13 carbon atoms, and 1 to 8 heteroatoms selected from O, S, and N. In some embodiments, the heteroaryl group has 5 to 10 ring-forming atoms including one to four heteroatoms. The heteroaryl group can also contain one to three oxo or thiono (i.e. =S) groups. In some embodiments, the heteroaryl group has 5 to 8 ring-forming atoms including one, two or three heteroatoms. For example, the term 5-membered heteroaryl” refers to a monocyclic heteroaryl group as defined above with 5 ring-forming atoms in the monocyclic heteroaryl ring; the term 6-membered heteroaryl” refers to a monocyclic heteroaryl group as defined above with 6 ring-forming atoms in the monocyclic heteroaryl ring; and the term 5- or 6-membered heteroaryl” refers to a monocyclic heteroaryl group as defined above with 5 or 6 ring-forming atoms in the monocyclic heteroaryl ring. For another example, term 5or 10-membered heteroaryl” refers to a monocyclic or bicyclic heteroaryl group as defined above with 5, 6, 7, 8, 9 or 10 ring-forming atoms in the monocyclic or bicyclic heteroaryl ring. A heteroaryl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
Examples of monocyclic heteroaryls include those with 5 ring-forming atoms including one to three heteroatoms or those with 6 ring-forming atoms including one, two or three nitrogen heteroatoms. Examples of fused bicyclic heteroaryls include two fused 5- and/or 6-membered monocyclic rings including one to four heteroatoms.
Examples of heteroaryl groups include pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, thienyl, furyl, imidazolyl, pyrrolyl, oxazolyl (e.g., 1,3-oxazolyl, 1,2-oxazolyl), thiazolyl (e.g., 1,2thiazolyl, 1,3-thiazolyl), pyrazolyl, tetrazolyl, triazolyl (e.g., 1,2,3-triazolyl, 1,2,4-triazolyl), oxadiazolyl (e.g., 1,2,3-oxadiazolyl), thiadiazolyl (e.g., 1,3,4-thiadiazolyl), quinolyl, isoquinolyl, benzothienyl, benzofuryl, indolyl, 1 /7-imidazo[4,5-c]pyridinyl, imidazo[1,2-a]pyridinyl, 1H10 pyrrolo[3,2-c]pyridinyl, imidazo[1,2-a]pyrazinyl, imidazo[2,1-c][1,2,4]triazinyl, imidazo[1,5a]pyrazinyl, imidazo[1,2-a]pyrimidinyl, 1 H-indazolyl, 9H-purinyl, imidazo[1,2-a]pyrimidinyl, [1,2,4]triazolo[1,5-a]pyrimidinyl, [1,2,4]triazolo[4,3-b]pyridazinyl, isoxazolo[5,4-c]pyridazinyl, isoxazolo[3,4-c]pyridazinyl, pyridone, pyrimidone, pyrazinone, pyrimidinone, 7H-imidazol-2(3/-/)one, ïH-pyrrole-2,5-dione, 3-oxo-2H-pyridazinyl, 1 H-2-oxo-pyrimidinyl, 1H-2-oxo-pyridinyl,
2,4(1 H,3/-/)-dioxo-pyrimidinyl, 1 /-/-2-oxo-pyrazinyl, and the like. The heteroaryl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term “heterocycloalkyl” refers to a monocyclic or polycyclic [including 2 or more rings that are fused together, including spiro, fused, or bridged Systems, for example, a bicyclic ring system], saturated or unsaturated, non-aromatic 4- to 15-membered ring system 20 (such as a 4- to 14-membered ring system, 4- to 10-membered ring system, 5- to 10-membered ring system, 4- to 7-membered ring system, 4- to 6-membered ring system, or 5- to 6membered ring system), including 1 to 14 ring-forming carbon atoms and 1 to 10 ring-forming heteroatoms each independently selected from O, S and N. For example, ; the term 4- to 14membered heterocycloalkyl” refers to a monocyclic or polycyclic, saturated or unsaturated, non25 aromatic 4- to 14-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from O, S and N; and the term “4- to 10-membered heterocycloalkyl” refers to a monocyclic or polycyclic, saturated or unsaturated, non-aromatic 4to 10-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from O, S and N. For another example, the term 4- to 6-membered heterocycloalkyl” refers to a monocyclic or polycyclic, saturated or unsaturated, non-aromatic 4to 6-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from O, S and N; and the term 5- to 6-membered heterocycloalkyl” refers to a monocyclic or polycyclic, saturated or unsaturated, non-aromatic 5- to 6-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from O, S and N. The heterocycloalkyl group optionally can be substituted by 1 or more (e.g., 1 to
5) suitable substituents. The heterocycloalkyl group can also optionally include one to three oxo or thiono groups.
Examples of such heterocycloalkyl rings include azetidinyl, tetrahydrofuranyl, imidazolidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxazolidinyl, thiazolidinyl, pyrazolidinyl, thiomorpholinyl, tetrahydrothiazinyl, tetrahydrothiadiazinyl, morpholinyl, oxetanyl, tetrahydrodiazinyl, oxazinyl, oxathiazinyl, quinuclidinyl, chromanyl, isochromanyl, benzoxazinyl, 7-azabicyclo[2.2.1]heptan-1-yl, 7-azabicyclo[2.2.1]heptan-2-yl, 7-azabicyclo[2.2.1]heptan-7-yl,
2-azabicyclo[2.2.1]heptan-3-on-2-yl, 3-azabicyclo[3.1,0]hexanyl, 3-azabicyclo[4.1.0]heptanyl and the like. Further examples of heterocycloalkyl rings include tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, imidazolidin-1-yl, imidazolidin-2-yl, imidazolidin-4-yl, pyrrolidin-1 -yl, pyrrolidin-2-yl, pyrrolidin-3-yl, piperidin-1 -yl, piperidin-2-yl, piperidin-3-yl, piperidin-4-yl, piperazin-1-yl, piperazin-2-yl, 1,3-oxazolidin-3-yl, 1,4-oxazepan-1-yl, isothiazolidinyl, 1,3thiazolidin-3-yl, 1,2-pyrazolidin-2-yl, 1,2-tetrahydrothiazin-2-yl, 1,3-thiazinan-3-yl, ΐ,2tetrahydrodiazin-2-yl, 1,3-tetrahydrodiazin-1-yl, 1,4-oxazin-4-yl, oxazolidinonyi, 2-oxo-piperidinyl (e.g., 2-oxo-piperidin-1-yl), and the like. Also included in the définition of heterocycloalkyl are moieties that hâve one or more aromatic rings (including aryl and heteroaryl) fused to the nonaromatic heterocycloalkyl ring, for example pyridinyl, pyrimidinyl, thiophenyl, pyrazolyl, phthalimidyl, naphthalimidyl, and benzo dérivatives of the nonaromatic heterocycloalkyl rings. Examples of such aromatic-fused heterocycloalkyl groups include indolinyl, isoindolinyl, isoindolin-1 -one-3-yl, 5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-yl, 6,7-dihydro-5/-/-pyrrolo[3,4aQpyrimidin-6-yl, 4,5,6,7-tetrahydrothieno[2,3-c]pyridine-5-yl, 5,6-dihydrothieno[2,3-c]pyridin7(4H)-one-5-yl, 1,4,5,6-tetrahydropyrrolo[3,4-c]pyrazol-5-yl, and 3,4-dihydroisoquinolin-1(2A/)one-3-yl groups. The heterocycloalkyl group is optionally substituted by 1 or more (e.g., 1 to 5) suitable substituents. Examples of heterocycloalkyl groups include 5- or 6-membered monocyclic rings and 9- or 10-membered fused bicyclic rings.
As used herein, the term halo or halogen group is defined to include fluorine, chlorine, bromine or iodine.
As used herein, the term “haloalkyl” refers to an alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by a halogen atom). For example, the term “C^ haloalkyl” refers to a Ci^ alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by a halogen atom). For another example, the term “Cm haloalkyl” refers to a Cm alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by a halogen atom); the term “C^ haloalkyl” refers to a Ci_3 alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by a halogen atom); and the term “C^ haloalkyl” refers to a alkyl group (i.e. methyl or ethyl) having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by a halogen atom). For yet another example, the term “Ci haloalkyl” refers
to a methyl group having one, two, or three halogen substituents. Examples of haloalkyl groups include CF3, C2F5, CHF2, CH2F, CH2CF3, CH2CI and the like.
As used herein, the term “halocycloalkyl” refers to a cycloalkyl group having one or more halogen substituents (up to perhalocycloalkyl, i.e., every hydrogen atom of the cycloalkyl group has been replaced by a halogen atom). For example, the term “C3.4 halocycloalkyl” refers to a cyclopropyl or cyclobutyl group having one or more halogen substituents. An example of halocycloalkyl is 2-fluorocyclopropan-1-yl.
As used herein, the term “alkoxy” or “alkyloxy” refers to an -O-alkyl group. For example, the term “C^ alkoxy” or “C^e alkyloxy” refers to an -O-(Ci_6 alkyl) group; and the term “C^ alkoxy” or “CM alkyloxy” refers to an -O-(C1.4 alkyl) group; For another example, the term “Ci.2 alkoxy” or “Ci_2 alkyloxy” refers to an -O-(Ci_2 alkyl) group. Examples of alkoxy include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), tert-butoxy, and the like. The alkoxy or alkyloxy group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used here, the term “haloalkoxy” refers to an -O-haloalkyl group. For example, the term “Ci_6 haloalkoxy” refers to an -O-(Ci_6 haloalkyl) group. For another example, the term “Ci_4 haloalkoxy” refers to an -O-(C1.4 haloalkyl) group; and the term “Ci-2 haloalkoxy” refers to an -O(Ci.2 haloalkyl) group. For yet another example, the term “C! haloalkoxy” refers to a methoxy group having one, two, or three halogen substituents. An example of haloalkoxy is -OCF3 or OCHF2.
As used herein, the term “cycloalkoxy” or “cycloalkyloxy” refers to an -O-cycloalkyl group. For example,, the term “C3.7 cycloalkoxy” or “C3.7 cycloalkyloxy” refers to an -O-(C3.7 cycloalkyl) group. For another example,, the term “C3.6 cycloalkoxy” or “C3.6 cycloalkyloxy” refers to an -O-(C3-6 cycloalkyl) group. Examples of cycloalkoxy include C3.6 cycloalkoxy (e.g., cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexanoxy, and the like). The cycloalkoxy or cycloalkyloxy group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used here, the term “C6-i0 aryloxy” refers to an -O-(C6.i0 aryl) group. An example of a C6.10 aryloxy group is -O-phenyl [i.e., phenoxy]. The C6_i0aryloxy y group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term “fluoroalkyl” refers to an alkyl group having one or more fluorine 30 substituents (up to perfluoroalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by fluorine). For example, the term “Ci_2 fluoroalkyl” refers to a Cv2 alkyl group having one or more fluorine substituents (up to perfluoroalkyl, i.e., every hydrogen atom of the Ον2 alkyl group has been replaced by fluorine). For another example, the term “Ci fluoroalkyl” refers to a Ci alkyl group (i.e., methyl) having 1,2, or 3 fluorine substituents). Examples of fluoroalkyl groups include CF3, C2F5, CH2CF3, CHF2, CH2F, and the like.
As used here, the term “fluoroalkoxy” refers to an -O-fluoroalkyl group. For example, the term “Ci_2 fluoroalkoxy” refers to an -O-Ci.2 fluoroalkyl group. For another example, the term “Ci fluoroalkoxy” refers to a methoxy group having one, two, or three fluorine substituents. An example of Ci fluoroalkoxy is -OCF3 or -OCHF2.
As used herein, the term hydroxylalkyl or hydroxyalkyl refers to an alkyl group having one or more (e.g., 1,2, or 3) OH substituents. The term “C^ hydroxylalkyl” or “C^ hydroxyalkyl refers to a alkyl group having one or more (e.g., 1,2, or 3) OH substituents. The term “Cm hydroxylalkyl” or “Cm hydroxyalkyl” refers to a Cm alkyl group having one or more (e.g., 1,2, or 3) OH substituents; the term “C-i-3 hydroxylalkyl” or “C1-3 hydroxyalkyl” refers to a Ci-3 alkyl group having one or more (e.g., 1,2, or 3) OH substituents; and the term “Cm hydroxylalkyl” or “Cm hydroxyalkyl” refers to a Cm alkyl group having one or more (e.g., 1, 2, or 3) OH substituents. An example of hydroxylalkyl is -CH2OH or -CH2CH2OH.
As used herein, the term “cyanoalkyl” refers to an alkyl group having one or more (e.g., 1,2, or 3) -CN (i.e. -C^N or cyano) substituents. For example, The term “Cm cyanoalkyl” refers to a Cm alkyl group having one or more (e.g., 1, 2, or 3) -CN substituents. An Example of cyanoalkyl is -CH2-CN or -CH2CH2-CN.
As used herein, the term “oxo” refers to =0. When an oxo is substituted on a carbon atom, they together form a carbonyl moiety [-C(=O)-J. When an oxo is substituted on a sulfur atom, they together form a sulfinyl moiety (-S(=O)-]; when two oxo groups are substituted on a sulfur atom, they together form a sulfonyl moiety [-S(=O)2-J.
As used herein, the term “optionally substituted” means that substitution is optional and therefore includes both unsubstituted and substituted atoms and moieties. A “substituted” atom or moiety indicates that any hydrogen on the designated atom or moiety can be replaced with a sélection from the indicated substituent group (up to that every hydrogen atom on the designated atom or moiety is replaced with a sélection from the indicated substituent group), provided that the normal valency of the designated atom or moiety is not exceeded, and that the substitution results in a stable compound. For example, if a methyl group (i.e., CH3) is optionally substituted, then up to 3 hydrogen atoms on the carbon atom can be replaced with substituent groups.
As used herein, unless specified, the point of attachment of a substituent can be from any suitable position of the substituent. For example, piperidinyl can be piperidin-1 -yl (attached through the N atom of the piperidinyl), piperidin-2-yl (attached through the C atom at the 2position of the piperidinyl), piperidin-3-yl (attached through the C atom at the 3-position of the piperidinyl), or piperidin-4-yl (attached through the C atom at the 4-position of the piperidinyl). For another example, pyridinyl (or pyridyl) can be 2-pyridinyl (or pyridin-2-yl), 3-pyridinyl (or pyridin-3-yl), or 4-pyridinyl (or pyridin-4-yl).
When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any of the ring-forming atoms in that ring that are substitutable (i.e., bonded to one or more hydrogen atoms), unless otherwise specifized or otherwise implicit from the context. For example, as shown in Formula a-101 below, R9 may be
bonded to either of the two ring carbon atoms each of which bears a hydrogen atom (but not shown), but not to the N to which R9A is bonded (even wherein R9A is H). For another example, as shown in Formula a-102 below, R9 may be bonded to either of the two ring carbon atoms each of which bears a hydrogen atom (but not shown), but not to the N that is shown to be bonded to a H atom. For yet another example, as shown in Formula a-103 below, R12 may be bonded to either of the two ring carbon atoms each of which bears a hydrogen atom (but not shown), but not the the ring carbon atom that is shown to be bonded to a H atom.
a-101
R12 H ξ Ή Au a-103
When a substituted or optionally substituted moiety is described without indicating the atom via which such moiety is bonded to a substituent, then the substituent may be bonded via any appropriate atom in such moiety. For example in a substituted arylalkyl, a substituent on the arylalkyl [e.g., (C6-io arylj-C^ alkyl-] can be bonded to any carbon atom on the alkyl part or on the aryl part of the arylalkyl. Combinations of substituents and/or variables are permissible only if such combinations resuit in stable compounds.
As noted above, the compounds of Formula I may exist in the form of pharmaceutically acceptable salts such as acid addition salts and/or base addition salts of the compounds of Formula I. The phrase “pharmaceutically acceptable salt(s)”, as used herein, unless otherwise indicated, includes acid addition or base salts which may be présent in the compounds of
Formula I.
Pharmaceutically acceptable salts of the compounds of Formula I include the acid addition and base salts thereof.
Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulfate/sulfate, borate, camphorsulfonate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulfate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stéarate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinofoate salts.
Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnésium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
Hemisalts of acids and bases may also be formed, for example, hemisulfate and hemicalcium salts.
For a review on suitable salts, see Handbook of Pharmaceutical Salts: Properties, Sélection, and Use” by Stahl and Wermuth (Wiley-VCH, 2002). Methods for making pharmaceutically acceptable salts of compounds of Formula I are known to one of skill in the art.
As used herein the terms “Formula I”, “Formula I or pharmaceutically acceptable salts thereof”, “pharmaceutically acceptable salts of the compound or the sait [of Formula I]” are defined to include ail forms of the compound of Formula I, including hydrates, solvatés, isomers (including for example rotational stereoisomers), crystalline and non-crystalline forms, isomorphs, polymorphe, métabolites, and prodrugs thereof.
As it is known to the person skilled in the art, amine compounds (i.e., those comprising one or more nitrogen atoms), for example tertiary amines, can form A/-oxides (also known as amine oxides or amine /V-oxides). An /V-oxide has the formula of (R100R200R300)N+-O~ wherein the parent amine (R100R200R300)N can be for example, a tertiary amine (for example, each of R100, R200, R300 is independently alkyl, arylalkyl, aryl, heteroaryl, or the like), a heterocyclic or heteroaromatic amine [for example, (R100R200R300)N together forms 1 -alkylpiperidine, 1alkylpyrrolidine, 1-benzylpyrrolidine, or pyridine]. For instance, an imine nitrogen, especially heterocyclic or heteroaromatic imine nitrogen, or pyridine-type nitrogen (4=N-|-) atom [such as a nitrogen atom in pyridine, pyridazine, or pyrazine], can be /V-oxidized to form the /V-oxide ocomprising the group ( Thus, a compound according to the présent invention comprising one or more nitrogen atoms (e.g., an imine nitrogen atom) may be capable of forming an /V-oxide thereof (e.g., mono-/V-oxides, bis-/V-oxides or multi-/V-oxides, or mixtures thereof depending on the number of nitrogen atoms suitable to form stable /V-oxides).
As used herein, the term “/V-oxide(s)” refer to ail possible, and in particular ail stable, Noxide forms of the amine compounds (e.g., compounds comprising one or more imine nitrogen atoms) described herein, such as mono-/V-oxides (including different isomers when more than one nitrogen atom of an amine compound can form a mono-/V-oxide) or multi-/V-oxides (e.g., bis-/V-oxides), or mixtures thereof in any ratio.
Compounds of Formula I and their salts described herein further include /V-oxides thereof.
Compounds of Formula I (including salts thereof) may exist in a continuum of solid states ranging from fully amorphous to fully crystalline. The term ‘amorphous’ refers to a state in which the material lacks long-range order at the molecular level and, depending upon température, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from apparent solid to a material with liquid properties occurs, which is characterised by a change of state, typically second order (‘glass transition’). The term ‘crystalline’ refers to a solid phase in which the material has a regular ordered internai structure at the molecular level and gives a distinctive Xray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order (‘melting point’).
Compounds of Formula I (including salts thereof) may exist in unsolvated and solvated forms. When the solvent or water is tightly bound, the complex will hâve a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvatés and hygroscopic compounds, the water/solvent content will be dépendent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
The compounds of Formula I (including salts thereof) may exist as clathrates or other complexes (e.g., co-crystals). Included within the scope of the invention are complexes such as clathrates, drug-host inclusion complexes wherein the drug and host are présent in stoichiometric or non-stoichiometric amounts. Also included are complexes of the compounds of Formula I containing two or more organic and/or inorganic components, which may be in stoichiometric or non-stoichiometric amounts. The resulting complexes may be ionized, partially ionized, or non-ionized. Co-crystals are typically defined as crystalline complexes of neutral molecular constituents that are bound together through non-covalent interactions, but could also be a complex of a neutral molécule with a sait. Co-crystals may be prepared by melt crystallization, by recrystallization from solvents, or by physically grinding the components together; see O. Almarsson and M. J. Zaworotko, Chem. Commun. 2004, 17, 1889-1896. For a general review of multi-component complexes, see J. K. Haleblian, J. Pharm. Soi. 1975, 64, 1269-1288.
The compounds of the invention (including salts thereof) may also exist in a mesomorphic state (mesophase or liquid crystal) when subjected to suitable conditions. The mesomorphic state is intermediate between the true crystalline state and the true liquid state (either melt or solution). Mesomorphism arising as the resuit of a change in température is described as ‘thermotropic’ and that resulting from the addition of a second component, such as water or another solvent, is described as ‘lyotropic’. Compounds that hâve the potential to form lyotropic mesophases are described as ‘amphiphilic’ and consist of molécules which possess an ionic (such as -COO~Na+, -COOK+, or -SO3'Na+) or non-ionic (such as -N'N+(CH3)3) polar head group. For more information, see Crystals and the Polarizing Microscope by N. H. Hartshorne and A. Stuart, 4,h Edition (Edward Arnold, 1970).
The invention also relates to prodrugs of the compounds of Formula I. Thus certain dérivatives of compounds of Formula I which may hâve little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of Formula I having the desired activity, for example, by hydrolytic cleavage. Such dérivatives are referred to as “prodrugs”. Further information on the use of prodrugs may be found in Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Sériés (T. Higuchi and W. Stella) and
Bioreversible Carriers in Drug Design, Pergamon Press, 1987 (Ed. E. B. Roche, American Pharmaceutical Association).
Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities présent in the compounds of Formula I with certain moieties known to those skilled in the art as ‘pro-moieties’ as described, for example, in Design of Prodrugs by H. Bundgaard (Elsevier, 1985), or in Prodrugs: Challenges and Reward, 2007 édition, edited by Valentino Stella, Ronald Borchardt, Michael Hageman, Reza Oliyai, Hans Maag, Jefferson Tilley, pages 134-175 (Springer, 2007).
Moreover, certain compounds of Formula I may themselves act as prodrugs of other compounds of Formula I.
Also included within the scope of the invention are métabolites of compounds of Formula I, that is, compounds formed in vivo upon administration of the drug.
The compounds of Formula I (including salts thereof) include ail stereoisomers and tautomers. Stereoisomers of Formula I include cis and trans isomers, optical isomers such as R and S enantiomers, diastereomers, géométrie isomers, rotational isomers, atropisomers, and conformational isomers of the compounds of Formula I, including compounds exhibiting more than one type of isomerism; and mixtures thereof (such as racemates and diastereomeric pairs). Also included are acid addition or base addition salts wherein the counterion is opticaliy active, for example, D-lactate or L-lysine, or racemic, for example, DL-tartrate or DL-arginine.
In some embodiments, the compounds of Formula I (including salts thereof) may hâve asymmetric carbon atoms. The carbon-carbon bonds of the compounds of Formula I may be depicted herein using a solid line (------), a solid wedge ( —), or a dotted wedge (.......11111
). The use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that ail possible stereoisomers (e.g., spécifie enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of either a solid or dotted wedge to depict bonds to asymmetric carbon atoms is meant to indicate that only the stereoisomer shown is meant to be included. It is possible that compounds of Formula I may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that ail possible stereoisomers are meant to be included. For example, unless stated otherwise, it is intended that the compounds of Formula I can exist as enantiomers and diastereomers or as racemates and mixtures thereof. The use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of Formula I and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is présent.
In some embodiments, the compounds of Formula I (including salts thereof) may exist in and/or be isolated as atropisomers (e.g., one or more atropenantiomers). Those skilled in the art would recognize that atropisomerism may exist in a compound that has two or more aromatic rings (for example, two aromatic rings linked through a single bond). See e.g.,
Freedman, T. B. et al., Absolute Configuration Détermination of Chiral Molécules in the Solution State Using Vibrational Circular Dichroism. Chirality 2003, 15, 743-758; and Bringmann, G. et al., Atroposelective Synthesis of Axially Chiral Biaryl Compounds. Angew. Chem., Int. Ed. 2005, 44, 5384-5427.
When any racemate crystallizes, crystals of different types are possible. One type is the racemic compound (true racemate) wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. Another type is a racemic mixture or conglomerate wherein two forms of crystal are produced in equal or different molar amounts each comprising a single enantiomer.
The compounds of Formula I (including salts thereof) may exhibit the phenomena of tautomerism and structural isomerism. For example, the compounds of Formula I may exist in several tautomeric forms, including the enol and imine form, the amide and imidic acid form, and the keto and enamine form and géométrie isomers and mixtures thereof. Ail such tautomeric forms are included within the scope of the compounds of Formula I. Tautomers may exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer prédominâtes.
Even though one tautomer may be described, the présent invention includes ail tautomers of the compounds of Formula I. For example, when one of the following two tautomers of the invention is disclosed in the experimental section herein, those skilled in the art would readily recognize that the invention also includes the other.
For another example, when one of the following three tautomers of the invention is disclosed in the experimental section herein, those skilled in the art would readily recognize that the invention also includes each of the others.
O OH O
The présent invention includes ail pharmaceutically acceptable isotopically-labelled compounds of Formula I (including salts thereof) wherein one or more atoms are replaced by
atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which prédominâtes in nature.
Examples of isotopes suitable for inclusion in the compounds of the invention (including salts thereof) include isotopes of hydrogen, such as 2H and 3H, carbon, such as 11C, 13C and 5 14C, chlorine, such as 36CI, fluorine, such as 18F, iodine, such as 123l and 125l, nitrogen, such as 13N and 15N, oxygen, such as 150,17O and 18O, phosphorus, such as 32P, and sulphur, such as 35S.
Certain isotopically-labelled compounds of Formula I, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e., 3H, and carbon-14, i.e., 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of détection.
Substitution with heavier isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. 15 Substitution with positron-emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
Isotopically-labeled compounds of Formula I (including salts thereof) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Préparations using an 20 appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein L1 is O.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein L1 is S.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein L1 is NH, alkyl), 14(-0^ alkyl-C3.4 cycloalkyl), or N(C3.6 cycloalkyl). In a further embodiment, L1 is NH.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein Q1 is Q1a. In a further embodiment, X1 is O.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I is a compound of Formula IA-1, IA-2, IA-3, IA-4, IA-5, IA-6, IA-7, IA-8, IA-9, or IA-10:
IA-9
IA-10.
In one embodiment of a compound of Formula I or a pharmaceutically acceptable sait thereof wherein Q1 is Q1a, or in one embodiment of a compound of Formula IA-1, IA-2, IA-3, ΙΑ4, IA-5, IA-6, IA-7, IA-8, IA-9, or IA-10, or a pharmaceutically acceptable sait thereof, each R9 is independently selected from the group consisting of -CN, C^ alkyl, Ci_4 haloalkyl, C3.4 cycioalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the C14 alkyl of R9 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, CM alkoxy, and C^ haloalkoxy; and wherein each of the C3.4 cycioalkyl, cyclopropylmethyl, and cyclobutylmethyl of R9 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN,
C-j-4 alkyl, CM haloalkyl, C-m hydroxylalkyl, cyanoalkyl, alkoxy, and C14 haloalkoxy; and
R9A is selected from the group consisting of H, C1.3 alkyl, C^ hydroxylalkyl, allyl, S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -C(R14)2-OH, -C(R14)2-OS(=O)2H, C(R14)2-OP(=O)(OH)2, -C(R14)2-OR15, and -C(R14)2-OC(=O)-R15. In a further embodiment, each R9 is independently selected from the group consisting of C^ alkyl, C^ haloalkyl, and cyclopropyl. In a yet further embodiment, each R9 is independently selected from the group consisting of C^ alkyl and cyclopropyl. In a still further embodiment, each R9 is independently methyl or ethyl. In a yet still further embodiment, each R9 is methyl.
In one embodiment of a compound of Formula IA-1 or IA-6, or a pharmaceutically acceptable sait thereof, R9A is selected from the group consisting of H, C^ alkyl, C1.3 hydroxylalkyl, and allyl. In a further embodiment, R9A is not H.
In one embodiment of a compound of Formula IA-1 or IA-6, or a pharmaceutically acceptable sait thereof, R9A is selected from the group consisting of H, -S(=O)2N(R5)(R6), C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -C(R14)2-OH, -C(R14)2-OS(=O)2H, -C(R14)2OP(=O)(OH)2j -C(R14)2-OR15, and -C(R14)2-OC(=O)-R15. In a further embodiment, R9A is selected from the group consisting of H, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, C(=O)-OR8, -CH2-OH, -CH2-OS(=O)2H, -CH2-OP(=O)(OH)2i -CH2-OR15, and -CH2-OC(=O)-R15. In a yet further embodiment, R9A is not H.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein Q1 is Q1b. In a further embodiment, X1 is O.
In another further embodiment, X2 is O. In a yet further embodiment, each of X1 and X2 is O.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I is a compound of Formula IB-1, IB-2, IB-3, IB-4, or IB-5, IB-6, IB-7, IB-8, IB-9, or IB-10:
IB-9
IB-10.
In one embodiment of a compound of Formula I or a pharmaceutically acceptable sait thereof of the présent invention wherein Q1 is Q1b, or in one embodiment of a compound of
Formula IB-1, IB-2, IB-3, IB-4, IB-5, IB-6, IB-7, IB-8, IB-9, or IB-10, or a pharmaceutically acceptable sait thereof, of the présent invention:
R10 is selected from the group consisting of -CN, Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the alkyl of R10 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R10 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and CV4 haloalkoxy;
R10A is selected from the group consisting of H, Cm alkyl, Cm hydroxylalkyl, C2.4 alkenyl, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -C(R14)2-OH, -C(R14)2-OS(=O)2H, C(R14)2-OP(=O)(OH)2, -C(R14)2-OR15, and -C(R14)2-OC(=O)-R15; and
R1ob is selected from the group consisting of Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R10B is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R10B is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, CV4 cyanoalkyl, Cm alkoxy, and Cm haloalkoxy.
In a further embodiment of the above compound of Formula I or a pharmaceutically acceptable sait thereof wherein Q1 is Q1b, or in a further embodiment of the above compound of Formula IB-1, IB-2, IB-3, IB-4, IB-5, IB-6, IB-7, ΙΒ-8, IB-9, or IB-10, or a pharmaceutically acceptable sait thereof, each of R10 and R10B is independently selected from the group consisting of Cmalkyl, Cmhaloalkyl, and cyclopropyl. In a yet further embodiment, each of R10 and R10B is independently selected from the group consisting of Cmalkyl and cyclopropyl. In a still further embodiment, each of R10 and R10B is independently methyl or ethyl. In a yet still further embodiment, each of R10 and R10B is methyl.
In one embodiment of a compound of Formula IB-1 or IB-6, or a pharmaceutically acceptable sait thereof, R10A is selected from the group consisting of H, Cm alkyl, Cm hydroxylalkyl, and C2.4 alkenyl (e.g., allyl). In a further embodiment, R10A is not H.
In one embodiment of a compound of Formula IB-1 or IB-6, or a pharmaceutically acceptable sait thereof, R10A is selected from the group consisting of H, -S(=O)2N(R5)(R6), C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -C(R14)2-OH, -C(R14)2-OS(=O)2H, -C(R14)2OP(=O)(OH)2j -C(R14)2-OR15, and -C(R14)2-OC(=O)-R15. In a further embodiment, R10A is selected from the group consisting of H, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, 17657
C(=O)-OR8, -CH2-OH, -CH2-OS(=O)2H, -CH2-OP(=O)(OH)2j -CH2-OR15, and -CH2-OC(=O)-R15.
In a yet further embodiment, R10A is not H.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein Q1 is Q1c. In a further embodiment, X1 is O.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I is a compound of
Formula IC-1, IC-2, IC-3, IC-4, IC-5, or IC-6:
R1 R3 (R11)t3
IC-1
R1 R3
IC-3
IC-5
In one embodiment of a compound of Formula I or a pharmaceutically acceptable sait thereof of the présent invention wherein Q1 is Q1c, or in one embodiment of a compound of Formula IC-1, IC-2, IC-3, IC-4, IC-5, or IC-6, or a pharmaceutically acceptable sait thereof, of the présent invention:
each R11 is independently selected from the group consisting of -CN, C^ alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the CM alkyl of R11 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Ci-4 alkoxy, and Ci_4 haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R11 is optionally substituted with one or
more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, CM cyanoalkyl, Cm alkoxy, and Cm haloalkoxy; and
R11A is selected from the group consisting of Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R11A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R11A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and C^.4 haloalkoxy.
In a further embodiment of the above compound of Formula I or a pharmaceutically acceptable sait thereof wherein Q1 is Q1c, or in a further embodiment of the above compound of Formula IC-1, IC-2, IC-3, IC-4, IC-5, or IC-6, or a pharmaceutically acceptable sait thereof, each of R11 and R11A is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl. In a yet further embodiment, each of R11 and R11A is independently selected from the group consisting of Cmalkyl and cyclopropyl. In a still further embodiment, each of R11 and R11A is independently methyl or ethyl. In a yet still further embodiment, each of R11 and R11A is methyl.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein Q1 is Q1d. In a further embodiment, X1 is O.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I is a compound of
Formula ID-1, ID-2, ID-3, ID-4, ID-5, ID-6, ID-7, ID-8, ID-9, or ID-10:
ID-3
ID-4
ID-9
ID-10.
In one embodiment of a compound of Formula I or a pharmaceutically acceptable sait thereof wherein Q1 is Q1d, or in one embodiment of a compound of Formula ID-1, ID-2, ID-3, ID4, ID-5, ID-6, ID-7, ID-8, ID-9, or ID-10, or a pharmaceutically acceptable sait thereof, each R12 10 is independently selected from the group consisting of -CN, alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Ci.4 alkyl of R12 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Ci_4 alkoxy, and haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R12 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, C^ alkyl, haloalkyl, hydroxylalkyl, cyanoalkyl, alkoxy, and Ci.4 haloalkoxy; and R12A is selected from the group consisting of C-m alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the alkyl of R12A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, alkoxy, and Ον4 haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R12A is optionally substituted with one or more substituents each
independently selected from the group consisting of halogen, -OH, -CN, Ον4 alkyl, Ci_4 haloalkyl, Cv4 hydroxylalkyl, cyanoalkyl, C14 alkoxy, and CM haloalkoxy. In a further embodiment, each of R12 and R12A is independently selected from the group consisting of 0^3 alkyl, haloalkyl, and cyclopropyl. In a yet further embodiment, each of R12 and R12A is independently selected from the group consisting of Co alkyl and cyclopropyl. In a sill further embodiment, each of R12 and R12A is independently methyl or ethyl. In a yet still further embodiment, each of R12 and R12A is methyl.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein Q1 is Q1e. In a further embodiment, X1 is O.
An embodiment of the présent invention is a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I is a compound of Formula IE-1, IE-2, IE-3, IE-4, or IE-5, IE-6, IE-7, IE-8, IE-9, or IE-10:
IE-5
IE-6
IE-9
IE-10.
In one embodiment of a compound of Formula I or a pharmaceutically acceptable sait thereof wherein Q1 is Q1e, or in one embodiment of a compound of Formula IE-1, IE-2, IE-3, IE4, IE-5, IE-6, IE-7, IE-8, IE-9, or IE-10, or a pharmaceutically acceptable sait thereof, each R13 is independently selected from the group consisting of -CN, Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R13 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R13 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy; and R13A is selected from the group consisting of Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R13A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R13A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, alkyl, Cm haloalkyl, Ci-4 hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy. In a further embodiment, each of R13 and R13A is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl. In a yet further embodiment, each of R13 and R13A is independently selected from the group consisting of Cm alkyl and cyclopropyl. In a still further embodiment, each of R13 and R13A is independently methyl or ethyl. In a yet still further embodiment, each of R13 and R13A is methyl.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10, i.e.., a compound of
ID-8, ID-9, ID-10, IE-1, IE-2, IE-3, IE-4, IE-5, IE-6, IE-7, IE-8, IE-9, or IE-10), or a pharmaceutically acceptable sait thereof, each of R1 and R2 is independently selected from the group consisting of H, halogen, -CN, Cm alkyl, Cm haloalkyl, Cm alkoxy, Cm haloalkoxy, C3.6 cycloalkyl, -0(=0)-(0,.4 alkyl), -C(=O)OH, and 0(=0)-0-(0,.4 alkyl), wherein each of the 0,-6 alkyl and C3.6 cycloalkyl is optionally substituted with 1, 2, 3, 4, or 5 substituents each independently selected from halogen, -OH, -CN, C,.4 alkyl, 0,.4 haloalkyl, Cm alkoxy, and 0,-4 haloalkoxy. In a further embodiment, each of R1 and R2 is independently selected from the group consisting of H, halogen, -0,.4 alkyl, 0,.4 alkoxy, and 03.4 cycloalkyl, wherein each of the 0,-4 alkyl and Ci.4 alkoxy of R1 and R2 is optionally substituted with 1,2, 3, 4, or 5 substituents each independently selected from halogen, -OH, Cm alkoxy, and Cm haloalkoxy; and wherein the C3.4 cycloalkyl of R1 and R2 is optionally substituted with 1,2, 3, 4, or 5 substituents each independently selected from halogen, -OH, Cm alkyl, Cm haloalkyl, C,.4 alkoxy, and C,.4 haloalkoxy. In a yet further embodiment, each of R1 and R2 is independently H, methyl, or halogen (e.g., F). In a still further embodiment, each of R1 and R2 is independently H or halogen (e.g., F). In a yet further embodiment, each of R1 and R2 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, each of R3 and R4 is independently selected from the group consisting of H, halogen, -CN, - C,.4 alkyl, C,.4 alkoxy, and C3.4 cycloalkyl, wherein each of the C,.4 alkyl and Cm alkoxy of R3 and R4 is optionally substituted with 1,2, 3, 4, or 5 substituents each independently selected from halogen, -OH, Cm alkoxy, and Cm haloalkoxy; and wherein the C3. 4 cycloalkyl of R3 and R4 is optionally substituted with 1,2, 3, 4, or 5 substituents each independently selected from halogen, -OH, Cm alkyl, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy. In a further embodiment, each of R3 and R4 is independently H, F, Cl, CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cmalkoxy. In a yet further embodiment, R3is H; and R4 is H, halogen, or methyl, wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cmalkoxy. In a still further embodiment, R3 is H and R4 is methyl.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas
IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, each of R1 and R2 is independently H, methyl, or halogen (e.g., F or Cl);
and each of R3 and R4 is independently H, halogen (e.g., F or Cl), CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cmalkoxy. In a further embodiment, each of R1, R2, and
R3 is H, and R4 is H, halogen, or methyl. In a yet further embodiment, R4 is H or methyl. In a still further embodiment, R4 is methyl.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, each of R1 and R3 is independently H, halogen, -CN, methyl, or methoxy, wherein each of the methyl and methoxy is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cm alkoxy; and R2 and R4 together with the two carbon atoms to which they are attached form a fused 5- or 6-membered heteroaryl, a fused 5- or 6-membered heterocycloalkyl ring, a fused 5or 6-membered cycloalkyl ring, or a fused benzene ring, wherein each of the fused rings is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halo, -CN, -OH, Cm alkyl, Cm alkoxy, Cm haloalkyl, and Cm haloalkoxy, and wherein the fused heterocycloalkyl ring or fused cycloalkyl ring is further optionally substituted with 1,2, or 3 oxo. In a further embodiment, each of R1 and R3 is independently H, halogen, CN, methyl, Cm fluoroakyl, methoxy, or Ci fluoroalkoxy. In a yet further embodiment, R2 and R4 together with the two carbon atoms to which they are attached form an optionally substituted fused 5- or 6-membered heteroaryl. In a still further embodiment, each of R1 and R3 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, each of T1, T2, T3, and T4 is independently selected from the group consisting of H, halogen, -CN, Cm alkyl, Cm haloalkyl, C2.4 alkenyl, Cm alkoxy, Cm haloalkoxy, and C3.4 cycloalkyl, wherein each of the Cm alkyl, C2_4 alkenyl, and Cm alkoxy of T1, T2, T3, and T4 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein the C3.4 cycloalkyl of T1, T2, T3, and T4 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, CM cyanoalkyl, Cm alkoxy, and Cm haloalkoxy. In a further embodiment, each of T1, T2, T3, andT4 is independently selected from the group consisting of H, halogen, CV4 alkyl, Cm hydroxylalkyl, Cm haloalkyl, Cm alkoxy, CM haloalkoxy, C3.4 cycloalkyl, and C3.4 halocycloalkyl. In a further embodiment, at least one of T1, T2, T3, and T4 is other than H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is other than H. In a further embodiment, T1 is selected from the group consisting of halogen, C3.4 cycloalkyl, Cm alkyl, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy, wherein each of the C3.4 cycloalkyl and C}.4 alkyl is optionally substituted with one or more substituents each independently selected from the group consisting of halogen and -OH. In a yet further embodiment, T1 is selected from the group consisting of halogen, C3.4 cycloalkyl, C3.4 halocycloalkyl, CM alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm alkoxy, and Cm haloalkoxy.
φ In a still further embodiment, T1 is selected from the group consisting of halogen, cyclopropyl, halocyclopropyl, methyl, ethyl, haloalkyl, C^ hydroxylalkyl, methoxy, ethoxy, and haloalkoxy. In a further embodiment, T1 is selected from the group consisting of halogen, cyclopropyl, halocyclopropyl, methyl, C/ haloalkyl, methoxy, and haloalkoxy. In a further embodiment, T1 is selected from the group consisting of C3.4 cycloalkyl, C3.4 halocycloalkyl, CV4 alkyl, and C14 haloalkyl.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is selected from the group consisting of C14 alkyl and Ον4 haloalkyl.
In a further embodiment, T1 is selected from the group consisting of methyl, ethyl, and haloalkyl. In a further embodiment, T1 is C^ haloalkyl (e.g., C^ fluoroalkyl).
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is selected from the group consisting of Ci_4 alkoxy, and Cf4 haloalkoxy. In a further embodiment, T1 is selected from the group consisting of methoxy, ethoxy, and Ομ2 haloalkoxy. In a further embodiment, T1 is C^ haloalkoxy (e.g., Ci_2 fluoroalkoxy).
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is halogen.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is selected from the group consisting of C3.4 cycloalkyl and C3.4 halocycloalkyl. In a further embodiment, T1 is C3.4 cycloalkyl.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas
IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T2 is selected from the group consisting of H, halogen, -CN, C3.4 cycloalkyl, C3.4 halocycloalkyl, Cw alkyl, Ci_4 haloalkyl, Cw alkoxy, and ΰν4 haloalkoxy, wherein each of the C3.4 cycloalkyl and Cv4 alkyl is optionally substituted with one or more substituents each independently selected from the group consisting of halogen and -OH. In a further embodiment, T2 is selected from the group consisting of H, halogen, -CN, C3.4 cycloalkyl, C3.4 halocycloalkyl, CV4 alkyl, CV4 haloalkyl, Cj_4 hydroxylalkyl, Ον4 alkoxy, and C^ haloalkoxy. In a yet further embodiment, T2 is selected from the group consisting of H, halogen, methyl, ethyl, Ct-2 haloalkyl, Ci_2 hydroxylalkyl, C1-2 alkoxy, and Ci_2 haloalkoxy. In a still further embodiment,
T2 is selected from the group consisting of H, halogen, methyl, -CH2OH, and Ci haloalkyl. In a yet still further embodiment, T2 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T3 is selected from the group consisting of H, halogen, -CN, C3.4 cycloalkyl, C3.4 halocycloalkyl, C^ alkyl, CM haloalkyl, CM hydroxylalkyl, alkoxy, and Ci_4 haloalkoxy. In a further embodiment, T3 is selected from the group consisting of H, halogen, methyl, -CH2OH, and haloalkyl (e.g., Ci fluoroalkyl). In a yet further embodiment, T3 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T4 is H, halogen, methyl, -CH2OH, or Ci haloalkyl. In a further embodiment, T4 is H or F. In a yet further embodiment, T4 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, each of T1, T2, and T3 is independently selected from the group consisting of H, halogen, Ci_2 alkyl, C^ haloalkyl, CV2 hydroxylalkyl, alkoxy, C^ haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In a further embodiment, each of T1, T2, and T3 is independently selected from the group consisting of H, halogen, methyl, Ci haloalkyl, -CH2OH, cyclopropyl, methoxy, and Ci haloalkoxy; and T4 is H. In a yet further embodiment, each of T1, T2, and T3 is independently selected from the group consisting of H, halogen (F, Cl, Br, or I), methyl, Ci fluoroalkyl (e.g., CF3 or CHF2), -CH2OH, cyclopropyl, methoxy, and Ci fluoroalkoxy (e.g., -OCF3 or -OCHF2); and T4 is H. In a still further embodiment, T1 is other than H and at least one of T2 and T3 is H. In a further embodiment, each of T2 and T3 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is selected from the group consisting of halogen (F, Cl, Br, or I), methyl, -CH2OH, Ci fluoroalkyl (e.g., CF3 or CHF2), methoxy, Ci fluoroalkoxy (e.g., -OCF3 or OCHF2), cyclopropyl, and fluorocyclopropyl; and each of T2, T3, and T4 is H.
In one embodiment of a compound of Formula I (e.g., a compound of one of Formulas IA-1 to IA-10, IB-1 to IB-10, IC-1 to IC-6, ID-1 to ID-10, and IE-1 to IE-10), or a pharmaceutically acceptable sait thereof, T1 is selected from the group consisting of halogen, Ci_2 alkyl, Ci.2 haloalkyl, Ci_2 hydroxylalkyl, Ci_2 alkoxy, Ci_2 haloalkoxy, cyclopropyl, and halocyclopropyl; each of T2 and T3 is independently selected from the group consisting of H, halogen, C^ alkyl, Ci_2 haloalkyl, Ci_2 hydroxylalkyl, Ci.2 alkoxy, Ci_2 haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In one further embodiment, one of T2 and T3 is H and the other is not H. In another further embodiment, T2 is H and T3 is not H. In yet another further embodiment, T2 is not H and T3 is H.
In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-1 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-2 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-3 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-4 or a sait thereof. In one
embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-5 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of
Formula IA-6 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-7 or a sait thereof. In one embodiment, the compound of Formula
I or a sait thereof is a compound of Formula IA-8 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-9 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IA-10 or a sait thereof.
In one embodiment, the compound of Formula I or a sait thereof is a compound of
Formula IB-1 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-2 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-3 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-4 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-5 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-6 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-7 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-8 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-9 or a sait thereof. In one 20 embodiment, the compound of Formula I or a sait thereof is a compound of Formula IB-10 or a sait thereof...
In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IC-1 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IC-2 or a sait thereof. In one embodiment, the compound of Formula 25 I or a sait thereof is a compound of Formula IC-3 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IC-4 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IC-5 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IC-6 or a sait thereof.
In one embodiment, the compound of Formula I or a sait thereof is a compound of
Formula ID-1 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-2 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-3 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-4 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-5 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-6 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-7 or a sait thereof. In one embodiment, the compound of Formula
I or a sait thereof is a compound of Formula ID-8 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-9 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula ID-10 or a sait thereof.
In one embodiment, the compound of Formula I or a sait thereof is a compound of
Formula IE-1 or a sait thereof. In one embodiment, the compound of Formula l or a sait thereof is a compound of Formula IE-2 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-3 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-4 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-5 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-6 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-7 or a sait thereof. In one embodiment, the compound of Formula or a sait thereof is a compound of Formula IE-8 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-9 or a sait thereof. In one embodiment, the compound of Formula I or a sait thereof is a compound of Formula IE-10 or a sait thereof.
In one embodiment of a compound of Formula IA-1, IA-2, IA-3, IA-4, ΙΑ-5, IA-6, ΙΑ-7, IA8, IA-9, or IA-10, or a pharmaceutically acceptable sait thereof, each R9 is independently selected from the group consisting of Ο1<3 alkyl, CA haloalkyl, and cyclopropyl; each of R1 and R2 is independently H, methyl, or halogen (e.g., F); each of R3 and R4 is independently H, F, Cl, CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cualkoxy; T1 is selected from the group consisting of halogen, Ci-2 alkyl, CV2 haloalkyl, CA hydroxylalkyl, CA alkoxy, CA haloalkoxy, cyclopropyl, and halocyclopropyl; each of T2 and T3 is independently selected from the group consisting of H, halogen, CA alkyl, CA haloalkyl, C^ hydroxylalkyl, C^ alkoxy, C^ haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In one further embodiment, each R9 is independently selected from the group consisting of CA alkyl and cyclopropyl; each of R1 and R2 is H; R3is H; and R4 is methyl. In one yet further embodiment, the compound or a sait thereof is a compound of Formula IA-1 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-2 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-3 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ΙΑ-4 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-5 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-6 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-7 or a sait thereof. In another yet further embodiment, the compound or a sait φ thereof is a compound of Formula IA-8 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-9 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IA-10 or a sait thereof.
In one embodiment of a compound of Formula IB-1,1B-2,1B-3,1B-4, IB-5, IB-6, IB-7, IB8, IB-9, or IB-10, or a pharmaceutically acceptable sait thereof, each of R10 and R10B is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl; each of R1 and R2 is independently H, methyl, or halogen (e.g., F); each of R3and R4 is independently H, F, Cl, CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cm alkoxy; T1 is selected from the group consisting of halogen, C}.2 alkyl, Cm haloalkyl, CV2 hydroxylalkyl, C;.2 alkoxy, Ci_2 haloalkoxy, cyclopropyl, and halocyclopropyl; each of T2 and T3 is independently selected from the group consisting of H, halogen, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, CV2 alkoxy, C^2 haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In one further embodiment, each of R10 and R108 is independently selected from the group consisting of C-|.3alkyl and cyclopropyl; each of R1 and R2 is H; R3 is H; and R4 is methyl. In one yet further embodiment, the compound or a sait thereof is a compound of Formula IB-1 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-2 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-3 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-4 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-5 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-6 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ΙΒ-7 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-8 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB-9 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IB10 or a sait thereof.
In one embodiment of a compound of Formula IC-1, IC-2, IC-3, IC-4, IC-5 or IC-6, or a pharmaceutically acceptable sait thereof, each of R11 and R11A is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl; each of R1 and R2 is independently H, methyl, or halogen (e.g., F); each of R3and R4 is independently H, F, Cl, CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cm alkoxy; T1 is selected from the group consisting of halogen, Ci_2 alkyl, Cm haloalkyl, C^.2 hydroxylalkyl, Ci_2 alkoxy, Cm haloalkoxy, cyclopropyl, and halocyclopropyl; each of T2 and T3 is independently selected from the group consisting of H, halogen, Ci.2 alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cv 2 alkoxy, Cî-2 haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In one further embodiment, each of R11 and R11A is independently selected from the group consisting of CV3 alkyl and cyclopropyl; each of R and R2 is H; R3 is H; and R4 is methyl. In one yet further embodiment, the compound or a sait thereof is a compound of Formula IC-1 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IC-2 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IC-3 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IC-4 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IC-5 or a sait thereof.
In one embodiment of a compound of Formula ID-1, ID-2, ID-3, ID-4, ID-5, ID-6, ID-7, ID8, ID-9, or ID-10, or a pharmaceutically acceptable sait thereof, each of R12 and R12A is independently selected from the group consisting of alkyl, Ci_3 haloalkyl, and cyclopropyl; each of R1 and R2 is independently H, methyl, or halogen (e.g., F); each of R3and R4 is independently H, F, Cl, CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Ci-4 alkoxy; T1 is selected from the group consisting of halogen, C/.2 alkyl, C^ haloalkyl, Ci_2 hydroxylalkyl, C^ alkoxy, 0^2 haloalkoxy, cyclopropyl, and halocyclopropyl; each of T2 and T3 is independently selected from the group consisting of H, halogen, 0^2 alkyl, haloalkyl, CV2 hydroxylalkyl, CK2 alkoxy, C).2 haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In one further embodiment, each of R12 and R12A is independently selected from the group consisting of C^salkyl and cyclopropyl; each of R1 and R2 is H; R3 is H; and R4 is methyl. In one yet further embodiment, the compound or a sait thereof is a compound of Formula ID-1 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-2 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-3 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-4 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-5 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-6 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-7 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-8 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID-9 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula ID10 or a sait thereof.
In one embodiment of a compound of Formula IE-1, IE-2, IE-3, IE-4, IE-5, IE-6, IE-7, IE8, IE-9, or IE-10, or a pharmaceutically acceptable sait thereof, each of R13 and R13A is independently selected from the group consisting of Ci_3 alkyl, haloalkyl, and cyclopropyl; each of R1 and R2 is independently H, methyl, or halogen (e.g., F); each of R3and R4 is independently H, F, Cl, CN, or methyl wherein the methyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and CM alkoxy; T1 is selected from the group consisting of halogen, Ον2 alkyl, Ci_2 haloalkyl, hydroxylalkyl, Ci_2 alkoxy, C^ haloalkoxy, cyclopropyl, and halocyclopropyl; each of T2 and T3 is independently selected from the group consisting of H, halogen, C^ alkyl, C^ haloalkyl, CV2 hydroxylalkyl, C>2 alkoxy, haloalkoxy, cyclopropyl, and halocyclopropyl; and T4 is H. In one further embodiment, each of R13 and R13A is independently selected from the group consisting of Ci.3 alkyl and cyclopropyl; each of R1 and R2 is H; R3 is H; and R4 is methyl. In one yet further embodiment, the compound or a sait thereof is a compound of Formula IE-1 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-2 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-3 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-4 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-5 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-6 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-7 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-8 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE-9 or a sait thereof. In another yet further embodiment, the compound or a sait thereof is a compound of Formula IE10 or a sait thereof.
In one embodiment, the invention also provides one or more of the compounds described in Examples 1-81 in the Examples section of the subject application, and pharmaceutically acceptable salts of the compounds or the /V-oxides.
One embodiment of the prevent invention provides a compound selected from: (-)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1/7,3/7)-dione;
(-)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1/7,3/7)-dione;
6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-5-ethyl-1 -methylpyrimidine-2,4(1/7,3H)dione;
(-)-1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2-yl]oxy}phenyl)pyrimidine2,4(1/7,3/7)-dione;
(-)-6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1 /7,3/7)-dione;
6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]phenyl}-1,5-dimethylpyrimidine-2,4(1/7,3H)dione;
(-)-6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy)-2-methylphenyl)-1,5-dimethylpyrimidine2,4(1/7,3/7)-dione;
(+)-5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6-dimethylpyridazin3(2H)-one;
6-{4-[(3-chloropyridin-2-yl)sulfanyl]-2-methylphenyl}-1,5-dimethylpyrimidine-2,4(1 /7,3/7)dione;
5-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-4,6-dimethylpyridazin-3(2/7)one;
5-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl)-4,6-dimethylpyridazin-3(2/7)-one;
5-{4-[(3-iodopyridin-2-yl)oxy]-2-methylphenyl}-4,6-dimethylpyridazin-3(2/7)-one;
(-)-6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine-2,4(1 /7,3/7)dione;
5-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl)-4,6-dimethylpyridazin-3(2/7)-one; 5-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-4,6-dimethylpyridazin-3(2/7)15 one;
5- (4-{[4-methoxy-3-(trifluoromethyl)pyridin-2-yl]oxy)-2-methylphenyl)-4,6dimethylpyridazin-3(2/7)-one;
(+)-4,6-dimethyl-5-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2-yl]oxy)phenyl)pyridazin3(2/7)-one;
6-{4-[(3-cyclopropylpyridin-2-yl)oxy]phenyl}-1,5-dimethylpyrimidine-2,4(1 H,3/7)-dione;
6- {4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl)-1,5-dimethylpyrazin-2(1H)-one;
6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidin-2(1/7)one; and
1-cyclopropyl-6-(4-((3-(difluoromethyl)pyridin-2-yl)oxy)-2-methylphenyl)-525 methylpyrimidine-2,4(1 /7,3/7)-dione, or a pharmaceutically acceptable sait thereof.
The présent invention also provides compositions (e.g., pharmaceutical compositions) comprising a compound of Formula I (including a pharmaceutically acceptable sait thereof). Accordingly, in one embodiment, the invention provides a pharmaceutical composition comprising (a therapeutically effective amount of) a compound of Formula I (or a pharmaceutically acceptable sait thereof) and optionally comprising a pharmaceutically acceptable carrier. In one further embodiment, the invention provides a pharmaceutical composition comprising (a therapeutically effective amount of) a compound of Formula I (or a pharmaceutically acceptable sait thereof), optionally comprising a pharmaceutically acceptable carrier and, optionally, at least one additional médicinal or pharmaceutical agent (such as an antipsychotic agent or anti-schizophrenia agent described below). In one embodiment, the additional médicinal or pharmaceutical agent is an anti-schizophrenia agent as described below.
The pharmaceutically acceptable carrier may comprise any conventional pharmaceutical carrier or excipient. Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents (such as hydrates and solvatés). The pharmaceutical compositions may, if desired, contain additional ingrédients such as flavorings, binders, excipients and the like. Thus for oral administration, tablets containing various excipients, such as citric acid, may be employed together with various disintegrants such as starch, alginic acid and certain complex silicates and with binding agents such as sucrose, gelatin and acacia. Additionally, lubricating agents such as magnésium stéarate, sodium lauryl sulfate and talc are often useful for tableting purposes. Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules. Non-limiting examples of materials, therefore, include lactose or milk sugar and high molecular weight polyethylene glycols. When aqueous suspensions or élixirs are desired for oral administration, the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, éthanol, propylene glycol, glycerin, or combinations thereof.
The pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulation, solution or suspension, for parentéral injection as a stérile solution, suspension or émulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
Exemplary parentéral administration forms include solutions or suspensions of active compounds in stérile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms may be suitably buffered, if desired.
The pharmaceutical composition may be in unit dosage forms suitable for single administration of précisé dosages. One of ordinary skill in the art would appreciate that the composition may be formulated in sub-therapeutic dosage such that multiple doses are envisioned.
In one embodiment the composition comprises a therapeutically effective amount of a compound of Formula I (or a pharmaceutically acceptable sait thereof) and a pharmaceutically acceptable carrier.
Compounds of Formula I (including pharmaceutically acceptable salts thereof) are D1 modulators. In some embodiments, a compound of Formula I is a D1 agonist [i.e., binding (having affinity for) and activating D1 receptorsj. In some embodiments, using dopamine as a reference full D1 agonist, a compound of Formula I is a superagonist (i.e., a compound that is capable of producing a greater maximal response than the endogenous D1 agonist, dopamine, for a D1 receptor, and thus exhibiting an efficacy of more than about 100%, for example 120%). In some embodiments, using dopamine as a reference full agonist, a compound of Formula I is a full D1 agonist (i.e., having an efficacy of about 100%, for example, 90%-100%, compared to that of dopamine). In some embodiments, using dopamine as a reference full D1 agonist, a compound of Formula I is a partial agonist [i.e., a compound having only partial efficacy (i.e., less than 100%, for example 10%-80% or 50%-70%) at a D1 receptor relative to the full agonist, dopamine, although it bmds and activâtes a D1 receptor], A D1 agonist (including superagonist, full agonist, and partial agonist) can agonize or partially agonize an activity of D1. In some embodiments, the EC50 of a compound of Formula I with respect to D1 is less than about 10 μΜ, 5 μΜ, 2 μΜ, 1 μΜ, 500 nM, 200 nM, 100 nM, 50, 40, 30, 20, 10, 5, 2, or 1 nM.
The présent invention further provides a method for modulating (such as agonizing or partially agonizing) an activity of D1 receptor (either in vitro or in vivo), comprising contacting (including incubating) the D1 receptor with a compound of Formula I (such as one selected from Examples 1-81), or a pharmaceutically acceptable sait thereof.
Another embodiment of the invention includes a method for treating a D1-mediated (or D1-associated) disorder, comprising administering to a mammal (e.g., a human) in need thereof an amount of a compound of Formula I (including a pharmaceutically acceptable sait thereof) effective in modulating (e.g., agonizing or partially agonizing) D1.
The compounds of Formula I used for treatment of a D1-mediated disorder also include pharmaceutically acceptable salts of the compounds.
D1-mediated (or D1-associated) disorders include neurological disorders [such as Tourette’s syndrome; tardive dyskinesia; Parkinson’s disease (including e.g., cognitive impairment associated with PD); cognitive disorders [including amnesia, age-related cognitive décliné, dementia [e.g., senile dementia, Alzheimer’s-associated dementia, HIV-associated dementia, Huntington’s-associated dementia, Lewy body dementia, vascular dementia, frontotemporal dementia, drug-related dementia (for example, dementia associated with pharmacotherapy therapy such as D2 antagonist therapy)], delirium, and cognitive impairment (e.g., cognitive impairment associated with AD or cognitive impairment associated with PD,), and mild cognitive impairment}; Huntington's chorea/ disease; and restless leg syndrome (RLS)]; psychiatrie disorders [such as cognitive impairment (e.g., cognitive impairment associated with schizophrenia or cognitive impairment associated with pharmacotherapy therapy (e.g., D2 antagonist therapy)); anxiety (including acute stress disorder, generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder, and obsessive-compulsive disorder); factitious disorder (including acute hallucinatory mania); impulse control disorders/impulsivity (including compulsive gambling and intermittent explosive disorder); mood disorders (including bipolar I disorder, bipolar II disorder, mania, mixed affective state, dépréssion {e.g., age-related dépréssion, major dépréssion, chronic dépréssion, seasonal dépréssion, psychotic dépréssion, postpartum dépréssion, and treatment résistant dépréssion (TRD)}; psychomotor disorders; psychotic disorders [including schizophrenia (including, for example, cognitive and négative symptoms in schizophrenia), schizoaffective disorder, schizophreniform, and delusional disorder]; substance abuse and drug dependence (including narcotic dependence, alcoholism, amphétamine dependence, cocaine addiction, nicotine φ dependence, and drug withdrawal syndrome); drug abuse relapse, eating disorders (including anorexia, bulimia, binge eating disorder, overeating, hyperphagia, and pagophagia); autism spectrum disorder (e.g., autism); chronic apathy, anhedonia, chronic fatigue, seasonal affective disorder, and pédiatrie psychiatrie disorders (including attention déficit disorder, attention déficit 5 hyperactive disorder (ADHD), conduct disorder, and autism)], endocrine disorders (such as hyperprolactinemia), or other disorders including drowsiness, excessive daytime sleepiness, cachexia, inattention, sexual dysfunction (e.g., erectile dysfunction, post-SSRI sexual dysfunction), pain, migraine, systemic lupus erythematosus (SLE), hyperglycemia, atherosclerosis, dislipidemia, obesity, diabètes, sepsis, post-ischemic tubular necrosis, rénal failure, hyponatremia, résistant edema, narcolepsy, cardiovascular disease (e.g., hypertension), congestive heart failure, postoperative ocula hypotonia, sleep disorders, and serotonin syndrome.
Another embodiment of the invention provides a method for treating neurological disorders [such as Tourette’s syndrome; tardive dyskinesia; Parkinson’s disease; cognitive disorders [including amnesia, senile dementia, HIV-associated dementia, Alzheimer’sassociated dementia, Huntington’s-associated dementia, Lewy body dementia, vascular dementia, drug-related dementia (for example, cognitive impairment associated with D2 antagonist therapy), delirium, and mild cognitive impairment)}; RLS; and Huntington's chorea/ disease], psychiatrie disorders [such as anxiety (including acute stress disorder, generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive-compulsive disorder); factitious disorder (including acute hallucinatory mania); impulse control disorders/impulsivity (including compulsive gambling and intermittent explosive disorder); mood disorders (including bipolar I disorder, bipolar II disorder, mania, mixed affective state, major dépréssion, chronic dépréssion, seasonal dépréssion, psychotic dépréssion, and postpartum dépréssion); psychomotor disorders; psychotic disorders (including schizophrenia, schizoaffective disorder, schizophreniform, and delusional disorder); drug dependence (including narcotic dependence, alcoholism, amphétamine dependence, cocaine addiction, nicotine dependence, and drug withdrawal syndrome); eating disorders (including anorexia, bulimia, binge eating disorder, hyperphagia, and pagophagia); and pédiatrie psychiatrie disorders (including attention déficit disorder, attention deficit/hyperactive disorder, conduct disorder, and autism)], or endocrine disorders (such as hyperprolactinemia) in a mammal, for example a human, comprising administering to said mammal a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
Another embodiment of the invention includes a method for treating a disorder in a mammal (e.g., a human), which method comprises administering to said mammal a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, wherein the disorder is selected from schizophrenia (e.g., cognitive and négative symptoms in schizophrenia), cognitive impairment [e.g., cognitive impairment associated with schizophrenia, cognitive impairment associated with AD, cognitive impairment associated with
PD, cognitive impairment associated with pharmacotherapy therapy (e.g., D2 antagonist therapy), and mild cognitive impairment], attention déficit hyperactivity disorder (ADHD), impulsivity, compulsive gambling, an eating disorder (e.g., anorexia, bulimia, binge eating disorder, overeating, hyperphagia, and pagophagia), autism spectrum disorder, mild cognitive impairment (MCI), age-related cognitive décliné, dementia (e.g., senile dementia, HIVassociated dementia, Alzheimer’s dementia, Lewy body dementia, vascular dementia, or frontotemporal dementia), restless leg syndrome (RLS), Parkinson’s disease, Huntington’s chorea, anxiety, dépréssion (e.g., age-related dépréssion), major dépressive disorder (MDD), treatment résistant dépréssion (TRD), bipolar disorder, chronic apathy, anhedonia, chronic fatigue, post-traumatic stress disorder, seasonal affective disorder, social anxiety disorder, postpartum dépréssion, serotonin syndrome, substance abuse and drug dependence, drug abuse relapse, Tourette’s syndrome, tardive dyskinesia, drowsiness, excessive daytime sleepiness, cachexia, inattention, sexual dysfunction (e.g., erectile dysfunction or post-SSRI sexual dysfunction), migraine, systemic lupus erythematosus (SLE), hyperglycemia, atherosclerosis, dislipidemia, obesity, diabètes, sepsis, post-ischemic tubular necrosis, rénal failure, hyponatremia, résistant edema, narcolepsy, hypertension, congestive heart failure, postoperative ocular hypotonia, sleep disorders, and pain.
Another embodiment of the invention includes a method for treating schizophrenia (e.g., cognitive and négative symptoms in schizophrenia or cognitive impairment associated with schizophrenia) or psychosis in a mammal, for example a human, comprising administering to said mammal (e.g., a human) a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
Another embodiment of the invention includes a method for treating schizophrenia (e.g., cognitive and négative symptoms in schizophrenia or cognitive impairment associated with schizophrenia) in a mammal, for example a human, comprising administering to said mammal a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
Another embodiment of the invention includes a method for the treatment of cognitive impairment [e.g., cognitive impairment associated with schizophrenia, cognitive impairment associated with AD, or cognitive impairment associated with PD] in a mammal, for example a human, comprising administering to said mammal a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
Another embodiment of the invention includes a method for treating AD (e.g., treating cognitive impairment associated with AD), PD (e.g., treating cognitive impairment associated with PD), RLS, dépréssion, or MDD in a mammal, for example a human, comprising administering to said mammal a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
The term therapeutically effective amount as used herein refers to that amount of the compound (including a pharmaceutically acceptable sait thereof) being administered which will relieve to some extent one or more of the symptoms of the disorder being treated. In reference to the treatment of a D1-mediated disorder (e.g., schizophrenia), a therapeutically effective amount refers to that amount which has the effect of relieving to some extent (or, for example, eliminating) one or more symptoms associated with a D1-mediated disorder (e.g., schizophrenia, or cognitive and négative symptoms in schizophrenia, or cognitive impairment associated with schizophrenia).
The term treating, as used herein, unless otherwise indicated, means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term treatment, as used herein, unless otherwise indicated, refers to the act of treating as treating is defined herein. The term “treating” also includes adjuvant and neo-adjuvant treatment of a subject.
Administration of the compounds of Formula I may be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intranasal routes, inhaled routes, intraduodenal routes, parentéral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion), topical, and rectal administration.
In one embodiment of the présent invention, the compounds of Formula I may be administered/effected by oral routes.
Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parentéral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to physically discrète units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The spécifications for the dosage unit forms of the invention are dictated by a variety of factors such as the unique characteristics of the therapeutic agent and the particular therapeutic or prophylactic effect to be achieved. In one embodiment of the présent invention, the compounds of Formula I may be used to treat humans.
It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It is to be further understood that for any particular subject, spécifie dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamie parameters, which may include clinical effects such as toxic effects and/or laboratory values. Thus, the présent invention encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regimens for administration of the chemotherapeutic agent is well-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
The amount of the compound of Formula I or a pharmaceutically acceptable sait thereof administered will be dépendent on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discrétion of the prescribing physician. Generally, an effective dosage is in the range of about 0.0001 to about 50 mg per kg body weight per day, for example about 0.01 to about 10 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.007 mg to about 3500 mg/day, for example about 0.7 mg to about 700 mg/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adéquate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several small doses for administration throughout the day.
As used herein, the term “combination therapy” refers to the administration of a compound of Formula I or a pharmaceutically acceptable sait thereof together with an at least one additional pharmaceutical or médicinal agent (e.g., an anti-schizophrenia agent), either sequentially or simultaneously.
The présent invention includes the use of a combination of a compound of Formula I (or a pharmaceutically acceptable sait thereof) and one or more additional pharmaceutically active agent(s). If a combination of active agents is administered, then they may be administered sequentially or simultaneously, in separate dosage forms or combined in a single dosage form. Accordingly, the présent invention also includes pharmaceutical compositions comprising an amount of: (a) a first agent comprising a compound of Formula I (including an A/-oxide thereof or a pharmaceutically acceptable sait of the compound or the /V-oxide); (b) a second pharmaceutically active agent; and (c) a pharmaceutically acceptable carrier, vehicle or diluent.
Various pharmaceutically active agents may be selected for use in conjunction with the compounds of Formula I (including or pharmaceutically acceptable salts thereof), depending on the disease, disorder, or condition to be treated. Pharmaceutically active agents that may be used in combination with the compositions of the présent invention include, without limitation:
(i) acetylcholinesterase inhibitors such as donepezil hydrochloride (ARICEPT, MEMAC); or Adenosine A2a receptor antagonists such as Preladenant (SCH 420814) or SCH 412348;
(ii) amyloid-β (or fragments thereof), such as AB^sConjugated to pan HLA DR-binding epitope (PADRE) and ACC-001 (Elan/Wyeth);
(iii) antibodies to amyloid-B (or fragments thereof), such as bapineuzumab (also known as AAB-001) and AAB-002 (Wyeth/Elan);
(iv) amyloid-lowering or -inhibiting agents (including those that reduce amyloid production, accumulation and fibrillization) such as colostrinin and bisnorcymserine (also known as BNC);
(v) alpha-adrenergic receptor agonists such as clonidine (CATAPRES);
(vi) beta-adrenergic receptor blocking agents (beta blockers) such as carteolol;
(vii) anticholinergics such as amitriptyline (ELAVIL, ENDEP);
(viii) anticonvulsants such as carbamazepine (TEGRETOL, CARBATROL);
(ix) antipsychotics, such as lurasidone (also known as SM-13496; Dainippon Sumitomo);
(x) calcium channel blockers such as nilvadipine (ESCOR, NIVADIL);
(xi) catechol O-methyltransferase (COMT) inhibitors such as tolcapone (TASMAR);
(xii) central nervous system stimulants such as caffeine;
(xiii) corticosteroids such as prednisone (STERAPRED, DELTASONE);
(xiv) dopamine receptor agonists such as apomorphine (APOKYN);
(xv) dopamine receptor antagonists such as tetrabenazine (NITOMAN, XENAZINE, dopamine D2 antagonist such as Quetiapine);
(xvi) dopamine reuptake inhibitors such as nomifensine maleate (MERITAL);
(xvii) gamma-aminobutyric acid (GABA) receptor agonists such as baclofen (LIORESAL, KEMSTRO);
(xviii) histamine 3 (H3) antagonists such as ciproxifan;
(xix) immunomodulators such as glatiramer acetate (also known as copolymer-1; COPAXONE);
(xx) immunosuppressants such as methotrexate (TREXALL, RHEUMATREX);
(xxi) interferons, including interferon beta-1a (AVONEX, REBIF) and interferon beta-1b (BETASERON, BETAFERON);
(xxii) levodopa (or its methyl or ethyl ester), alone or in combination with a DOPA decarboxylase inhibitor (e.g., carbidopa (SINEMET, CARBILEV, PARCOPA));
(xxiii) /V-methyl-D-aspartate (NMDA) receptor antagonists such as memantine (NAMENDA, AXURA, EBIXA);
(xxiv) monoamine oxidase (MAO) inhibitors such as selegiline (EMSAM);
(xxv) muscarinic receptor (particularly M1 subtype) agonists such as bethanechol chloride (DUVOID, URECHOLINE);
(xxvi) neuroprotective drugs such as 2,3,4,9-tetrahydro-1 H-carbazol-3-one oxime;
(xxvii) nicotinic receptor agonists such as epibatidine;
(xxviii) norepinephrine (noradrenaline) reuptake inhibitors such as atomoxetine (STRATTERA);
(xxix) phosphodiesterase (PDE) inhibitors, for example,PDE9 inhibitors such as BAY 736691 (Bayer AG) and PDE 10 (e.g. PDE10A) inhibitors such as papaverine;
(xxx) other PDE inhibitors including (a) PDE1 inhibitors (e.g., vinpocetine), (b) PDE2 inhibitors (e.g., erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA)), (c) PDE4 inhibitors (e.g., rolipram), and (d) PDE5 inhibitors (e.g., sildenafil (VIAGRA, REVATIO));
(xxxi) quinolines such as quinine (including its hydrochloride, dihydrochloride, sulfate, bisulfate and gluconate salts);
(xxxii) β-secretase inhibitors such as WY-25105;
(xxxiii) γ-secretase inhibitors such as LY-411575 (Lilly);
(xxxiv) serotonin (5-hydroxytryptamine) 1A (5-HT1A) receptor antagonists such as spiperone;
(xxxv) serotonin (5-hydroxytryptamine) 4 (5-HT4) receptor agonists such as PRX-03140 (Epix);
(xxxvi) serotonin (5-hydroxytryptamine) 6 (5-HT6) receptor antagonists such as mianserin (TORVOL, BOLVIDON, NORVAL);
(xxxvii) serotonin (5-HT) reuptake inhibitors such as alaproclate, citalopram (CELEXA, CIPRAMIL);
(xxxviii) trophic factors, such as nerve growth factor (NGF), basic fibroblast growth factor (bFGF; ERSOFERMIN), neurotrophin-3 (NT-3), cardiotrophin-1, brain-derived neurotrophic factor (BDNF), neublastin, meteorin, and glial-derived neurotrophic factor (GDNF), and agents that stimulate production of trophic factors, such as propentofylline;
and the like.
The compound of Formula I (including a pharmaceutically acceptable sait thereof) is optionally used in combination with another active agent. Such an active agent may be, for example, an atypical antipsychotic or an anti-Parkinson’s disease agent or an anti-Alzheimer’s agent. Accordingly, another embodiment of the invention provides methods of treating a D1mediated disorder (e.g., a neurological and psychiatrie disorder associated with D1), comprising administering to a mammal an effective amount of a compound of Formula I (including an Noxide thereof or a pharmaceutically acceptable sait of the compound or the /V-oxide) and further comprising administering another active agent.
As used herein, the term “another active agent” refers to any therapeutic agent, other than the compound of Formula I (including or a pharmaceutically acceptable sait thereof) that is useful for the treatment of a subject disorder. Examples of additional therapeutic agents include antidepressants, antipsychotics (such as anti-schizophrenia), anti-pain, anti-Parkinson’s disease agents, anti-LID (levodopa-induced dyskinesia), anti-Alzheimer’s and anti-anxiety agents. Examples of particular classes of antidepressants that can be used in combination with the compounds of the invention include norepinephrine reuptake inhibitors, sélective serotonin reuptake inhibitors (SSRIs), NK-1 receptor antagonists, monoamine oxidase inhibitors (MAOIs), réversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, a-adrenoreceptor antagonists, and atypical antidepressants. Suitable norepinephrine reuptake inhibitors include tertiary amine tricyclics and secondary amine tricyclics. Examples of suitable tertiary amine tricyclics and secondary amine tricyclics include amitriptyhne, clomipramine, doxepin, imipramine, trimipramine, dothiepin, butriptyline, iprindole, lofepramine, nortriptyline, protriptyline, amoxapine, desipramine and maprotiline. Examples of suitable sélective serotonin reuptake inhibitors include fluoxetine, fluvoxamine, paroxetine, and sertraline. Examples of monoamine oxidase inhibitors include isocarboxazid, phenelzine, and tranylcyclopramine. Examples of suitable réversible inhibitors of monoamine oxidase include moclobemide. Examples of suitable serotonin and noradrenaline reuptake inhibitors of use in the présent invention include venlafaxine. Examples of suitable atypical anti-depressants include bupropion, lithium, nefazodone, trazodone and viloxazine. Examples of anti-Alzheimer’s agents include Dimebon, NMDA receptor antagonists such as memantine; and cholinestérase inhibitors such as donepezil and galantamine. Examples of suitable classes of anti-anxiety agents that can be used in combination with the compounds of the invention include benzodiazépines and serotonin 1A (5-HT1A) agonists or antagonists, especially 5-HT1A partial agonists, and corticotropin releasing factor (CRF) antagonists. Suitable benzodiazépines include alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, halazepam, lorazépam, oxazepam, and prazepam. Suitable 5-HT1A receptor agonists or antagonists include buspirone, flesinoxan, gepirone, and ipsapirone. Suitable atypical antipsychotics include paliperidone, bifeprunox, ziprasidone, rispéridone, aripiprazole, olanzapine, and quetiapine. Suitable nicotine acétylcholine agonists include ispronicline, varenicline and MEM 3454. Anti-pain agents include pregabalin, gabapentin, clonidine, neostigmine, baclofen, midazolam, ketamine and ziconotide. Examples of suitable anti-Parkinson’s disease agents include L-DOPA (or its methyl or ethyl ester), a DOPA decarboxylase inhibitor (e.g., carbidopa (SINEMET, CARBILEV, PARCOPA), an Adenosine A2a receptor antagonist [e.g., Preladenant (SCH 420814) or SCH 412348], benserazide (MADOPAR), α-methyldopa, monofluoromethyldopa, difluoromethyldopa, brocresine, or m-hydroxybenzylhydrazine), a dopamine agonist [such as apomorphine (APOKYN), bromocriptine (PARLODEL), cabergoline (DOSTINEX), dihydrexidine, dihydroergocryptine, fenoldopam (CORLOPAM), lisuride (DOPERGIN), pergolide (PERMAX), piribedil (TRIVASTAL, TRASTAL), pramipexole (MIRAPEX), quinpirole, ropinirole (REQUIP), rotigotine (NEUPRO), SKF-82958 (GlaxoSmithKIine), and sarizotan], a monoamine oxidase (MAO) inhibitor [such as selegiline (EMSAM), selegiline hydrochloride (L-deprenyl, ELDEPRYL, ZELAPAR), dimethylselegilene, brofaromine, phenelzine (NARDIL), tranylcypromine (PARNATE), moclobemide (AURORIX, MANERIX), befloxatone, safinamide, isocarboxazid (MARPLAN), nialamide (NIAMID), rasagiline (AZILECT), iproniazide (MARSILID, IPROZID, IPRONID), CHF-3381 (Chiesi Farmaceutici), iproclozide, toloxatone (HUMORYL, PERENUM), bifemelane, desoxypeganine, harmine (also known as telepathine or banasterine), harmaline, linezolid (ZYVOX, ZYVOXID), and pargyline (EUDATIN, SUPIRDYL)], a catechol O47 methyltransferase (COMT) inhibitor [such as tolcapone (TASMAR), entacapone (COMTAN), and tropolone], an /V-methyl-D-aspartate (NMDA) receptor antagonist [such as amantadine (SYMMETREL)j, antichohnergics [such as amitriptyhne (ELAVIL, ENDEP), butriptyline, benztropine mesylate (COGENTIN), trihexyphenidyl (ARTANE), diphenhydramine (BENADRYL), orphenadrine (NORFLEX), hyoscyamine, atropine (ATROPEN), scopolamine (TRANSDERM-SCOP), scopolamine methylbromide (PARMINE), dicycloverine (BENTYL, BYCLOMINE, DIBENT, DILOMINE, tolterodine (DETROL), oxybutynin (DITROPAN, LYRINEL XL, OXYTROL), penthienate bromide, propantheline (PRO-BANTHINE), cyclizine, imipramine hydrochloride (TOFRANIL), imipramine maleate (SURMONTIL), lofepramine, desipramine (NORPRAMIN), doxepin (SINEQUAN, ZONALON), trimipramine (SURMONTIL), and glycopyrrolate (ROBINUL)], or a combination thereof. Examples of anti-schizophrenia agents include ziprasidone, rispéridone, olanzapine, quetiapine, aripiprazole, asenapine, blonanserin, or iloperidone.
As noted above, the compounds of Formula I (including pharmaceutically acceptable salts thereof) may be used in combination with one or more additional anti-schizophrenia agents which are described herein. When a combination therapy is used, the one or more additional anti-schizophrenia agents may be administered sequentially or simultaneously with the compound of the invention. In one embodiment, the additional anti-schizophrenia agent is administered to a mammal (e.g., a human) prior to administration of the compound of the invention. In another embodiment, the additional anti-schizophrenia agent is administered to the mammal after administration of the compound of the invention. In another embodiment, the additional anti-schizophrenia agent is administered to the mammal (e.g., a human) simultaneously with the administration of the compound of the invention (or an /V-oxide thereof or a pharmaceutically acceptable sait of the foregoing).
The invention also provides a pharmaceutical composition for the treatment of schizophrenia in a mammal, including a human, which comprises an amount of a compound of Formula I (or a pharmaceutically acceptable sait thereof), as defined above (including hydrates, solvatés and polymorphs of said compound or pharmaceutically acceptable salts thereof), in combination with one or more (for example one to three) anti-schizophrenia agents such as ziprasidone, rispéridone, olanzapine, quetiapine, aripiprazole, asenapine, blonanserin, or iloperidone, wherein the amounts of the active agent and the combination when taken as a whole are therapeutically effective for treating schizophrenia.
The invention also provides a pharmaceutical composition for the treatment of
Parkinson’s disease in a mammal (including cognition impairment associated with PD), including a human, which comprises an amount of a compound of Formula I (or a pharmaceutically acceptable sait thereof), as defined above (including hydrates, solvatés and polymorphs of said compound or pharmaceutically acceptable salts thereof), in combination with one or more (for example one to three) anti-Parkinson’s disease agents such as L-DOPA,
wherein the amounts of the active agent and the combination when taken as a whole are therapeutically effective for treating Parkinson’s disease.
It will be understood that the compounds of Formula I depicted above are not limited to a particular stereoisomer (e.g. enantiomer or atropisomer) shown, but also include ail stereoisomers and mixtures thereof.
DETAILED DESCRIPTION OF THE INVENTION
Compounds of the invention, including salts thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
The reactions for preparing compounds of the invention can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the températures at which the reactions are carried out, e.g., températures that can range from the solvents freezing température to the solvent's boiling température. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Préparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the sélection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic résonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographie methods such as highperformance liquid chromatography (HPLC) or thin layer chromatography (TLC).
Compounds of Formula I and intermediates thereof may be prepared according to the following reaction schemes and accompanying discussion. Unless otherwise indicated, R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R10B, R11, R11A, T1, T2, T3, T4, Q1, and X1, and structural Formula I in the reaction schemes and discussion that follow are as defined above. In general, the compounds of this invention may be made by processes which include processes analogous to those known in the chemical arts, particularly in light of the description provided herein. Certain processes for the manufacture of the compounds of this invention and intermediates thereof are provided as further features of the invention and are illustrated by the following reaction schemes. Other processes are described in the experimental section. The schemes and examples provided herein (including the corresponding description) are for illustration only, and not intended to limit the scope of the présent invention.
Scheme 1
Scheme 1 refers to préparation of compounds of Formula 1-5 (i.e., compounds of
Formula I wherein L1 is O). Referring to Scheme 1, compounds of Formula 1-1 [where Lg1 is a suitable leaving group such as halo (e.g., F, Cl or Br)] and 1-2 [wherein Z1 can be, e.g., halogen (e.g., Br or I) or trifluoromethanesulfonate (triflate)] are commercially available or can be made by methods described herein or other methods well known to those skilled in the art. A compound of Formula 1-3 can be prepared by coupling a compound of Formula 1-1 with a compound of Formula 1-2 under suitable conditions. The coupling can be accomplished, for example, by heating a mixture of a compound of Formula 1-1 with a compound of Formula 1-2 in the presence of a base, such as Cs2CO3, in an appropriate solvent, such as dimethyl sulfoxide (DMSO). Altematively, a metal-catalyzed (such as using a palladium or copper catalyst) coupling may be employed to accomplish the aforesaid coupling. In this variant of the coupling, a mixture of a compound of Formula 1-1 and a compound of Formula 1-2 can be heated in the presence of a base (such as Cs2CO3), a métal catalyst [such as a palladium catalyst, e.g., Pd(OAc)2], and a ligand [such as 1,T-binaphthalene-2,2‘diylbis(diphenylphosphane) (BINAP)] in an appropriate solvent, such as 1,4-dioxane. A compound of Formula 1-3 can subsequently be reacted with a compound of Formula Q1-Z2 [wherein Z2 can be Br; B(OH)2; B(OR)2 wherein each R is independently H or CA alkyl, or wherein the two (OR) groups, together with the B atom to which they are attached, form a 5- to 10-membered heterocycloalkyl optionally substituted with one or more CA alkyl; a trialkyltin moiety; or the like] by a metal-catalyzed (such as using a palladium catalyst) coupling reaction to obtain a compound of Formula I. Compounds of Formula Q1-Z2 are commercially available or can be made by methods described herein or by methods analogous to those described in the chemical art. Alternatively, a compound of Formula 1-3 can be converted to a compound of Formula 1-4 (wherein Z2 is defined as above). For example, a compound of Formula 1-3 (wherein Z1 is halogen such as Br or I) can be converted to a compound of Formula 1-4 [wherein Z2 is B(OH)2; B(OR)2 wherein each R is independently H or C^e alkyl, or wherein the two (OR) groups, together with the B atom to which they are attached, form a 5- to 10membered heterocycloalkyl or heteroaryl optionally substituted with one or more alkyl] by methods described herein or other methods well known to those skilled in the art. In this example, this reaction can be accomplished, for example, by reacting a compound of Formula 1-3 (wherein Z1 is halogen such as Br) with 4,4,4',4',5,5,5,,5'-octamethyl-2,2'-bi-1,3,2dioxaborolane, a suitable base (such as potassium acetate), and a palladium catalyst {such as [1,T-bis(diphenylphosphino)ferrocene]dichloropalladium(ll)} in a suitable solvent such as 1,4dioxane. In another example, a compound of Formula 1-3 (wherein Z1 is halogen such as Br) can be converted to a compound of Formula 1-4 (wherein Z2 is a trialkyltin moiety) by alternate methods described herein or other methods well known to those skilled in the art. In this example, this reaction can be accomplished, for example, by reacting a compound of Formula 1-3 (wherein Z1 is halogen such as Br) with a hexaalkyldistannane (such as hexamethyldistannane) in the presence of a palladium catalyst [such as tetrakis(triphenylphosphine)paliadium(0)] in a suitable solvent such as 1,4-dioxane. A compound of Formula 1-4 can then be reacted with a compound of Formula Q1-Z1 (wherein Z1 is defined as above) by a metal-catalyzed (such as using a palladium catalyst) coupling reaction to obtain a compound of Formula I. Compounds of Formula Q1-Z1 are commercially available or can be made by methods described herein or by methods analogous to those described in the chemical art. The type of reaction employed dépends on the sélection of Z1 and Z2. For example, when Z1 is halogen or triflate and the Q1-Z2 reagent is a boronic acid or boronic ester, a Suzuki reaction may be used [A. Suzuki, J. Organomet. Chem. 1999, 576, 147-168; N. Miyaura and A. Suzuki, Chem. Rev. 1995, 95, 2457-2483; A. F. Littke et al., J. Am. Chem. Soc. 2000, 122, 4020-4028]. In some spécifie embodiments, an aromatic iodide, bromide, or triflate of Formula 1-3 is combined with an aryl or heteroaryl boronic acid or boronic ester of Formula Q1-Z2 and a suitable base, such as potassium phosphate, in a suitable organic solvent such as tetrahydrofüran (THF). A palladium catalyst is added, such as S-Phos precatalyst {also known as chloro(2-dicyclohexylphosphino-2',6'-dimethoxy-1,T-biphenyl)[2-(2aminoethylphenyl)]palladium(ll) - tert-butyl methyl ether adduct], and the reaction mixture is heated. Alternatively, when Z1 is halogen or triflate and Z2 is trialkyltin, a Stille coupling may be employed [V. Farina et al., Organic Reactions 1997, 50, 1-652]. More specifically, a compound of Formula 1-3 (wherein Z1 is Br, I, or triflate ) may be combined with a compound of Formula φ Q1-Z2 (wherein the Q1-Z2 compound is a Q1-stannane compound) in the presence of a palladium catalyst, such as dichlorobis(triphenylphosphine)palladium(ll), in a suitable organic solvent such as toluene, and the reaction may be heated. Where Z1 is Br, I, or triflate and Z2 is Br or I, a
Negishi coupling may be used [E. Erdik, Tetrahedron 1992, 48, 9577-9648], More specifically, a compound of Formula 1-3 (wherein Z1 is Br, I, or triflate) may be transmetallated by treatment with 1 to 1.1 équivalents of an alkyllithium reagent followed by a solution of 1.2 to 1.4 équivalents of zinc chloride in an appropriate solvent such as THF at a température ranging from -80 °C to -65 °C. After warming to a température between 10 °C and 30 °C, the reaction mixture may be treated with a compound of Formula Q1-Z2 (wherein Z2 is Br or I), and heated at
50 °C to 70 °C with addition of a catalyst such as tetrakis(triphenylphosphine)paliadium(0). The reaction may be carried out for times ranging from 1 to 24 hours to yield the compound of Formula 1-5.
Scheme 1’
Similar to the chemical transformations described in Scheme 1, compounds of Formula I can be prepared starting from compounds of Formula V-3 according to Scheme 1’
Scheme 2
2-4
1-1
Scheme 2 also refers to préparation of compounds of Formula 1-5. Referring to Scheme
2, compounds of Formula 1-5 may be prepared utilizing analogous chemical transformations to those described in Scheme 1, but with a different ordering of steps. Compounds of Formula 2-1 [wherein Pg1 is a suitable protecting group such as methyl, benzyl, tetrahydropyranyl (THP), or te/t-butyldimethyl (TBS)] are commercially available or can be made by methods described herein or other methods well known to those skilled in the art. A compound of Formula 2-1 can be converted to a compound of Formula 2-2 either directly or after conversion to a compound of Formula 2-3 using methods analogous to those described in Scheme 1. A compound of Formula 2-2 may then be deprotected, using appropriate conditions depending on the sélection of the Pg1 group, to obtain a compound of Formula 2-4, which in turn can be coupled with a compound of Formula 1-1 in Scheme 1 to afford a compound of Formula 1-5. The coupling conditions employed may be analogous to those described for the préparation of a compound of Formula 1-3 in Scheme 1.
Scheme 3
T3 A1a
Scheme 3 refers to a préparation of a compound of Formula 3-5 wherein A1 is a moiety of Formula A1a or a suitable protecting group Pg2. (e.g., methyl, benzyl, THP, or TBS). Referring to Scheme 3, compounds of Formula 3-1 are commercially available or can be made by methods described herein or other methods well known to those skilled in the art. A compound of Formula 3-2 can be prepared by reacting an arylketone of Formula 3-1 with an alkyl nitrite (e.g., isoamyl nitrite) in the presence of an acid (such as hydrochloric acid). The resulting oxime of Formula 3-2 can be converted to the diketone of Formula 3-3 upon treatment with formaldéhyde (or its équivalent such as métaformaldéhyde or polyformaldéhyde) in the presence of an acid (such as an aqueous hydrochloric acid solution). Diketones of Formula 3-3 can be reacted with glycinamide or a sait thereof (such as an acetic acid sait) in the presence of a base such as sodium hydroxide to obtain pyrazinones of Formula 3-4. Alkylation of the pyrazinone nitrogen to obtain a compound of Formula 3-5 can be achieved by treatment of a compound of Formula 3-4 with a base [such as lithium diisopropylamide (LDA), lithium bis(trimethylsilyl)amide (LHMDS), and the like] and a compound of the formula R -Z3 [wherein Z3 is an acceptable leaving group such as Cl, Br, I, methanesulfonate (mesylate), and the like and wherein R11A is for example C^alkyl (e.g., methyl)]. Suitable reaction solvents typically can be selected from polar aprotic solvents such as A/,A/-dimethylformamide (DMF), 1,4-dioxane, or
THF.
(a) heat (b) S; heat
A1 is Pg2 or a moiety of A1a:
Alternatively, a compound of Formula 3-5 may be prepared as in Scheme 4 wherein L1 is
O, NH, N(Ci-4 alkyl) and N(C3.6 cycloalkyl). Referring to Scheme 4, compounds of Formula 4-1 and 4-2 are commercially available or can be made by methods described herein or other methods well known to those skilled in the art. A compound of Formula 4-3 can be prepared by coupling a compound of Formula 4-1 with a compound of Formula 4-2. The aforesaid coupling may be accomplished by reacting a compound of Formula 4-1 with a compound of Formula 4-2 in the presence of a suitable base (such as potassium carbonate), a suitable catalyst [such as tetrakis(triphenylphosphine)palladium(0)], and a suitable solvent (such as éthanol). A compound of Formula 4-3 can be reacted with maleic anhydride and hydrogen peroxide in a solvent (such as dichloromethane) to provide a compound of Formula 4-4, which may contain a mixture of Noxide regioisomers. A compound of Formula 4-5 can be prepared from a compound of Formula 4-4 by heating with acetic anydride; the initial product can be saponified using a base (such as NaOH) in a suitable polar solvent (such as water or methanol). A compound of Formula 3-5 can be prepared from a compound of Formula 4-5 by reaction with a suitable base (such as LDA,
LHMDS and the like), lithium bromide, and a compound of the formula R11A-Z3 (wherein Z3 is an acceptable leaving group such as Cl, Br, I, mesylate, and the like). Suitable reaction solvents typically can be selected from polar aprotic solvents (such as DMF, 1,4-dioxane, or THF).
O
5-5 T3
Scheme 5 refers to a préparation of a compound of Formula 5-5 wherein L1 is O, NH, N(CV4 alkyl) and N(C3.6 cycloalkyl) and A1 is a moiety of Formula A1a or a Pg2 (such as a benzyl group). Referring to Scheme 5, compounds of Formula 5-1 and 5-2 are commercially available or can be made by methods described herein or other methods well known to those skilled in the art. A compound of Formula 5-3 can be prepared by coupling a compound of Formula 5-1 with an enol trifluoromethanesulfonate of Formula 5-2. The aforesaid coupling may be accomplished by reacting a compound of Formula 5-1 with a trifluoromethanesulfonate of Formula 5-2 in the presence of a suitable base (such as potassium carbonate or sodium carbonate), a suitable catalyst [such as palladium (II) acetate], optionally a suitable ligand (such as tricyclohexylphosphine), and optionally a suitable phase-transfer catalyst such as tetrabutylammonium chloride. Suitable reaction solvents typically can be selected from polar aprotic solvents such as 1,4-dioxane or THF. A compound of Formula 5-3 can be reacted with 1 to 5 équivalents of a suitable base [such as 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU)] under an oxygen atmosphère to obtain a compound of Formula 5-4. Suitable reaction solvents typically can be selected from polar aprotic solvents such as DMF, 1,4-dioxane, or THF. A compound of Formula 5-5 can be obtained by reacting a compound of Formula 5-4 with hydrazine in a suitable solvent such as 1-butanol.
Scheme 6
Scheme 6 refers to a préparation of a compound of Formula 6-5. Referring to Scheme 6, a compound of Formula 6-1 can be prepared as described in Scheme 5, wherein Pg2 is a suitable protecting group (such as benzyl). A compound of Formula 6-1 can be converted to a suitably protected compound of Formula 6-2 using methods described herein or other methods well known to those skilled in the art, wherein Pg3 is a suitable protecting group (such as THP) that can be removed under orthogonal reaction conditions to Pg2. A compound of Formula 6-3 can be prepared by sélective removal of Pg2 under suitable deprotection conditions depending 10 on the sélection of Pg2. For example, when Pg2 is a benzyl group, it can be removed by treatment with palladium (10% on carbon) under hydrogénation condition in a suitable solvent, such as methanol and ethyl acetate. Using the aforementioned reaction conditions described in Scheme 1, a compound of Formula 6-3 can be coupled with a reagent of Formula 1-1 to yield a compound of Formula 6-4. A compound of Formula 6-5 can be obtained by removing Pg3 under 15 suitable deprotection conditions depending on the sélection of Pg3. For example, when Pg3 is
THP, it can be removed under acidic conditions, such as hydrogen chloride in a suitable solvent, such as dichloromethane.
Scheme 7 refers to a préparation of a compound of Formula 7-5 [wherein R10 is, for example, Co alkyl (e.g., methyl ); R10B is, for example, H or Ον3 alkyl (e.g., methyl ); and Pg4is a suitable protecting group [e.g., 2-(trimethylsilyl)ethoxymethyl (SEM), tert-butoxycarbonyl (Boc), or benzyloxymethyl acetal (BOM)]. Referring to Scheme 7, compounds of Formula 2-3 and 7-1 are commercially available or can be prepared by methods described herein or other methods well known to those skilled in the art. A compound of Formula 7-2 can be prepared by coupling a compound of Formula 2-3 with a compound of Formula 7-1, in the presence of a suitable base (such as potassium carbonate) and a suitable catalyst (such as [1,1’bis(diphenylphosphino)ferrocene]dichloropalladium(ll)}. A compound of Formula 7-3 can be prepared by sélective removal of Pg2 under suitable de-protection conditions depending on the sélection of Pg2. For example, when Pg2 is a benzyl group, it can be removed by treatment with palladium (10% on carbon) under hydrogénation condition in a suitable solvent, such as methanol and ethyl acetate. Using the aforementioned reaction conditions described in Scheme
1, a compound of Formula 7-3 can be coupled with a reagent of Formula 1-1 to yield a compound of Formula 7-4. Alternatively, a compound of Formula 7-4 can be prepared from intermediate 1-4, following the coupling conditions described in Scheme 1. A compound of Formula 7-5 can then be obtained from a compound of Formula 7-4 by removing Pg4 under 20 suitable deprotection conditions that are known to those skilled in the art.
Scheme 8 refers to a préparation of a compound of Formula 8-1 [wherein R10 is, for example, Cmalkyl (e.g., methyl); R10B is, for example, H or Cmalkyl (e.g., methyl)]. Referring to
Scheme 8, compounds of Formula 8-1 can be prepared by treating a compound of Formula 7-5 with a suitable thianation reagent, such as Lawesson’s reagent [2,4-bis(4-methoxyphenyl)1,3,2,4-dithiadiphosphetane-2,4-dithione] or phosphorus pentasulfide, in a suitable solvent such as toluene.
Scheme 9
Scheme 9 refers to préparation of compounds of Formula 9-5 and 9-6. Referring to Scheme 9, compounds of Formula 9-1 are commercially available or can be made by methods described herein or other methods well known to those skilled in the art. A compound of Formula 9-1 can be converted to a compound of Formula 9-2 either directly or after conversion 15 to a compound of Formula 9-3 using methods analogous to those described in Scheme 1. The nitro group of a compound of Formula 9-2 can then be converted to an amine via hydrogénation in the presence of a suitable catalyst, such as palladium (10% on carbon), to yield a compound of Formula 9-4. A compound of Formula 9-4 can then be coupled with a compound of Formula 1-1 in Scheme 1 to afford a compound of Formula 9-5. The coupling conditions employed may 20 be analogous to those described for the préparation of a compound of Formula 1-3 in Scheme
1. A compound of Formula 9-6 can be prepared via /V-alkylation of a compound of formula 9-5
using a reagent of Y-Z3, wherein Y is Cm alkyl, or C3.6 cycloalkyl, and Z3 is an acceptable leaving group such as Cl, Br, I, mesylate, and the like.
Scheme 10
Scheme 10 refers to préparation of compounds of Formula 10-4. Referring to Scheme
10, a compound of Formula 10-1 can be prepared via triflation of a compound of Formula 2-4 (Scheme 2) using a suitable reagent such as trifluoromethanesulfonic anhydride in the presence of a suitable base such as triethylamine. A compound of Formula 10-1 can be converted to a compound of Formula 10-2 by coupling with potassium thioacetate, in the presence of a suitable 10 métal catalyst, such as tris(dibenzylideneacetone)dipalladium(0), and a suitable ligand, such as (R)-(-)-1-[(Sp)-2-(dicyclohexylphosphino)ferrocenyl]ethyldi-tert-butylphosphine, in a suitable solvent, such as toluene. A compound of Formula 10-2 can then be hydrolyzed to obtain a compound of Formula 10-3, which in turn can be coupled with a compound of Formula 1-1 in Scheme 1 to afford a compound of Formula 10-4. The coupling conditions employed may be 15 analogous to those described for the préparation of a compound of Formula 1-3 in Scheme 1. A compound of Formula 10-4 may then be deprotected, using appropriate conditions depending on the sélection of the Pg1 group, to obtain a compound of Formula I.
Additional starting materials and intermediates useful for making the compounds of the présent invention can be obtained from chemical vendors such as Sigma-Aldrich or can be 20 made according to methods described in the chemical art.
Those skilled in the art can recognize that in ail of the Schemes described herein, if there are functional (reactive) groups présent on a part of the compound structure such as a substituent group, for example R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R10B, R11, R11A, T1, T2, T3, T4, Q1, and X1 etc., further modification can be made if appropriate and/or desired, using 25 methods well known to those skilled in the art. For example, a -CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to an amide; a carboxylic acid can be converted to an ester, which in turn can be reduced to an alcohol, which in turn can be further modified. For another example, an OH group can be converted into a better leaving group such as a methanesulfonate, which in turn is suitable for nucleophilic substitution, such as by a φ cyanide ion (CN’). For another example, an -S- can be oxidized to -S(=O)- and/or -S(=O)2-. For yet another example, an unsaturated bond such as C=C or C^C can be reduced to a saturated bond by hydrogénation. In some embodiments, a primary amine or a secondary amine moiety (présent on a substituent group such as R3, R4, R9, R10, etc.) can be converted to an amide, sulfonamide, urea, or thiourea moiety by reacting it with an appropriate reagent such as an acid chloride, a sulfonyl chloride, an isocyanate, or a thioisocyanate compound. One skilled in the art will recognize further such modifications. Thus, a compound of Formula I having a substituent that contains a functional group can be converted to another compound of Formula I having a different substituent group.
Similarly, those skilled in the art can also recognize that in ail of the schemes described herein, if there are functional (reactive) groups présent on a substituent group such as R3, R4, R9, R10, etc., these functional groups can be protected/deprotected in the course of the synthetic scheme described here, if appropriate and/or desired. For example, an OH group can be protected by a benzyl, methyl, or acetyl group, which can be deprotected and converted back to the OH group in a later stage of the synthetic process. For another example, an NH2 group can be protected by a benzyloxycarbonyl (Cbz) or Boc group; conversion back to the NH2 group can be carried out at a later stage of the synthetic process via deprotection.
As used herein, the term “reacting” (or “reaction” or “reacted”) refers to the bringing together of designated chemical reactants such that a chemical transformation takes place generating a compound different from any initially introduced into the system. Reactions can take place in the presence or absence of solvent.
Compounds of Formula I may exist as stereoisomers, such as atropisomers, racemates, enantiomers, or diastereomers. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate using, for example, chiral high pressure liquid chromatography (HPLC). Altematively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound contains an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to one skilled in the art. Chiral compounds of Formula I (and chiral precursors thereof) may be obtained in enantiomerically enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0% to 50% 2-propanol, typically from
2% to 20%, and from 0% to 5% of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. Stereoisomeric conglomérâtes may be separated by conventional techniques known to those skilled in the art. See, e.g., Stereochemistry of Organic Compounds by E. L. Eliel and S. H. Wilen (Wiley, New York, 1994), the disclosure of which is φ incorporated herein by reference in its entirety. Suitable stereoselective techniques are wellknown to those of ordinary skill in the art.
Where a compound of Formula I contains an alkenyl or alkenylene (alkylidene) group, géométrie cis/trans (or Z/E) isomers are possible. Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallization. Salts of the présent invention can be prepared according to methods known to those of skill in the art.
The compounds of Formula I that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. Although such salts must be 10 pharmaceutically acceptable for administration to animais, it is often désirable in practice to initially isolate the compound of the présent invention from the reaction mixture as a pharmaceutically unacceptable sait and then simply convert the latter back to the free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition sait. The acid addition salts of the basic 15 compounds of this invention can be prepared by treating the basic compound with a substantially équivalent amount of the selected minerai or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or éthanol. Upon évaporation of the solvent, the desired solid sait is obtained. The desired acid sait can also be precipitated from a solution of the free base in an organic solvent by adding an appropriate minerai or organic acid 20 to the solution.
If the inventive compound is a base, the desired pharmaceutically acceptable sait may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic 25 acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, isonicotinic acid, lactic acid, pantothenic acid, bitartric acid, ascorbic acid, 2,5dihydroxybenzoic acid, gluconic acid, saccharic acid, formic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, and pamoic [i.e., 4,4'methanediylbis(3-hydroxynaphthalene-2-carboxylic acid)] acid, a pyranosidyl acid, such as 30 glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as ethanesulfonic acid, or the like.
Those compounds of Formula I that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include the 35 alkali métal or alkaline earth métal salts, and particularly the sodium and potassium salts.
These salts are ail prepared by conventional techniques. The chemical bases which are used as reagents to préparé the pharmaceutically acceptable base salts of this invention are those which form non-toxic base salts with the acidic compounds of Formula I. These salts may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali métal hydroxide or alkaline earth métal hydroxide, or the like. These salts can also be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, for example under reduced pressure. Altematively, they may also be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali métal alkoxide together, and then evaporating the resulting solution to dryness in the same manner as before. In either case, stoichiometric quantities of reagents are, for example, employed in order to ensure completeness of reaction and maximum yields of the desired final product.
Pharmaceutically acceptable salts of compounds of Formula I (including compounds of Formula la or Ib) may be prepared by one or more of three methods:
(i) by reacting the compound of Formula I with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of Formula I or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or (iii) by converting one sait of the compound of Formula I to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column.
Ail three reactions are typically carried out in solution. The resulting sait may precipitate out and be collected by filtration or may be recovered by évaporation of the solvent. The degree of ionization in the resulting sait may vary from completely ionized to almost non-ionized.
Polymorphs can be prepared according to techniques well-known to those skilled in the art, for example, by crystallization.
When any racemate crystallizes, crystals of two different types are possible. The first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. The second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer.
While both of the crystal forms présent in a racemic mixture may hâve almost identical physical properties, they may hâve different physical properties compared to the true racemate. Racemic mixtures may be separated by conventional techniques known to those skilled in the art - see, for example, Stereochemistry of Organic Compounds by E. L. Eliel and S. H. Wilen (Wiley, New York, 1994).
The invention also includes isotopically labeled compounds of Formula I wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Isotopically labeled compounds of Formula I (or pharmaceutically acceptable salts thereof or Noxides thereof) can generally be prepared by conventional techniques known to those skilled in φ the art or by processes analogous to those described herein, using an appropriate isotopically labeled reagent in place of the non-labeled reagent otherwise employed.
Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities présent in the compounds of Formula I with certain moieties known to 5 those skilled in the art as ‘pro-moieties’ as described, for example, in Design of Prodrugs by H. Bundgaard (Elsevier, 1985).
The compounds of Formula I should be assessed for their biopharmaceutical properties, such as solubility and solution stability (across pH), permeability, etc., in order to select the most appropriate dosage form and route of administration for treatment of the proposed indication.
Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as précipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
They may be administered alone or in combination with one or more other compounds of 15 the invention or in combination with one or more other drugs (or as any combination thereof).
Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable excipients. The term “excipient” is used herein to describe any ingrédient other than the compound(s) of the invention. The choice of excipient will to a large extent dépend on factors such as the particular mode of administration, the effect of the 20 excipient on solubility and stability, and the nature of the dosage form.
Pharmaceutical compositions suitable for the delivery of compounds of the présent invention (or pharmaceutically acceptable salts thereof) and methods for their préparation will be readily apparent to those skilled in the art. Such compositions and methods for their préparation may be found, for example, in Remington’s Pharmaceutical Sciences, 19th Edition 25 (Mack Publishing Company, 1995).
The compounds of the invention (including pharmaceutically acceptable salts thereof and /V-oxides thereof) may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual, or sublingual administration by which the compound enters the blood stream directly from the mouth.
Formulations suitable for oral administration include solid, semi-solid and liquid Systems such as tablets; soft or hard capsules containing multi- or nano-particulates, liquids, or powders;
lozenges (including liquid-filled); chews; gels; fast dispersing dosage forms; films; ovules;
sprays; and buccal/mucoadhesive patches.
Liquid formulations include suspensions, solutions, syrups and élixirs. Such formulations may be employed as fillers in soft or hard capsules (made, for example, from gelatin or hydroxypropyl methyl cellulose) and typically comprise a carrier, for example, water, éthanol, polyethylene glycol, propylene glycol, methyl cellulose, or a suitable oil, and one or more
emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.
The compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described by Liang and Chen, Expert Opinion in Therapeutic 5 Patents 2001, 11, 981-986.
For tablet dosage forms, depending on dose, the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form. In addition to the drug, tablets generally contain a disintegrant. Examples of disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl cellulose, starch, pregelatinized starch and sodium alginate. Generally, the disintegrant will comprise from 1 weight % to 25 weight %, for example, from 5 weight % to 20 weight % of the dosage form.
Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When présent, surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
Tablets also generally contain lubricants such as magnésium stéarate, calcium stéarate, zinc stéarate, sodium stearyl fumarate, and mixtures of magnésium stéarate with sodium lauryl sulfate. Lubricants generally comprise from 0.25 weight % to 10 weight %, for example, from 0.5 weight % to 3 weight % of the tablet.
Other possible ingrédients include anti-oxidants, colorants, flavoring agents, preservatives and taste-masking agents.
Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant.
Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may altematively be wet-, dry-, or melt-granulated, melt-congealed, or extruded before tabletting. The final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated.
The formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets, Vol.
1, by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
Consumable oral films for human or veterinary use are typically pliable water-soluble or water-swellable thin film dosage forms which may be rapidly dissolving or mucoadhesive and typically comprise a compound of Formula I, a film-forming polymer, a binder, a solvent, a humectant, a plasticizer, a stabilizer or emulsifier, a viscosity-modifying agent and a solvent. Some components of the formulation may perform more than one function.
The compound of Formula I (or pharmaceutically acceptable salts thereof or /V-oxides thereof) may be water-soluble or insoluble. A water-soluble compound typically comprises from 1 weight % to 80 weight %, more typically from 20 weight % to 50 weight %, of the solutés. Less soluble compounds may comprise a smaller proportion of the composition, typically up to 30 weight % of the solutés. Altematively, the compound of Formula I may be in the form of multiparticulate beads.
The film-forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocolloids and is typically présent in the range 0.01 to 99 weight %, more typically in the range 30 to 80 weight %.
Other possible ingrédients include anti-oxidants, colorants, flavorings and flavor enhancers, preservatives, salivary stimulating agents, cooling agents, co-solvents (including oils), émollients, bulking agents, anti-foaming agents, surfactants and taste-masking agents.
Films in accordance with the invention are typically prepared by evaporative drying of thin aqueous films coated onto a peelable backing support or paper. This may be done in a drying oven or tunnel, typically a combined coater dryer, or by freeze-drying or vacuuming.
Solid formulations for oral administration may be formulated to be immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic and coated particles are to be found in Verma et al., Pharmaceutical Technology On-line, 25(2), 1-14 (2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may also be administered directly into the blood stream, into muscle, or into an internai organ. Suitable means for parentéral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous. Suitable devices for parentéral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
Parentéral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (for example to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a stérile non-aqueous solution or
as a dried form to be used in conjunction with a suitable vehicle such as stérile, pyrogen-free water.
The préparation of parentéral formulations under stérile conditions, for example, by lyophilization, may readily be accomplished using standard pharmaceutical techniques well 5 known to those skilled in the art.
The solubility of compounds of Formula I (including pharmaceutically acceptable salts thereof) used in the préparation of parentéral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents.
Formulations for parentéral administration may be formulated to be immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus compounds of the invention may be formulated as a suspension or as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and semi-solids and suspensions comprising drug15 loaded poly(DL-lactic-coglycolic acid) (PLGA) microspheres.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may also be administered topically, (intra)dermally, or transdermally to the skin or mucosa. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, 20 fibers, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, minerai oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Pénétration enhancers may be incorporated. See e.g., Finnin and Morgan, J. Pharm. Sci. 1999, 88, 955-958.
Other means of topical administration include delivery by electroporation, iontophoresis, 25 phonophoresis, sonophoresis and microneedle or needle-free (e.g., Powderject™, Bioject™, etc.) injection.
Formulations for topical administration may be formulated to be immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention (including pharmaceutically acceptable salts thereof) can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone; as a mixture, for example, in a dry blend with lactose; or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler, as an aérosol spray from a pressurized container, pump, spray, atomizer (for example an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3heptafluoropropane, or as nasal drops. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
The pressurized container, pump, spray, atomizer, or nebulizer contains a solution or suspension of the compound(s) of the invention comprising, for example, éthanol, aqueous éthanol, or a suitable alternative agent for dispersing, solubihzing, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
Prior to use in a dry powder or suspension formulation, the drug product is micronized to a size suitable for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
Capsules (made, for example, from gelatin or hydroxypropyl methyl cellulose), blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as L-leucine, mannitol, or magnésium stéarate. The lactose may be anhydrous or in the form of the monohydrate. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
A suitable solution formulation for use in an atomizer using electrohydrodynamics to produce a fine mist may contain from 1 pg to 20 mg of the compound of the invention per actuation and the actuation volume may vary from 1 pL to 100 pL. A typical formulation may comprise a compound of Formula I or a pharmaceutically acceptable sait thereof, propylene glycol, stérile water, éthanol and sodium chloride. Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
Suitable flavors, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those formulations of the invention intended for inhaled/intranasal administration.
Formulations for inhaled/intranasal administration may be formulated to be immédiate and/or modified release using, for example, PGLA. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
In the case of dry powder inhalers and aérosols, the dosage unit is determined by means of a valve which delivers a metered amount. Units in accordance with the invention are typically arranged to administer a metered dose or “puff” containing from 0.01 to 100 mg of the compound of Formula I. The overall daily dose will typically be in the range 1 pg to 200 mg, which may be administered in a single dose or, more usually, as divided doses throughout the day.
The compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
Formulations for rectal/vaginal administration may be formulated to be immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may also be administered directly to the eye or ear, typically in the form of drops of a micronized suspension or solution in isotonie, pH-adjusted, stérile saline. Other formulations suitable for ocular and aurai administration include ointments, gels, biodégradable (e.g., absorbable gel sponges, collagen) and non-biodegradable (e.g., silicone) implants, wafers, lenses and particulate or vesicular Systems, such as niosomes or liposomes. A polymer such as crossedlinked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropyl methyl cellulose, hydroxyethyl cellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
Formulations for ocular/aural administration may be formulated to be immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted, or programmed release.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may be combined with soluble macromolecular entities, such as cyclodextrin and suitable dérivatives thereof or polyethylene glycol-containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration.
Drug-cyclodextrin complexes, for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion complexes may be used. As an alternative to direct complexation with the drug, the cyclodextrin may be used as an auxiliary additive, i.e., as a carrier, diluent, or solubilizer. Most commonly used for these purposes are alpha-, beta- and gamma-cyclodextrins, examples of which may be found in International Patent Applications Nos. WO 91/11172, WO 94/02518 and WO 98/55148.
Since the présent invention has an aspect that relates to the treatment of the disease/conditions described herein with a combination of active ingrédients which may be administered separately, the invention also relates to combining separate pharmaceutical compositions in kit form. The kit comprises two separate pharmaceutical compositions: a compound of Formula I a prodrug thereof or a sait of such compound or prodrug and a second compound as described above. The kit comprises means for containing the separate compositions such as a container, a divided bottle or a divided foil packet. Typically the kit comprises directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are for example administered in different dosage forms (e.g., oral and parentéral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
An example of such a kit is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses hâve the size and shape of the tablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a resuit, the tablets or capsules are sealed in the recesses between the plastic foil and the sheet. In some embodiments, the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
It may be désirable to provide a memory aid on the kit, e.g., in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested. Another example of such a memory aid is a calendar printed on the card, e.g., as follows First Week, Monday, Tuesday, etc.... Second Week, Monday, Tuesday,... etc. Other variations of memory aids will be readily apparent. A daily dose can be a single tablet or capsule or several pills or capsules to be taken on a given day. Also, a daily dose of Formula I compound can consist of one tablet or capsule while a daily dose of the second compound can consist of several tablets or capsules and vice versa. The memory aid should reflect this.
In another spécifie embodiment of the invention, a dispenser designed to dispense the daily doses one at a time in the order of their intended use is provided. For example, the dispenser is equipped with a memory aid, so as to further facilitate compliance with the regimen. An example of such a memory aid is a mechanical counter which indicates the number of daily doses that has been dispensed. Another example of such a memory aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
The invention will be described in greater detail by way of spécifie examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters that can be changed or modified to yield essentially the same results. Additional compounds within the scope of this invention may be prepared using the methods illustrated in these Examples, either alone or in combination with techniques generally known in the art. In the following Examples and Préparations, “DMSO means dimethyl sulfoxide, “N” where referring to concentration means Normal, “M” means molar, “mL” means milliliter, “mmol” means millimoles, “umol” means micromoles, “eq.” means équivalent, “°C” means degrees Celsius, “MHz” means mégahertz, HPLC means high-performance liquid chromatography.
EXAMPLES
Experiments were generally carried out under inert atmosphère (nitrogen or argon), particularly in cases where oxygen- or moisture-sensitive reagents or intermediates were employed. Commercial solvents and reagents were generally used without further purification. Anhydrous solvents were employed where appropriate, generally AcroSeal® products from Acros Organics or DriSolv® products from EMD Chemicals. In other cases, commercial solvents were passed through columns packed with 4Â molecular sieves, until the following QC standards for water were attained: a) <100 ppm for dichloromethane, toluene, N,Ndimethylformamide and tetrahydrofuran; b) <180 ppm for methanol, éthanol, 1,4-dioxane and diisopropylamine. For very sensitive reactions, solvents were further treated with metallic sodium, calcium hydride or molecular sieves, and distilled just prior to use. Products were generally dried under vacuum before being carried on to further reactions or submitted for biological testing. Mass spectrometry data is reported from either liquid chromatography-mass spectrometry (LCMS), atmospheric pressure chemical ionization (APCI) or gas chromatography-mass spectrometry (GCMS) instrumentation. Chemical shifts for nuclear magnetic résonance (NMR) data are expressed in parts per million (ppm, δ) referenced to residual peaks from the deuterated solvents employed. In some examples, chiral séparations were carried out to separate atropisomers (or atropenantiomers) of certain compounds of the invention. In some examples, the optical rotation of an atropisomer was measured using a polarimeter. According to its observed rotation data (or its spécifie rotation data), an atropisomer (or atropenantiomer) with a clockwise rotation was designated as the (+)-atropisomer [or the (+) atropenantiomer] and an atropisomer (or atropenantiomer) with a counter-clockwise rotation was designated as the (-)-atropisomer [or the (-) atropenantiomer].
Reactions proceeding through détectable intermediates were generally followed by LCMS, and allowed to proceed to full conversion prior to addition of subséquent reagents. For synthèses referencing procedures in other Examples or Methods, reaction conditions (reaction time and température) may vary. In general, reactions were followed by thin-layer chromatography or mass spectrometry, and subjected to work-up when appropriate. Purifications may vary between experiments: in general, solvents and the solvent ratios used for eluents/gradients were chosen to provide appropriate Rfs or rétention times.
Examples 1 and 2 (+)-6-{4-[(3-Cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1H,3H)-dione (1) and (-)-6-{4-[(3-Cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpyrimidine-2,4( 1iï,3iï)-dione (2)
Step 1. Synthesis of 6-amino-1,5-dimethylpyrimidine-2,4(1H,3H)-dione, hydrochloride sait (C1).
A solution of sodium methoxide in methanol (4.4 M, 27 mL, 119 mmol) was added to a solution of ethyl 2-cyanopropanoate (95%, 13.2 mL, 99.6 mmol) and 1-methylurea (98%, 8.26 g, 109 mmol) in methanol (75 mL), and the reaction mixture was heated at reflux for hours, then cooled to room température. After removal of solvent in vacuo, the residue was repeatedly evaporated under reduced pressure with acetonitrile (3 x 50 mL), then partitioned between acetonitrile (100 mL) and water (100 mL). Aqueous 6 M hydrochloric acid was slowly added until the pH had reached approximately 2; the resulting mixture was stirred for 1 hour.
The precipitate was collected via filtration and washed with tert-butyl methyl ether, affording the product as a white solid. Yield: 15.2 g, 79.3 mmol, 80%. LCMS m/z 156.1 [M+H]+. 1H NMR (400
MHz, DMSO-d6) δ 10.38 (brs, 1H), 6.39 (s, 2H), 3.22 (s, 3H), 1.67 (s, 3H).
Step 2. Synthesis of 6-bromo-1,5-dimethylpyrimidine-2,4(1H,3H)-dione (C2).
A 1:1 mixture of acetonitrile and water (120 mL) was added to a mixture of C1 (9.50 g, 49.6 mmol), sodium nitrite (5.24 g, 76 mmol), and copper(ll) bromide (22.4 g, 100 mmol) {Caution: bubbling and slight exotherm!}, and the reaction mixture was allowed to stir at room température for 66 hours. Addition of aqueous sulfuric acid (1 N, 200 mL) and ethyl acetate (100 mL) provided a precipitate, which was collected via filtration and washed with water and ethyl acetate to afford the product as a light yellow solid (7.70 g). The organic layer of the filtrate was concentrated to a smaller volume, during which additional precipitate formed; this was isolated via filtration and washed with 1:1 ethyl acetate / heptane to provide additional product (0.4 g). Total yield: 8.1 g, 37 mmol, 75%. GCMS m/z218, 220 [M+]. 1H NMR (400 MHz, DMSO-d6) δ 11.58 (br s, 1 H), 3.45 (s, 3H), 1.93 (s, 3H).
Step 3. Synthesis of 6-bromo-1,5-dimethyl-3-{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine2,4(1H,3H)-dione (C3).
To a mixture of C2 (21.9 g, 99.8 mmol) and 2-(trimethylsilyl)ethoxymethyl chloride (20 g, 120 mmol) in acetonitrile (400 mL) was added 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU, 18.3 g, 120 mmol), and the reaction mixture was stirred at 60 °C for 18 hours. Additional 2(trimethylsilyl)ethoxymethyl chloride (5 g, 30 mmol) and 1,8-diazabicyclo[5.4.0]undec-7-ene (4.6 g, 30 mmol) were added, and stirring was continued at 60 °C for 18 hours. After the mixture had been concentrated in vacuo, the residue was diluted with water (500 mL) and extracted with ethyl acetate (3 x 300 mL). The combined organic layers were concentrated; purification using chromatography on silica gel (Gradient: 20% to 50% ethyl acetate in petroleum ether) afforded the product as a colorless oil. Yield: 22.5 g, 64.4 mmol, 64%. 1H NMR (400 MHz, CDCI3) δ 5.41 (s, 2H), 3.61-3.72 (m, 5H), 2.13 (s, 3H), 0.93-1.02 (m, 2H), 0.00 (s, 9H).
Step 4. Synthesis of 6-[4-(benzyloxy)-2-methylphenyl]-1,5-dimethyl-3-{[2(trimethylsilyllethoxyJmethyllpyrimidine-EfifW^I-dione (C4).
To a mixture of C3 (10 g, 29 mmol), [4-(benzyloxy)-2-methylphenyl]boronic acid (10.4 g, 43.0 mmol) and césium carbonate (28 g, 86 mmol) in 1,4-dioxane (400 mL) was added [1,1’bis(diphenylphosphino)ferrocene]dichloropalladium(ll) (2.2 g, 3.0 mmol). The reaction mixture was heated at reflux for 4 hours, then filtered. The filtrate was concentrated, and the residue was purified by silica gel chromatography (Gradient: 10% to 20% ethyl acetate in petroleum ether) to provide the product as a light yellow solid. Yield: 10 g, 21 mmol, 72%. 1H NMR (400 MHz, CDCI3) δ 7.34-7.49 (m, 5H), 7.00 (d, half of AB quartet, J=Q.3 Hz, 1H), 6.91-6.97 (m, 2H), 5.50 (AB quartet, JAB=9.2 Hz, ΔνΑΒ=4.1 Hz, 2H), 5.10 (s, 2H), 3.73-3.79 (m, 2H), 3.03 (s, 3H), 2.15 (s, 3H), 1.65 (s, 3H), 1.00-1.06 (m, 2H), 0.03 (s, 9H).
Step 5. Synthesis of 6-(4-hydroxy-2-methylphenyl)-1,5-dimethyl-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(7H, SH)-dione (C5).
A mixture of C4 (10 g, 21 mmol) and palladium hydroxide (2 g, dry) in methanol (300 mL) was stirred at room température for 24 hours under 40 psi of hydrogen. After filtration of the reaction mixture, the filtrate was concentrated to provide the product as a light yellow solid.
Yield: 8.0 g, 21 mmol, 100%. Ή NMR (400 MHz, CDCI3) δ 6.92 (d, half of AB quartet, J=8.2 Hz,
1 H), 6.81-6.87 (m, 2H), 5.52 (AB quartet, JAB=9.5 Hz, Avab=2.7 Hz, 2H), 3.73-3.80 (m, 2H), 3.03 (s, 3H), 2.11 (s, 3H), 1.65 (s, 3H), 0.99-1.05 (m, 2H), 0.01 (s, 9H).
Step 6. Synthesis of 2-chloro-3-cyclopropylpyridine (C6).
To a mixture of 2-chloro-3-iodopyridine (2.39 g, 9.98 mmol), cyclopropylboronic acid (860 mg, 10 mmol) and potassium carbonate (4.14 g, 30.0 mmol) in 1,4-dioxane (50 mL) was added tetrakis(trïphenylphosphine)palladium(0) (1.16 g, 1.00 mmol). The reaction mixture was stirred at 120 °C for 4 hours, then diluted with ethyl acetate (50 mL) and filtered. The filtrate was concentrated and the residue was purified by silica gel chromatography (Gradient: 10% to 30% ethyl acetate in petroleum ether) to afford the product as a colorless oil. Yield: 1 g, 6 mmol, 60%. Ή NMR (400 MHz, CDCI3) δ 8.20 (dd, J=4.7, 1.8 Hz, 1H), 7.24-7.28 (m, 1H), 7.14 (brdd,
J=7.6, 4.8 Hz, 1H), 2.12-2.21 (m, 1H), 1.04-1.11 (m, 2H), 0.67-0.72 (m, 2H).
Step 7. Synthesis of 6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethyl-
3-{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,3H)-dione (C7).
Palladium(ll) acetate (61 mg, 0.27 mmol) and di-tert-butyl[3,4,5,6-tetramethyl-2',4',6'tri(propan-2-yl)biphenyl-2-yl]phosphane (130 mg, 0.27 mmol) were added to a mixture of C6 (615 mg, 4.00 mmol), C5 (1.0 g, 2.6 mmol) and césium carbonate (2.6 g, 8.0 mmol) in 1,4dioxane (25 mL). The reaction mixture was stirred at 120 °C under microwave irradiation for 5 hours, then diluted with ethyl acetate (50 mL) and filtered. After removal of solvents in vacuo, the residue was purified via silica gel chromatography (Gradient: 0% to 25% ethyl acetate in petroleum ether) to provide the product as a yellow gum. Yield: 900 mg, 1.8 mmol, 69%. LCMS m/z 494.1 [M+H]+. Ή NMR (400 MHz, CDCI3) δ 8.02 (dd, J=4.8, 1.8 Hz, 1H), 7.30 (dd, J=7.4,
1.8 Hz, 1H), 7.11-7.14 (m, 1H), 7.08-7.10 (m, 2H), 7.01 (dd, J=7.5, 4.8 Hz, 1H), 5.51 (AB quartet, JAB=9.3 Hz, Avab=3.8 Hz, 2H), 3.74-3.80 (m, 2H), 3.08 (s, 3H), 2.18 (s, 3H), 2.16-2.24 (m, 1H), 1.70 (s, 3H), 1.00-1.06 (m, 4H), 0.74-0.79 (m, 2H), 0.03 (s, 9H).
Step 8. Synthesis of (+)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,530 dimethylpyrimidine-2,4( 1H,3H)-dione (1) and (-)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2methylphenyl}-1,5-dimethylpyrimidine-2,4(7H,3H)-dione (2).
Trifluoroacetic acid (1.5 mL) was added to a solution of C7 (875 mg, 1.77 mmol) in dichloromethane (8 mL). The reaction mixture was stirred at room température for 2 hours and concentrated in vacuo-, the residue was dissolved in methanol (10 mL), treated with potassium carbonate (1.22 g, 8.83 mmol) and stirred at room température for 18 hours. After removal of solids via filtration, the filtrate was concentrated under reduced pressure and partitioned between ethyl acetate and water. The aqueous layer was extracted three times with ethyl acetate, and the combined organic layers were washed sequentially with water and with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Gradient: 0% to 100% ethyl acetate m heptane) afforded a mixture of 1 and 2, which was separated via reversed phase chiral chromatography (Column: Chiral Technologies, Chiralpak IA; Gradient: heptane in éthanol). The first-eluting atropenantiomer, obtained as a solid that exhibited a positive (+) rotation, was designated as Example 1. Yield: 210 mg, 0.578 mmol, 33%. The second-eluting atropenantiomer, also obtained as a solid but with a négative (-) rotation, was designated as Example 2. Yield: 190 mg, 0.523 mmol, 30%. 1: LCMS m/z 364.2 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 7.94 (br d, J=5 Hz, 1H), 7.48 (br d, J=7.6 Hz, 1H), 7.22 (d, J=8.2 Hz, 1H), 7.03-7.14 (m, 3H), 3.04 (s, 3H), 2.20 (s, 3H), 2.15-2.23 (m, 1H), 1.63 (s, 3H), 0.99-1.06 (m, 2H), 0.75-0.82 (m, 2H). 2: LCMS m/z364.2 [M+Hf. 1H NMR (400 MHz, CD3OD) δ 7.94 (dd, J=4.8, 1.7 Hz, 1H), 7.48 (dd, J=7.5, 1.8 Hz, 1H), 7.22 (d, 4=8.3 Hz, 1H), 7.09-7.14 (m, 2H), 7.06 (dd, J=8.4, 2.3 Hz, 1H), 3.04 (s, 3H), 2.20 (s, 3H), 2.15-2.23 (m, 1H), 1.63 (s, 3H), 0.99-1.06 (m, 2H), 0.75-0.82 (m, 2H).
Examples 3 and 4 (-)-6-{4-[(3-Chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1\-\,3H)-dione (3) and (+)-6-{4-[(3-Chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpynmidine-2,4( 1H,3iï)-dione (4)
HO'
Step 1. Synthesis of 6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethyl-3-{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,3H)-dione (C8).
Césium carbonate (476 mg, 1.46 mmol) was added to a mixture of 3-chloro-2,5difluoropyridine (97%, 150 mg, 0.97 mmol) and C5 (366 mg, 0.972 mmol) in dimethyl sulfoxide (5 mL), and the reaction mixture was stirred at 80 °C for 6 hours. Water was added, and the mixture was extracted three times with ethyl acetate; the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 10% to 40% ethyl acetate in heptane) provided the product as a sticky solid. Yield: 414 mg, 0.818 mmol, 84%. H NMR (400 MHz, CDCIs) δ 7.96 (d, J=2.7 Hz, 1H), 7.64 (dd, J=7.1,2.7 Hz, 1H), 7.09-7.15 (m, 3H), 5.51 (AB quartet, JAB=9.3 Hz, ΔνΑΒ=3.4 Hz, 2H), 3.74-3.80 (m, 2H), 3.07 (s, 3H), 2.19 (s, 3H), 1.69 (s, 3H), 1.00-1.06 (m, 2H), 0.03 (s, 9H).
Step 2. Synthesis of (-)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpyrimidine-2,4( 1\-ï,3H)-dione (3) and (+)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2methylphenyl}-1,5-dimethylpyrimidine-2,4( 1iï,3H)-dione (4).
Trifluoroacetic acid (812 pl_, 10.9 mmol) was added to a solution of C8 (187 mg, 0.370 mmol) in dichloromethane (3.0 mL), and the reaction mixture was stirred at room température for 1 hour. Solvents were removed in vacuo, and the residue was taken up in tetrahydrofuran (4.5 mL) and treated with concentrated aqueous ammonium hydroxide (9 mL). After 4 hours, the reaction mixture was concentrated under reduced pressure, combined with the crude product from an identical reaction carried out on C8 (200 mg, 0.395 mmol), and purified via chromatography on silica gel (Gradient: 20% to 40% ethyl acetate in heptane), to provide the racemic product as a white solid. Yield: 219 mg, 0.583 mmol, 76%. This was separated into its atropenantiomers via chiral chromatography (Column: Phenomenex Lux Cellulose-1; Gradient: 50% to 100% éthanol in heptane). The first-eluting atropenantiomer, which was obtained as a white solid, exhibited a négative (-) rotation and was designated as Example 3. Yield: 25 mg, 66 pmol, 9%. The second-eluting atropenantiomer was also a white solid, but exhibited a positive (+) rotation; this was designated as Example 4. Yield: 62 mg, 160 pmol, 21%. 3: LCMS m/z 376.1, 378.0 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.35 (br s, 1H), 7.97 (d, J=2.7 Hz, 1H), 7.64 (dd, J=7.1,2.8 Hz, 1H), 7.11-7.16 (m, 3H), 3.04 (s, 3H), 2.20 (s, 3H), 1.67 (s, 3H). 4: LCMS m/z376.2, 378.2 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.46 (br s, 1H), 7.97 (d, J=2.7 Hz, 1H), 7.64 (dd, J=7.1, 2.7 Hz, 1H), 7.12-7.16 (m, 3H), 3.04 (s, 3H), 2.20 (brs, 3H), 1.67 (s, 3H).
Example 5 6-{4-[(3-Chloropyridin-2-yl)oxy]-2-methylphenyl}-1-ethyl-5-methylpyrimidine-2,4(1ï-\,3H)dione (5)
Ο ο
Step 1. Synthesis of 6-amino-1-ethyl-5-methylpyrimidine-2,4(1iï,3iï)-dione (C9).
Sodium hydride (1.84 g, 76.7 mmol) was added in portions to a solution of 1-ethylurea (5.7 g, 65 mmol) and ethyl 2-cyanopropanoate (7.5 g, 59 mmol) in methanol (60 mL) that had been cooled to 0 to 5 °C. The reaction mixture was stirred for 18 hours and then was concentrated in vacuo. Acetonitrile (200 mL) was added, and the mixture was again concentrated to dryness. The residue was diluted with a mixture of acetonitrile (100 mL) and water (30 mL); 12 M aqueous hydrochloric acid was added drop-wise until the pH was approximately 1 - 2. After the mixture had been stirred for 1 hour, the precipitate was collected via filtration and washed with tert-butyl methyl ether, affording the product as a white solid. Yield: 8.15 g, 48.2 mmol, 82%. 1H NMR (400 MHz, DMSO-rt6) δ 3.84 (q, J=6.9 Hz, 2H), 1.66 (s, 3H), 1.07 (t, J=7.0 Hz, 3H).
Step 2. Synthesis of 6-bromo-1 -ethyl-5-methylpyrimidine-2,4(114,3iï)-dione (C10).
To a solution of C9 (6.2 g, 36.6 mmol) in a 1:1 mixture of acetonitrile and water (70 mL) were added sodium nitrite (3.8 g, 55 mmol) and copper(ll) bromide (16.4 g, 73.4 mmol), and the reaction mixture was stirred for 18 hours at room température. A mixture of 1 N aqueous sulfuric acid (100 mL) and ethyl acetate (50 mL) was added, and stirring was continued for 1 hour, at which time the organic layer was separated, and the aqueous layer was extracted with dichloromethane (2 x 100 mL). The combined organic layers were concentrated in vacuo; silica gel chromatography (Gradient: 0% to 50% ethyl acetate in petroleum ether) provided the product as a green solid. Yield: 5.0 g, 21 mmol, 57%. Ή NMR (400 MHz, CDCI3) δ 8.87 (br s, 1H), 4.21 (q, J=7.0 Hz, 2H), 2.11 (s, 3H), 1.32 (t, J=7.Q Hz, 3H).
Step 3. Synthesis of 6-bromo-1-ethyl-5-methyl-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1H,3H)-dione (C11).
Compound C10 was converted to the product using the method described for synthesis of C3 in Examples 1 and 2. The product was obtained as a yellow gum. Yield: 1.28 g, 3.52 mmol, 17%. 1H NMR (400 MHz, CDCI3) δ 5.41 (s, 2H), 4.24 (q, J=7.1 Hz, 2H), 3.65-3.72 (m, 2H), 2.13 (s, 3H), 1.31 (t, J=7.1 Hz, 3H), 0.94-1.01 (m, 2H), 0.00 (s, 9H).
Step 4. Synthesis of 6-[4-(benzyloxy)-2-methylphenyl]-1 -ethyl-5-methyl-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,SH)-dione (C12).
Compound C11 was converted to the product using the method described for synthesis of C4 in Examples 1 and 2. The product was obtained as a yellow gum. Yield: 1.09 g, 2.27 mmol, 78%. 1H NMR (400 MHz, CDCI3) δ 7.34-7.49 (m, 5H), 7.05 (d, J=8.2 Hz, 1H), 6.91-6.97 (m, 2H), 5.50 (s, 2H), 5.10 (s, 2H), 3.79-3.89 (m, 1H), 3.74-3.80 (m, 2H), 3.23-3.34 (m, 1H),
2.15 (s, 3H), 1.62 (s, 3H), 1.00-1.07 (m, 5H), 0.03 (s, 9H).
Step 5. Synthesis of 1-ethyl-6-(4-hydroxy-2-methylphenyl)-5-methyl-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1H,3H)-dione (C13).
The product, obtained as a gray solid, was synthesized from C12 using the method described for synthesis of C5 in Examples 1 and 2. Yield: 800 mg, 2.05 mmol, 90%. 1H NMR (400 MHz, CDCI3) δ 6.99 (d, J=8.2 Hz, 1H), 6.79-6.85 (m, 2H), 5.51 (s, 2H), 3.79-3.89 (m, 1H), 3.73-3.80 (m, 2H), 3.24-3.34 (m, 1H), 2.12 (s, 3H), 1.62 (s, 3H), 0.99-1.06 (m, 5H), 0.02 (s, 9H).
Step 6. Synthesis of 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1-ethyl-5-methyl-3{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,3H)-dione (C14).
Césium carbonate (127 mg, 0.390 mmol) and C13 (50 mg, 0.13 mmol) were added to a solution of 2,3-dichloropyridine (38 mg, 0.26 mmol) in dimethyl sulfoxide (3 mL), and the reaction mixture was heated at 80 °C for 18 hours. After removal of solids via filtration, the filtrate was partitioned between ethyl acetate (20 mL) and water (20 mL), and the aqueous layer was extracted with ethyl acetate (2 x 20 mL). The combined organic layers were concentrated in vacuo and the residue was purified by préparative thin-layer chromatography on silica gel (Eluent: 3:1 petroleum ether / ethyl acetate) to afford the product as a yellow gum. Yield: 31 mg, 62 pmol, 48%. 1H NMR (400 MHz, CDCI3) δ 8.08 (dd, J=4.7, 1.4 Hz, 1H), 7.81 (dd, J=7.7, 1.4 Hz, 1H), 7.11-7.19 (m, 3H), 7.05 (dd, J=7.6, 4.9 Hz, 1H), 5.50 (s, 2H), 3.81-3.93 (m, 1H), 3.723.80 (m, 2H), 3.25-3.37 (m, 1H), 2.19 (s, 3H), 1.65 (s, 3H), 0.98-1.10 (m, 5H), 0.02 (s, 9H).
Step 7. Synthesis of 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1 -ethyl-3(hydroxymethyl)-5-methylpyrimidine-2,4( 1iï,3H)-dione (C15).
Compound C14 (31 mg, 62 pmol) was treated with trifluoroacetic acid (3 mL), and the reaction mixture was stirred at room température for 1 hour. Removal of solvent in vacuo provided the product (24.8 mg), which was used for the next step without further purification.
Step 8. Synthesis of 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1-ethyl-5methylpyrimidine-2,4( 1iï,3H)-dione (5).
To a solution of C15 (from the previous step, 24.8 mg, <62 pmol) in methanol (5 mL) was added potassium carbonate (83 mg, 0.60 mmol), and the reaction mixture was stirred at room température for 1 hour. After removal of solids via filtration, the filtrate was concentrated and the residue was purified by préparative thin-layer chromatography on silica gel (Eluent: 20:1 dichloromethane / methanol) to afford the product as a white solid. Yield: 7.7 mg, 21 pmol, 34% over two steps. LCMS m/z 372.0 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 8.07 (dd, J=4.8, 1.5 Hz, 1H), 7.98 (dd, J=7.8, 1.6 Hz, 1H), 7.32 (d, J=8.2 Hz, 1H), 7.12-7.21 (m, 3H), 3.78-3.89 (m, 1H), 3.27-3.38 (m, 1H, assumed; partially obscured by solvent peak), 2.21 (s, 3H), 1.60 (s, 3H), 1.07 (t, J=7.1 Hz, 3H).
Example 6
6-{4-[(3-Chloropyridin-2-yl)oxy]-2-methylphenyl}-5-ethyl-1 -methylpyrimidine-2,4(1H,3H)dione (6)
C16 C17
Step 1. Synthesis of 6-amino-5-ethyl-1-methylpyrimidine-2,4(1H,3iï)-dione (C16).
Ethyl 2-cyanobutanoate was reacted with 1-methylurea according to the method described for synthesis of C9 in Example 5. The product was obtained as a white solid. Yield:
5.95 g, 35.2 mmol, 66%. Ή NMR (400 MHz, DMSO-de) δ 10.36 (s, 1H), 6.41 (s, 2H), 3.22 (s, 3H), 2.22 (q, J=7.3 Hz, 2H), 0.87 (t, J=7.3 Hz, 3H).
Step 2. Synthesis of 6-bromo-5-ethyl-1-methylpyrimidine-2,4(1iï,3H)-dione (C17).
To a solution of C16 (5.95 g, 35.2 mmol) in a 1:1 mixture of acetonitrile and water (80 mL) were added sodium nitrite (3.6 g, 52 mmol) and copper(ll) bromide (15.7 g, 70.3 mmol), and the reaction mixture was stirred for 18 hours at room température. A mixture of 1 N aqueous sulfuric acid (100 mL) and ethyl acetate (50 mL) was added, and stirring was continued for 1 hour. The resulting solid was collected via filtration and the filter cake was washed with aqueous ethyl acetate, providing the product as a white solid (4 g). The organic layer of the filtrate was separated and the aqueous layer was extracted with dichloromethane (2 x 100 mL); the combined organic layers were concentrated in vacuo to afford additional product as a green solid (3 g). Yield: 7 g, 30 mmol, 85%. 1H NMR (400 MHz, CDCI3) δ 8.92 (br s, 1H), 3.62 (s, 3H), 2.58 (q, J=7.4 Hz, 2H), 1.09 (t, J=7.4 Hz, 3H).
Step 3. Synthesis of 6-bromo-5-ethyl-1-methyl-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(/H,3H)-dione (C18).
Compound C17 was converted to the product using the method described for synthesis of C3 in Examples 1 and 2. The product was obtained as a yellow gum. Yield: 3.1 g, 8.5 mmol, 28%. 1H NMR (400 MHz, CDCI3) δ 5.41 (s, 2H), 3.66 (s, 3H), 3.64-3.72 (m, 2H), 2.61 (q, J=7.4 Hz, 2H), 1.09 (t, J=7.4 Hz, 3H), 0.95-1.01 (m, 2H), 0.00 (s, 9H).
Step 4. Synthesis of 6-[4-(benzyloxy)-2-methylphenyl]-5-ethyl-1-methyl-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,3H)-dione (C19).
Compound C18 was converted to the product using the method employed for synthesis of C4 in Examples 1 and 2. The product was obtained as a yellow gum. Yield: 1.26 g, 2.62 mmol, 59%. 1H NMR (400 MHz, CDCI3) δ 7.34-7.49 (m, 5H), 7.03 (d, J=8.0 Hz, 1H), 6.91-6.97 (m, 2H), 5.47-5.54 (m, 2H), 5.10 (s, 2H), 3.73-3.80 (m, 2H), 3.00 (s, 3H), 2.18-2.29 (m, 1H),
2.16 (s, 3H), 1.86-1.97 (m, 1H), 0.99-1.07 (m, 2H), 0.91 (t, J=7.3 Hz, 3H), 0.03 (s, 9H).
Step 5. Synthesis of 5-ethyl-6-(4-hydroxy-2-methylphenyl)-1-methyl-3-{[2(trimethylsilyljethoxyJmethylIpyrimidine^AflH^ydione (C20).
The product, obtained as a gray solid, was synthesized from C19 using the method described for synthesis of C5 in Examples 1 and 2. Yield: 850 mg, 2.18 mmol, 83%. LCMS m/z
413.2 [M+Na+J. 1H NMR (400 MHz, CDCI3) δ 6.97 (d, J=7.9 Hz, 1H), 6.79-6.86 (m, 2H), 5.485.54 (m, 2H), 3.73-3.80 (m, 2H), 3.01 (s, 3H), 2.18-2.30 (m, 1H), 2.13 (s, 3H), 1.86-1.97 (m,
1H), 0.99-1.06 (m, 2H), 0.90 (t, J=7.3 Hz, 3H), 0.02 (s, 9H).
Step 6. Synthesis of 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-5-ethyl-1-methyl-3{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1l·V3l·^)-dione (C21).
A mixture of C20 (80 mg, 0.20 mmol), 2,3-dichloropyridine (45 mg, 0.30 mmol) and césium carbonate (199 mg, 0.611 mmol) in dimethyl sulfoxide (8 mL) was heated at 120 °C for hours. After addition of water and ethyl acetate, the mixture was extracted with ethyl acetate. The combined organic layers were dried, filtered, and concentrated under reduced pressure. Préparative thin-layer chromatography on silica gel (Eluent: 1:1 petroleum ether / ethyl acetate) afforded the product as a colorless oil. Yield: 82 mg, 0.16 mmol, 80%.
Step 7. Synthesis of 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-5-ethyl-1 methylpyrimidine-2,4( 1H,3H)-dione (6).
A solution of C21 (82 mg, 0.16 mmol) in trifluoroacetic acid (3 mL) was heated at 80 °C for 1 hour. After removal of solvent in vacuo, the residue was dissolved in methanol (5 mL), treated with potassium carbonate (68 mg, 0.49 mmol), and stirred at room température for 1 hour. The reaction mixture was filtered, and the filtrate was concentrated; purification via préparative thin-layer chromatography (Eluent: ethyl acetate) provided the product as a white solid. Yield: 28 mg, 75 pmol, 47%. LCMS m/z 372.0 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 8.07 (brd, J=4 Hz, 1H), 7.97 (d, J=7.5 Hz, 1H), 7.29 (d, J=8.3 Hz, 1H), 7.11-7.21 (m, 3H), 3.01 (s, 3H), 2.22 (s, 3H), 2.17-2.27 (m, 1H), 1.87-1.98 (m, 1H), 0.93 (t, J=7.3 Hz, 3H).
Examples 7 and 8 (-)-1,5-Dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2-yl]oxy}phenyl)pyrimidine2,4(1H,3ï-\)-dione (7) and (+)-1,5-Dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2yl]oxy}phenyl)pyrimidine-2,4(1iï,3H)-dione (8) i OH
Step 1. Synthesis of tert-butyl 4-bromo-3,5-dimethyl-2,6-dioxo-3,6-dihydropyrimidine1 (2iï)-carboxylate (C22).
Compound C2 (800 mg, 3.65 mmol), di-tert-butyl dicarbonate (99%, 966 mg, 4.38 mmol), triethylamine (0.62 mL, 4.4 mmol) and 4-(dimethylamino)pyridine (45 mg, 0.36 mmol) were combined in tetrahydrofuran (15 mL) and heated to 70 °C for 1 hour, then allowed to stir at room température for 18 hours. The reaction mixture was concentrated in vacuo, and the residue was purified via chromatography on silica gel (Gradient: 10% to 25% ethyl acetate in heptane) to provide the product as a white solid. Yield: 1.10 g, 3.45 mmol, 94%. 1H NMR (400
MHz, CDCI3) δ 3.64 (s, 3H), 2.12 (s, 3H), 1.61 (s, 9H).
Step 2. Synthesis of tert-butyl 4-[4-(benzyloxy)-2-methylphenyl]-3,5-dimethyl-2,6-dioxo3,6-dihydropyrimidine-1 (2H)-carboxyla te (C23).
A mixture of C22 (1.00 g, 3.13 mmol), [4-(benzyloxy)-2-methylphenyl]boronic acid (98%,
1.16 g, 4.68 mmol), chloro(2-dicyclohexylphosphino-2',6'-dimethoxy-1,1'-biphenyl)[2-(2- aminoethylphenyl)]palladium(ll) - tert-butyl methyl ether adduct (S-Phos precatalyst) (119 mg, 0.156 mmol), and césium carbonate (3.06 g, 9.39 mmol) in 2-methyltetrahydrofuran (10 mL) and water (3 mL) was heated at 50 °C for 66 hours. The reaction mixture was diluted with water and ethyl acetate, and then filtered to remove suspended solids. The filtrate was extracted several times with ethyl acetate, and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated in vacuo. The resulting solid was suspended in a 1:3 mixture of ethyl acetate and heptane, stirred for several minutes, and filtered, providing the product as a white solid. Yield: 970 mg, 2.22 mmol, 71%. LCMS m/z337.2 [(M-Boc)+H]+. 1H NMR (400 MHz, CDCI3) δ 7.34-7.48 (m, 5H), 6.91-7.01 (m, 3H), 5.10 (s, 2H), 3.01 (s, 3H), 2.16 (brs, 3H), 1.66 (s, 9H), 1.64 (s, 3H).
Step 3. Synthesis of 6-(4-hydroxy-2-methylphenyl)-1,5-dimethylpyrimidine-2,4(1iï,3\-\)dione (C24).
Compound C23 (250 mg, 0.573 mmol) was mixed with a 30% solution of hydrogen bromide in acetic acid (1 mL, 5 mmol) and allowed to stir for 18 hours at room température. After removal of acetic acid under reduced pressure, the residue was dissolved in a minimal quantity of éthanol and diluted with 4 M aqueous hydrochloric acid to provide a slightly cloudy mixture; this was evaporated to dryness, and the resulting solid was suspended in 4 N aqueous hydrochloric acid, stirred for several minutes, and filtered, affording the product as a yellow solid. Yield: 125 mg, 0.508 mmol, 89%. LCMS m/z 247.2 [M+H]+. 1H NMR (400 MHz, DMSO-de) δ 11.36 (brs, 1H), 9.71 (v brs, 1H), 6.99 (d, J=8.2 Hz, 1H), 6.76 (d, J=2.3 Hz, 1H), 6.72 (d, J=8.1, 2.3 Hz, 1H), 2.82 (s, 3H), 2.03 (s, 3H), 1.44 (s, 3H).
Step 4. Synthesis of 1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2yl]oxy}phenyl)pyrimidine-2,4( 1E\,3H)-dione (C25).
2-Chloro-3-(trifluoromethyl)pyridine (98%, 269 mg, 1.45 mmol), C24 (325 mg, 1.32 mmol) and césium carbonate (521 mg, 1.60 mmol) were combined in /V,A/-dimethylformamide (6 mL) and the resulting suspension was heated at 100 °C for 18 hours. After it had cooled to room température, the reaction mixture was diluted with aqueous 1 M hydrochloric acid and extracted several times with ethyl acetate. The combined organic layers were washed twice with water and once with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated in vacuo. The resulting solid was suspended in a 1:1 mixture of ethyl acetate and heptane, stirred for several minutes and collected by filtration, providing the product as a white solid. Yield: 440 mg, 1.12 mmol, 85%. LCMS m/z 392.2 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.31-8.36 (m, 2H), 8.05 (br d, J=7.5 Hz, 1H), 7.13-7.22 (m, 4H), 3.06 (s, 3H), 2.21 (s, 3H), 1.69 (s, 3H).
Step 5. Isolation of (-)-1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2^]οχγ}ρΐΊβηγΙ)ργηηΊίάΐηβ-2,4(1Μ,3Η)^ίοηβ (7) and (+)-1,5-dimethyl-6-(2-methyl-4-{[3(trifluoromethyl)pyridin-2-yl]oxy}phenyl)pyrimidine-2,4(1H,3H)-dione (8).
Racemate C25 (1.30 g, 3.32 mmol) was separated into its atropenantiomers via chiral chromatography (Column: Phenomenex Lux Cellulose-2; Gradient: heptane / éthanol). The firsteluting atropenantiomer, obtained as a tan solid that exhibited a négative (-) rotation, was designated as Example 7. Yield: 536 mg, 1.37 mmol, 41%. The second-eluting atropenantiomer, also obtained as a tan solid but with a positive (+) rotation, was designated as Example 8. Yield: 553 mg, 1.41 mmol, 42%. 7: LCMS m/z 392.2 [M+H]+. Ή NMR (400 MHz, CDCI3) δ 8.34 (ddq, J=4.9, 1.9, 0.6 Hz, 1H), 8.30 (br s, 1H), 8.05 (ddq, J=7.6,1.9, 0.7 Hz, 1H),
7.13- 7.21 (m, 4H), 3.06 (s, 3H), 2.21 (br s, 3H), 1.69 (s, 3H). 8: LCMS m/z 392.2 [M+H]+. Ή NMR (400 MHz, CDCI3) δ 8.34 (br d, J=4.9 Hz, 1 H), 8.30 (br s, 1 H), 8.05 (br d, J=7.5 Hz, 1 H),
7.13- 7.22 (m, 4H), 3.06 (s, 3H), 2.21 (br s, 3H), 1.69 (s, 3H).
Examples 9 and 10 (+)-6-{4-[(3-Chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1iï,3H)-dione (9) and (-)-6-{4-[(3-Chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpyrimidine-2,4( 1H,^r\)-dione (10)
Step 1. Synthesis of 6-bromo-3-(3,4-dimethoxybenzyl)-1,5-dimethylpyrimidine5 2,4(1H,3H)-dione (C26).
1,8-Diazabicyclo[5.4.0]undec-7-ene (98%, 5.57 mL, 36.5 mmol) was added to a suspension of C2 (4.00 g, 18.3 mmol) and 4-(chloromethyl)-1,2-dimethoxybenzene (5.16 g, 27.6 mmol) in acetonitrile (80 mL), and the reaction mixture was heated at 60 °C for 18 hours. After removal of solvent in vacuo, the residue was purified via silica gel chromatography (Gradient:
25% to 50% ethyl acetate in heptane) to afford the product as a white solid. Yield: 5.70 g, 15.4 mmol, 84%. Ή NMR (400 MHz, CDCI3) δ 7.08-7.12 (m, 2H), 6.80 (d, J=8.0 Hz, 1H), 5.07 (s,
2H), 3.88 (s, 3H), 3.85 (s, 3H), 3.65 (s, 3H), 2.14 (s, 3H).
Step 2. Synthesis of 3-(3,4-dimethoxybenzyl)-6-(4-hydroxy-2-methylphenyl)-1,5dimethylpyrimidine-2,4( 1H,3H)-dione (C27).
An aqueous solution of potassium carbonate (3.0 M, 14 mL, 42 mmol) was added to a mixture of C26 (5.00 g, 13.5 mmol), (4-hydroxy-2-methylphenyl)boronic acid (4.12 g,
27.1 mmol), [1,1’-bis(diphenylphosphino)ferrocene]dichloropalladium(ll), dichloromethane complex (98%, 1.13 g, 1.36 mmol) and 1,4-dioxane (120 mL). After the reaction mixture had been heated at 100 °C for 18 hours, it was cooled to room température, diluted with ethyl acetate and water, and filtered through diatomaceous earth. The organic layer from the filtrate
was washed sequentially with saturated aqueous sodium bicarbonate solution and with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated in vacuo. Purification using silica gel chromatography (Gradient: 25% to 75% ethyl acetate in heptane) afforded the product as a white foam. Yield: 2.71 g, 6.84 mmol, 51%. LCMS m/z397.2 [M+H]+. Ή NMR (400 MHz, CDCI3) δ 7.22 (d, J=2.0 Hz, 1H), 7.19 (dd, J=8.1,2.0 Hz,
1H), 6.93 (d, J=8.2 Hz, 1H), 6.83 (d, J=8.3 Hz, 1H), 6.80-6.82 (m, 1H), 6.76-6.80 (m, 1H), 5.16 (AB quartet, JAB=13.3 Hz, ΔνΑΒ=19·2 Hz, 2H), 3.91 (s, 3H), 3.87 (s, 3H), 3.02 (s, 3H), 2.11 (br s, 3H), 1.66 (s, 3H).
Step 3. Synthesis of 6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-3-(3,410 dimethoxybenzyl)-1,5-dimethylpyrimidine-2,4( 1H,3H)-dione (C28).
A mixture of 2,3-dichloro-5-methylpyridine (735 mg, 4.54 mmol), C27 (1.5 g, 3.8 mmol) and césium carbonate (2.46 g, 7.55 mmol) in dimethyl sulfoxide (36 mL) was stirred at 100 °C for 40 hours, and at 120 °C for a further 48 hours. The reaction mixture was diluted with water (300 mL) and extracted with ethyl acetate (3 x 200 mL); the combined organic layers were dried, 15 filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 10% to 60% ethyl acetate in petroleum ether) provided the product as a yellow solid. Yield: 1.7 g, 3.2 mmol, 84%. Ή NMR (400 MHz, CDCI3) δ 7.89-7.92 (m, 1H), 7.64-7.66 (m, 1H), 7.23 (brd, J=1.9 Hz, 1H), 7.20 (brdd, J=8.2, 1.9 Hz, 1H), 7.10-7.12 (br s, 1H), 7.06-7.09 (m, 2H), 6.83 (d, J=8.2 Hz, 1H),
5.16 (AB quartet, JAB=13.4 Hz, ΔνΑΒ=20.4 Hz, 2H), 3.91 (s, 3H), 3.87 (s, 3H), 3.06 (s, 3H), 2.32 20 (s, 3H), 2.16 (S, 3H), 1.68 (S, 3H).
Step 4. Synthesis of 6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpyrimidine-2,4( 1M,3H)-dione (C29).
This experiment was carried out in three batches. A mixture of C28 (600 mg, 1.15 mmol) and methoxybenzene (622 mg, 5.75 mmol) in trifluoroacetic acid (20 mL) was stirred at 120 °C 25 for 48 hours, then at 125 °C for another 48 hours. The three batches were combined, concentrated under reduced pressure, and purified via chromatography on silica gel (Gradient: 10% to 70% ethyl acetate in petroleum ether). The product was obtained as a light brown solid. Yield: 690 mg, 1.86 mmol, 54%. LCMS m/z 371.8, 373.9 [M+H]+. Ή NMR (400 MHz, CD3OD) δ 7.90-7.92 (m, 1H), 7.82-7.84 (m, 1H), 7.23 (d, J=8.4 Hz, 1H), 7.14 (br d, J=2.2 Hz, 1H), 7.08 (br 30 dd, J=8.2, 2.2 Hz, 1 H), 3.03 (s, 3H), 2.33 (br s, 3H), 2.20 (br s, 3H), 1.62 (s, 3H).
Step 5. Isolation of (+)-6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpyrimidine-2,4( 1H,3H)-dione (9) and (-)-6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2methylphenyl}-1,5-dimethylpyrimidine-2,4(ïH,3H)-dione (10).
Compound C29 (690 mg, 1.86 mmol) was separated into its atropenantiomers via supercritical fluid chromatography (Column: Chiral Technologies, Chiralcel OJ-H, 5 pm; Eluent: 7:3 carbon dioxide / methanol). The first-eluting atropenantiomer, obtained as a solid that exhibited a positive (+) rotation, was designated as Example 9. Yield: 240 mg, 0.645 mmol, 35%. The second-eluting atropenantiomer, also obtained as a solid but with a négative (-) rotation, was designated as Example 10. Yield: 250 mg, 0.672 mmol, 36%. 9: LCMS m/z 372.1,
374.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.36 (br s, 1H), 7.91-7.93 (m, 1H), 7.65-7.66 (m, 1H), 7.13-7.14 (m, 1H), 7.10-7.11 (m, 2H), 3.04 (s, 3H), 2.32-2.34 (m, 3H), 2.18-2.19 (m, 3H), 1.67 (s, 3H). 10: LCMS m/z 372.1,374.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.35 (br s, 1H), 7.91-7.93 (m, 1H), 7.65-7.66 (m, 1H), 7.13-7.14 (m, 1H), 7.10-7.11 (m, 2H), 3.04 (s, 3H), 2.33 (dd, J=0.7, 0.7 Hz, 3H), 2.19 (d, J=0.6 Hz, 3H), 1.67 (s, 3H).
Example 11
6-{4-[(3-Chloro-4-methylpyridin-2-yl)oxy]phenyl}-1!5-dimethylpyrimidine-2,4(1F\,3H)dione (11)
Step 1. Synthesis of tert-butyl 4-[4-(benzyloxy)phenyl]-3,5-dimethyl-2,6-dioxo-3,6dihydropyrimidine-1 (2H)-carboxylate (C30).
A solution of C22 (23.3 g, 73.0 mmol), [4-(benzyloxy)phenyl]boronic acid (25 g, 110 mmol), [1,r-bis(diphenylphosphino)ferrocene]dichloropalladium(ll) (2.68 g, 3.66 mmol), and césium carbonate (95.2 g, 292 mmol) in 2-methyltetrahydrofuran (360 mL) and water (120 mL) was purged with nitrogen and heated to 50 °C for 5 hours. After cooling to room température, the reaction mixture was stirred at room température for 18 hours, then diluted with water and ethyl acetate. The mixture was filtered, and the filtrate was extracted several times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated in vacuo. The resulting solid was combined with the solid collected from the initial filtration, and extracted several times with hot ethyl acetate; the combined ethyl acetate extracts were concentrated under reduced pressure. The residue was suspended in a 1:3 mixture of ethyl acetate and heptane, stirred for several minutes, and filtered, affording the product as a gray solid, which was used without additional purification. Yield: 21.8 g, 51.6 mmol, 71%. LCMS m/z 323.1 [(M-Boc)+H]+. 1H NMR (400 MHz, DMSO-d6), characteristic peaks: δ 7.46-7.51 (m, 2H), 7.42 (br dd, J=7.5, 7.4 Hz, 2H), 7.32-7.38 (m, 3H), 7.18 (brd, J=8.8 Hz, 2H), 5.16 (s, 2H), 2.92 (s, 3H), 1.54 (s, 9H).
Step 2. Synthesis of 6-(4-hydroxyphenyl)- 1,5-dimethylpyrimidine-2,4( 1H,3iï)-dione (031).
Compound C30 (21.8 g, 51.6 mmol) was mixed with a 30% solution of hydrogen bromide in acetic acid (100 mL, 520 mmol) and stirred at room température for 4 hours. Acetic acid was removed under reduced pressure, and the resulting oil was dissolved in a minimal quantity of éthanol and diluted with water, providing a slightly cloudy mixture. After this was evaporated to dryness, the resulting solid was suspended in water and stirred for several minutes. Filtration afforded the product as a tan solid, which was used without additional purification. Yield: 11.4 g, 49.1 mmol, 95%. LCMS m/z 233.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 11.34 (brs, 1H), 9.85 (br s, 1H), 7.14 (brd, J=8.6 Hz, 2H), 6.89 (br d, J=8.6 Hz, 2H), 2.88 (s, 3H), 1.50 (s, 3H).
Step 3. Synthesis of 6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]phenyl}-1,5dimethylpyrimidine-2,4(11-1,3H)-dione (11).
Césium carbonate (32.6 g, 100 mmol) was added to a mixture of C31 (11.4 g, 49.1 mmol) and 2,3-dichloro-4-methylpyridine (11.9 g, 73.4 mmol) in 1-methylpyrrolidin-2-one (100 mL), and the reaction mixture was heated at 140 °C for 24 hours. Additional 2,3-dichloro-4methylpyridine (4.0 g, 25 mmol) was added, and heating was continued for 24 hours. The reaction mixture was cooled to approximately 50 °C and poured into ice water (500 mL); the resulting suspension was stirred for 5 minutes and then filtered. The collected solid was dissolved in hot éthanol (600 mL), treated with charcoal and magnésium sulfate, and stirred under heating for 10 minutes. The hot mixture was filtered through diatomaceous earth, and the hot filtrate was diluted with heptane (400 mL) while stirring, then cooled to 0 °C. After stirring for 45 minutes at 0 °C, the mixture was filtered to afford the crude product as an off-white solid (11.75 g). The filtrate was concentrated under reduced pressure, suspended in diethyl ether, and filtered to provide a solid, which was extracted several times with hot ethyl acetate; the combined ethyl acetate extracts were concentrated in vacuo, yielding additional crude product (2 g). The two lots of crude product were combined and recrystallized from ethyl acetate / heptane to afford the final product as a white solid. Yield: 11.1 g, 31.0 mmol, 63%. LCMS m/z 358.2, 360.2 [M+H]+. 1H NMR (400 MHz, DMSO-de) δ 11.42 (brs, 1H), 8.00 (d, J=5.0 Hz, 1H), 7.42 (br d, J=8.8 Hz, 2H), 7.30 (br d, J=8.7 Hz, 2H), 7.21 (br d, J=5.0 Hz, 1H), 2.91 (s, 3H), 2.44 (s, 3H), 1.53 (s, 3H).
Example 12
6-(4-{[3-(Difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1-ethyl-5-methylpyrimidine2,4(1H,3H)-dione (12)
To a mixture of 2-chloro-3-(difluoromethyl)pyridine (15 g, 92 mmol) and césium carbonate (90 g, 280 mmol) in dimethyl sulfoxide (300 mL) was added 4-bromo-3-methylphenol (19.8 g, 106 mmol). The reaction mixture was stirred at 100 °C for 18 hours, then diluted with water (1 L) and extracted with ethyl acetate (5 x 200 mL). The combined organic layers were dried, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: 40:1 petroleum ether / ethyl acetate) afforded the product as a white solid. Yield: 27 g, 86 mmol, 93%. 1H NMR (400 MHz, CD3OD) 5 8.19 (br d, J=4 Hz, 1H), 8.07 (d, J=7.2 Hz, 1H), 7.56 (d, J=8.5 Hz, 1H),
7.19-7.25 (m, 1H), 7.10 (br d, J=2.5 Hz, 1H), 7.08 (t, JHF=54.8 Hz, 1H), 6.90 (dd, J=8.6, 2.6 Hz, 1H), 2.39 (s, 3H).
Step 2. Synthesis of 3-(difluoromethyl)-2-[3-methyl-4-(4,4,5,5-tetramethyl-1,3,2dioxaborolan-2-yl)phenoxy]pyridine (C33).
To a mixture of C32 (27 g, 86 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi-1,3,2dioxaborolane (32.8 g, 129 mmol) and potassium acetate (25.8 g, 263 mmol) in 1,4-dioxane (500 mL) was added [1,T-bis(diphenylphosphino)ferrocene]dichloropalladium(ll) (6.3 g, 8.6 mmol). The mixture was stirred at 100 °C for 18 hours, then filtered. After concentration of the filtrate under reduced pressure, the residue was purified via silica gel chromatography (Eluent:
petroleum ether) to provide the product as a yellow oil. Yield: 16 g, 44 mmol, 51%. LCMS m/z
362.0 [M+H]+. Ή NMR (400 MHz, CD3OD) 5 8.18-8.22 (m, 1H), 8.07 (brd, J=7 Hz, 1H), 7.75 (d,
Step 3. Synthesis of 6-bromo-3-(3,4-dimethoxybenzyl)-1-ethyl-5-methylpyrimidine2,4(1H,3H)-dione (C34).
Compound C10 was converted to the product according to the method used for synthesis of C26 in Examples 9 and 10. The product was obtained as a light yellow oil. Yield: 720 mg, 1.88 mmol, 84%. 1H NMR (400 MHz, CDCI3) δ 7.07-7.14 (m, 2H), 6.80 (d, J=8.2 Hz, 1H), 5.06 (S, 2H), 4.23 (q, J=7.0 Hz, 2H), 3.88 (s, 3H), 3.85 (s, 3H), 2.13 (s, 3H), 1.30 (t, J=7.0 Hz, 3H).
Step 4. Synthesis of 6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-3-(3,4dimethoxybenzyl)-1 -ethyl-5-methylpyrimidine-2,4(1b\,3H)-dione (C35).
To a mixture of C34 (57.5 mg, 0.150 mmol), C33 (108 mg, 0.299 mmol), and tetrakis(triphenylphosphine)palladium(0) (17 mg, 15 pmol) in a mixture of 1,4-dioxane (3 mL) and water (20 drops) was added barium hydroxide (77 mg, 0.45 mmol). The reaction mixture was stirred at 60 °C for 20 hours, then diluted with saturated aqueous ammonium chloride solution (20 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organic layers were dried, fîltered, and concentrated in vacuo. Préparative high-performance liquid chromatography afforded the product as a white solid. Yield: 30 mg, 56 pmol, 37%. LCMS m/z 538.0 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.28 (br d, J=4 Hz, 1H), 8.04 (d, J=7.2 Hz, 1H), 7.10-7.25 (m, 6H), 7.02 (t, JHf=55.1 Hz, 1H), 6.83 (d, J=8.2 Hz, 1H), 5.17 (s, 2H), 3.90 (s, 3H), 3.87 (s, 3H), 3.81-3.9 (m, 1H), 3.27-3.38 (m, 1H), 2.18 (s, 3H), 1.66 (s, 3H), 1.07 (t, J=7.0 Hz, 3H).
Step 5. Synthesis of 6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1-ethyl-5methylpyrimidine-2,4(7H,3H)-dione ( 12).
Compound C35 was deprotected using the method described for synthesis of C29 in Examples 9 and 10. In this case, purification was carried out via reversed phase highperformance liquid chromatography (Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 30% to 50% B). LCMS m/z 388.1 [M+H]+. 1H NMR (600 MHz, DMSO-c/6) δ 8.34 (br d, J=4.5 Hz, 1H), 8.13 (br d, J=7.2 Hz, 1H), 7.35 (d, J=8.3 Hz, 1H), 7.32 (dd, J=7.4, 5.0 Hz, 1H), 7.28 (t, JHf=54.4 Hz, 1H), 7.24 (brd, J=2.1 Hz, 1H), 7.18 (br dd, J=8.2, 2.3 Hz, 1H), 3.633.71 (m, 1H), 3.08-3.15 (m, 1H), 2.15 (s, 3H), 1.45 (s, 3H), 0.95 (t, J=7.0 Hz, 3H).
Example 13 (-)-6-(4-{[3-(Difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-1,5-dimethylpyrimidine2,4(1H,3H)-dione (13)
C36
Step 1. Synthesis of 2-chloro-3-(difluoromethoxy)pyridine (C36).
This reaction was carried out 3 times. A mixture of potassium carbonate (282 g, 2.04 mol) and /V,A/-dimethylformamide (750 mL) was heated to 100 °C and slowly treated, in a dropwise manner over 1 hour, with a solution of 2-chloropyridin-3-ol (66.7 g, 515 mmol) and sodium chloro(difluoro)acetate (200 g, 1.31 mol) in A/,A/-dimethylformamide (750 mL). After completion of the addition, the reaction mixture was stirred at 100 °C for 1 hour, then cooled to 25 °C and partitioned between water (10 L) and tert-butyl methyl ether (5 L). The aqueous layer was extracted with ethyl acetate (4 x 2.5 L), and the combined organic layers were washed with saturated aqueous sodium chloride solution (6 x 2.5 L), dried over sodium sulfate, filtered, and concentrated in vacuo. The combined crude products from the three reactions were purified via distillation at reduced pressure (30-40 °C, 1-5 mm Hg) to provide the product as a colorless oil.
Yield: 192 g, 1.07 mol, 69%. LCMS m/z 180.0 [M+H]+. Ή NMR (400 MHz, CDCI3) δ 8.26-8.30 (m, 1H), 7.60 (brd, J=8.2 Hz, 1H), 7.28 (brdd, J=8.0, 4.8 Hz, 1H), 6.60 (t, JHf=72.5 Hz, 1H).
Step 2. Synthesis of 3-[(benzyloxy)methyl]-6-bromo-1,5-dimethylpyrimidine-2,4(1iï,3iï)dione (C37).
1,8-Diazabicyclo[5.4.0]undec-7-ene (6.00 mL, 40.2 mmol) was added to a suspension of C2 (8.00 g, 36.5 mmol) and benzyl chloromethyl ether (95%, 5.86 mL, 40.2 mmol) in acetonitrile (100 mL). After 90 hours at room température, the reaction mixture was concentrated in vacuo, diluted with water, and extracted several times with ethyl acetate. The combined organic layers were washed sequentially with water and with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (Gradient: 10% to 25% ethyl acetate in heptane) afforded the product as a white solid. Yield: 10.1 g, 29.8 mmol, 82%. 1H NMR (400 MHz, CDCI3) δ 7.24-7.39 (m, 5H), 5.52 (s, 2H), 4.71 (s, 2H), 3.63 (s, 3H), 2.11 (s, 3H).
Step 3. Synthesis of 3-[(benzyloxy)methyl]-6-[4-(methoxymethoxy)-2-methylphenyl]-1,5dimethylpyrimidine-2,4(1H,3H)-dione (C38).
To a mixture of C37 (10.5 g, 31.0 mmol), [4-(methoxymethoxy)-2-methylphenyl]boronic acid (7.58 g, 38.7 mmol) and potassium carbonate (13 g, 94 mmol) in 1,4-dioxane (170 mL) was added [1,T-bis(diphenylphosphino)ferrocene]dichloropalladium(ll), dichloromethane complex (1.3 g, 1.6 mmol). The reaction mixture was stirred at 80 °C for 18 hours and filtered; the filtrate was concentrated in vacuo. Purification via silica gel chromatography (Gradient: 0% to 30% ethyl acetate in petroleum ether) provided the product as a yellow oil. Yield: 10.5 g, 25.6 mmol, 83%. 1H NMR (400 MHz, CDCI3) δ 7.25-7.46 (m, 5H), 6.93-7.02 (m, 3H), 5.60 (AB quartet, Jab=9-4 Hz, ΔνΑΒ=9.7 Hz, 2H), 5.22 (s, 2H), 4.79 (s, 2H), 3.52 (s, 3H), 3.00 (s, 3H), 2.12 (br s, 3H), 1.63 (s, 3H).
Step 4. Synthesis of 3-[(benzyloxy)methyl]-6-(4-hydroxy-2-methylphenyl)-1,5dimethylpyrimidine-2,4( 1H,3H)-dione (C39).
To a solution of C38 (9.0 g, 22 mmol) in tetrahydrofuran (70 mL) was added aqueous hydrochloric acid (8 M, 70 mL), and the reaction mixture was stirred at room température for 1 hour. After extraction with ethyl acetate (5 x 100 mL), the combined organic layers were concentrated in vacuo; silica gel chromatography (Gradient: 0% to 50% ethyl acetate in petroleum ether) afforded the product as a white solid. Yield: 6.3 g, 17 mmol, 77%. LCMS m/z 389.0 [M+Na+], 1H NMR (400 MHz, CDCIg) δ 7.43 (br d, J=7 Hz, 2H), 7.25-7.37 (m, 3H), 6.91 (d, J=7.9 Hz, 1H), 6.78-6.84 (m, 2H), 5.61 (AB quartet, JAB=9.4 Hz, ΔνΑΒ=9.2 Hz, 2H), 5.47 (s, 1H), 4.79 (s, 2H), 3.01 (s, 3H), 2.09 (s, 3H), 1.64 (s, 3H).
Step 5. Synthesis of 3-[(benzyloxy)methyl]-6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2methylphenyl)- 1,5-dimethylpyrimidine-2,4(1iï,3iï)-dione (C40).
A suspension of C39 (10 g, 27 mmol), C36 (5.88 g, 32.7 mmol), and césium carbonate (99%, 13.5 g, 41.0 mmol) in dimethyl sulfoxide (200 mL) was heated to 80 °C for 18 hours.
Compound C36 (2.9 g, 16 mmol) was added, and the reaction mixture was heated at 90 °C for 3 hours, then at 80 °C for 66 hours. After cooling to room température, the reaction mixture was diluted with water and extracted three times with ethyl acetate. The combined organic layers were washed with water (5 x 300 mL), washed with saturated aqueous sodium chloride solution (200 mL), dried over magnésium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Gradient: 25% to 50% ethyl acetate in heptane) provided the product as a viscous, light yellow oil. Yield: 10.8 g, 21.2 mmol, 78%. LCMS m/z510.2 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.05 (dd, J=4.9, 1.7 Hz, 1H), 7.61-7.65 (m, 1H), 7.40-7.44 (m, 2H), 7.307.36 (m, 2H), 7.24-7.29 (m, 1H), 7.11-7.16 (m, 2H), 7.10 (dd, J=7.9, 4.9 Hz, 1H), 7.08 (brd, J=8 Hz, 1H), 6.70 (t, JHf=73.5 Hz, 1H), 5.61 (AB quartet, JAb=9-5 Hz, Avab=9.2 Hz, 2H), 4.79 (br s, 2H), 3.04 (s, 3H), 2.16 (br s, 3H), 1.66 (s, 3H).
Step 6. Synthesis of (-)-6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-1,5dimethylpyrimidine-2,4(1iï,3H)-dione (13).
A mixture of C40 (10.8 g, 21.2 mmol) and trifluoroacetic acid (110 mL) was heated at 80 °C for 1 hour. The reaction mixture was concentrated in vacuo, treated with dichloromethane and concentrated again, then treated with tetrahydrofuran, concentrated under reduced pressure, and dried under high vacuum. The residue was diluted with tetrahydrofuran (50 mL), cooled in an ice bath, and treated with concentrated ammonium hydroxide (50 mL). The flask was removed from the ice bath and the reaction mixture was stirred at room température for 45 minutes; after removal of solvents in vacuo, purification via silica gel chromatography (Gradient: 25% to 100% ethyl acetate in heptane) provided a racemic mixture of 13 and its atropenantiomer. This was combined with material obtained from a similar reaction carried out on C40 (15.3 g, 30.0 mmol), and separated via supercritical fluid chromatography (Column: Phenomenex Lux Cellulose-2, 5 pm; Eluent: 3:2 carbon dioxide / methanol). The first-eluting atropenantiomer, which exhibited a négative (-) rotation, was assigned as atropenantiomer 13. Yield: 4.8 g, 12 mmol, 23%. This material was dissolved in hot ethyl acetate (200 mL) and slowly treated with heptane (100 mL) while maintaining the mixture at reflux. After slowly cooling to room température, the mixture was stirred at room température for 18 hours, then cooled to 0 °C and stirred for 30 minutes. Filtration afforded the product as a powdery white solid. Yield: 4.17 g, 10.7 mmol, 89% from the recrystallization. LCMS m/z390.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 11.45 (br s, 1H), 8.06 (dd, J=4.8, 1.5 Hz, 1H), 7.81 (br d, J=7.9 Hz, 1H), 7.32 (t, Jhf=73.4 Hz, 1H), 7.12-7.31 (m, 4H), 2.87 (s, 3H), 2.14 (s, 3H), 1.48 (s, 3H).
Examples 14 and 15 (-)-6-(4-{[3-(Difluoromethyl)pyndin-2-yl]oxy}-2-methylphenyl)-1,5-dimethylpyrimidine2,4(1iï,3iï)-dione (14) and (+)-6-(4-{[3-(Difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1,5dimethylpyrimidine-2,4( 1iï,3f\)-dione (15)
Step 1. Synthesis of 3-[(benzyloxy)methyl]-6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2methylphenyl)-1,5-dimethylpyrimidine-2,4(114,314)-dione (C41).
Compound C39 was reacted with 2-chloro-3-(difluoromethyl)pyridine using the method 5 described for synthesis of C40 in Example 13. The product was obtained as a white solid. Yield: 17.3 g, 35.1 mmol, 86%. LCMS m/z 494.2 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.27-8.31 (m, 1H), 8.02-8.07 (m, 1H), 7.41-7.46 (m, 2H), 7.32-7.37 (m, 2H), 7.26-7.31 (m, 1H), 7.08-7.21 (m, 4H), 7.03 (t, JHf=55.1 Hz, 1H), 5.62 (AB quartet, JAB=9.5 Hz, ΔνΑΒ=9·5 Hz, 2H), 4.80 (br s, 2H), 3.05 (s, 3H), 2.17 (br s, 3H), 1.68 (s, 3H).
Step 2. Synthesis of (-)-6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1,5dimethylpyrimidine-2,4( 1\4,3Y\)-dione ( 14) and (+)-6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2methylphenyl)-1,5-dimethylpyrimidine-2,4( 1l4,3iï)-dione (15).
Compound C41 was converted to a racemic mixture of the products using the method described for synthesis 13 in Example 13. This racemate was obtained as an off-white solid.
Yield: 12.1 g, 32.4 mmol, 92%. It was separated into its component atropenantiomers via supercritical fluid chromatography (Column: Phenomenex Lux Cellulose-2, 5 pm; Eluent: 55:45 carbon dioxide / methanol). The first-eluting atropenantiomer exhibited a négative (-) rotation, and was designated as Example 14 (5.15 g). This material was dissolved in hot ethyl acetate, concentrated to a volume of 50 mL, and allowed to crystallize at room température; 14 was isolated as a white solid, 3.35 g. The fîltrate was concentrated and similarly recrystallized to afford a white solid (450 mg). Combined yield of 14: 3.8 g, 10 mmol, 28%. The second-eluting atropenantiomer, obtained as an off-white solid exhibiting a positive (+) rotation, was designated as Example 15. Yield: 4.9 g, 13.1 mmol, 37%. 14: LCMS m/z 374.2 [M+H]+. 1H NMR (400 MHz,
CDCIg) 6 8.47 (brs, 1H), 8.27-8.31 (m, 1H), 8.02-8.07 (m, 1H), 7.12-7.21 (m, 4H), 7.03 (t,
JHf=55.0 Hz, 1 H), 3.06 (s, 3H), 2.21 (br s, 3H), 1.68 (s, 3H). 15: LCMS m/z374.0 [M+H]+. 1H
NMR (600 MHz, CDCI3) δ 8.98 (br s, 1H), 8.29 (br d, J=4.7 Hz, 1H), 8.04 (br d, J=7.5 Hz, 1H),
7.13-7.21 (m, 4H), 7.03 (t, JHf=55.1 Hz, 1H), 3.06 (s, 3H), 2.21 (s, 3H), 1.68 (s, 3H).
Examples 16 and 17 (+)-5-(4-{[3-(Difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6-dimethylpyridazin5 3(2iï)-one (16) and (-)-5-(4-{[3-(Difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,610 dimethylpyridazin-3(2H)-one (17)
F
F
N (+) > 1β
Step 1. Synthesis of 4-hydroxy-3,5-dimethylfuran-2(5H)-one (C42).
Méthylation of ethyl 3-oxopentanoate according to the method of D. Kalaitzakis et al.,
Tetrahedron: Asymmetry 2007, 18, 2418-2426, afforded ethyl 2-methyl-3-oxopentanoate;
subséquent treatment with 1 équivalent of bromine in chloroform provided ethyl 4-bromo-25 methyl-3-oxopentanoate. This crude material (139 g, 586 mmol) was slowly added to a 0 °C solution of potassium hydroxide (98.7 g, 1.76 mol) in water (700 mL). The internai reaction température rose to 30 °C during the addition. The reaction mixture was then subjected to vigorous stirring for 4 hours in an ice bath, at which point it was acidified via slow addition of concentrated hydrochloric acid. After extraction with ethyl acetate, the aqueous layer was saturated with solid sodium chloride and extracted three additional times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated under reduced pressure to afford a mixture of oil and solid (81.3 g). This material was suspended in chloroform (200 mL); the solids were removed via filtration and washed with chloroform (2 x 50 mL). The combined filtrâtes were concentrated in vacuo and treated with a 3:1 mixture of heptane and diethyl ether (300 mL). The mixture was vigorously swirled until some of the oil began to solidify. It was then concentrated under reduced pressure to afford an oily solid (60.2 g). After addition of a 3:1 mixture of heptane and diethyl ether (300 mL) and vigorous stirring for 10 minutes, filtration afforded the product as an off-white solid. Yield: 28.0 g, 219 mmol, 37%. 1H NMR (400 MHz, CDCI3) δ 4.84 (br q, 4=6.8
Hz, 1 H), 1.74 (br s, 3H), 1.50 (d, 4=6.8 Hz, 3H).
Step 2. Synthesis of 2,4-dimethyl-5-oxo-2,5-dihydrofuran-3-yl trifluoromethanesulfonate (C43).
Trifluoromethanesulfonic anhydride (23.7 mL, 140 mmol) was added portion-wise to a solution of C42 (15.0 g, 117 mmol) and /V,A/-diisopropylethylamine (99%, 24.8 mL, 140 mmol) in 25 dichloromethane (500 mL) at -20 °C, at a rate sufficient to maintain the internai reaction température below -10 °C. The reaction mixture was allowed to warm gradually from -20 °C to 0 °C over 5 hours. It was then passed through a plug of silica gel, dried over magnésium sulfate, and concentrated in vacuo. The residue was suspended in diethyl ether and filtered; the filtrate was concentrated under reduced pressure. Purification using silica gel chromatography (Gradient: 0% to 17% ethyl acetate in heptane) afforded the product as a pale yellow oil. Yield: 21.06 g, 80.94 mmol, 69%. 1H NMR (400 MHz, CDCI3) δ 5.09-5.16 (m, 1H), 1.94-1.96 (m, 3H), 1.56 (d, 4=6.6 Hz, 3H).
Step 3. Synthesis of 2-[4-(benzyloxy)-2-methylphenyl]-4,4,5,5-tetramethyl-1,3,2dioxaborolane (C44).
Benzyl 4-bromo-3-methylphenyl ether was converted to the product using the method described for synthesis of C33 in Example 12. The product was isolated as a yellow gel. Yield:
g, 46 mmol, 67%. 1H NMR (400 MHz, CDCI3) δ 7.73 (d, 4=8.0 Hz, 1H), 7.30-7.46 (m, 5H),
6.76-6.82 (m, 2H), 5.08 (s, 2H), 2.53 (s, 3H), 1.34 (s, 12H).
Step 4. Synthesis of 4-[4-(benzyloxy)-2-methylphenyl]-3,5-dimethylfuran-2(5H)-one (C45).
Compound C43 (5.0 g, 19 mmol), C44 (7.48 g, 23.1 mmol), tetrakis(trîphenylphosphine)palladium(0) (2.22 g, 1.92 mmol), and sodium carbonate (4.07 g,
38.4 mmol) were combined in 1,4-dioxane (100 mL) and water (5 mL), and heated at reflux for 2 hours. The reaction mixture was filtered and the filtrate was concentrated in vacuo. Purification using silica gel chromatography (Eluents: 10:1, then 5:1 petroleum ether / ethyl acetate) provided the product as a white solid. Yield: 5.8 g, 19 mmol, 100%. NMR (400 MHz, CDCI3) δ 7.33-7.49 (m, 5H), 6.98 (d, J=8.5 Hz, 1H), 6.94 (br d, J=2.5 Hz, 1H), 6.88 (br dd, J=8.3, 2.5 Hz, 10 1 H), 5.20 (qq, J=6.7, 1.8 Hz, 1 H), 5.09 (s, 2H), 2.21 (s, 3H), 1.78 (d, J=1.8 Hz, 3H), 1.31 (d,
J=6.8 Hz, 3H).
Step 5. Synthesis of 4-[4-(benzyloxy)-2-methylphenyl]-5-hydroxy-3,5-dimethylfuran2(5H)-one (C46).
A solution of C45 (5.4 g, 18 mmol) and 1,8-diazabicyclo[5.4.0]undec-7-ene (13.3 g, 87.4 15 mmol) in acetonitrile (100 mL) was cooled to -60 °C. Oxygen was bubbled into the reaction mixture for 20 minutes at -60 °C; the solution was then stirred at 50 °C for 18 hours. The reaction mixture was concentrated in vacuo and purified via silica gel chromatography (Eluent: 5:1 petroleum ether / ethyl acetate) to provide the product as a colorless oil. Yield: 3.5 g, 11 mmol, 61%. Ή NMR (400 MHz, CDCI3), characteristic peaks: δ 7.33-7.49 (m, 5H), 6.92-6.96 (m, 20 1 H), 6.88 (dd, J=8.5, 2.5 Hz, 1 H), 5.09 (s, 2H), 2.20 (s, 3H), 1.73 (s, 3H).
Step 6. Synthesis of 5-[4-(benzyloxy)-2-methylphenyl]-4,6-dimethylpyridazin-3(2H)-one (C47).
A mixture of C46 (3.5 g, 11 mmol) and hydrazine hydrate (85% in water, 1.9 g, 32 mmol) in n-butanol (60 mL) was heated at reflux for 18 hours. After removal of volatiles under reduced 25 pressure, the residue was stirred with ethyl acetate (20 mL) for 30 minutes, whereupon filtration provided the product as a white solid. Yield: 2.0 g, 6.2 mmol, 56%. 1H NMR (400 MHz, CDCI3) δ 10.93 (brs, 1H), 7.33-7.51 (m, 5H), 6.96 (s, 1H), 6.88-6.94 (m, 2H), 5.10 (s, 2H), 2.04 (s, 3H), 1.95 (s, 3H), 1.91 (s, 3H).
Step 7. Synthesis of 5-[4-(benzyloxy)-2-methylphenyl]-4,6-dimethyl-2-(tetrahydro-2iï30 pyran-2-yl)pyridazin-3(2H)-one (C48).
A mixture of C47 (17.8 g, 55.6 mmol), 3,4-dihydro-2/-/-pyran (233 g, 2.77 mol) and ptoluenesulfonic acid monohydrate (2.1 g, 11 mmol) in tetrahydrofuran (800 mL) was heated at reflux for 18 hours. Triethylamine (10 mL, 72 mmol) was added, and the mixture was concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 25% ethyl acetate in petroleum ether) afforded the product as a solid, presumed to be a mixture of diastereomeric atropisomers from its 1H NMR spectrum. Yield: 20 g, 49 mmol, 88%. 1H NMR (400 MHz,
CDCIg), characteristic peaks: δ 7.32-7.50 (m, 5H), 6.82-6.96 (m, 3H), 6.15 (br d, J=10.3 Hz, 1H),
Step 8. Synthesis of 5-(4-hydroxy-2-methylphenyl)-4,6-dimethyl-2-(tetrahydro-2H-pyran-
2-yl)pyridazin-3(2iï)-one (C49).
Palladium (10% on carbon, 1.16 g, 1.09 mmol) was added to a solution of C48 (1.47 g,
3.63 mmol) in methanol (30 mL) and ethyl acetate (10 mL), and the mixture was hydrogenated (50 psi) on a Parr shaker for 18 hours at room température. The reaction mixture was filtered through diatomaceous earth, and the filter pad was rinsed with ethyl acetate; the combined filtrâtes were concentrated in vacuo and triturated with heptane, affording the product as a white solid, judged to be a mixture of diastereomeric atropisomers from its 1H NMR spectrum. Yield: 1.01 g, 3.21 mmol, 88%. 1H NMR (400 MHz, CDCI3), characteristic peaks: δ 6.74-6.85 (m, 3H), 6.12-6.17 (m, 1H), 4.15-4.23 (m, 1H), 3.76-3.84 (m, 1H), 2.28-2.41 (m, 1H), 1.99 and 2.01 (2 s, total 3H), 1.97 and 1.98 (2 s, total 3H), 1.89 and 1.89 (2 s, total 3H).
Step 9. Synthesis of 5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6dimethyl-2-(tetrahydro-2H -pyran-2-yl)pyridazin-3(2H)-one (C50).
Compound C49 was reacted with 2-ch)oro-3-(difluoromethyl)pyridine using the method described for synthesis of C8 in Examples 3 and 4. The product was obtained as a white solid, presumed to be a mixture of diastereomeric atropisomers from its 1H NMR spectrum. Yield: 17.5 g, 39.6 mmol, 82%. LCMS m/z358.2 [(M - tetrahydropyran)+1], 1H NMR (400 MHz, DMSO-d6) δ 8.31-8.35 (m, 1H), 8.11-8.15 (m, 1H), 7.29 (t, JHf=54.5 Hz, 1H), 7.28-7.33 (m, 1H), 7.20-7.22 (m, 1H), 7.11-7.19 (m, 2H), 5.92-5.98 (m, 1H), 3.94-4.01 (m, 1H), 3.57-3.65 (m, 1H), 2.13-2.26 (m, 1 H), 2.02 and 2.03 (2 br s, total 3H), 1.93-2.0 (m, 1 H), 1.92 (s, 3H), 1.78 (s, 3H), 1.61 -1.74 (m, 2H), 1.48-1.58 (m, 2H).
Step 10. Synthesis of (+)-5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6dimethylpyridazin-3(2iï)-one (16) and (-)-5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2methylphenyl)-4,6-dimethylpyridazin-3(2H)-one ( 17).
Hydrogen chloride in 1,4-dioxane (4 M, 198 mL, 792 mmol) was added to a solution of C50 (17.5 g, 39.6 mmol) in dichloromethane (200 mL) and 1,4-dioxane (200 mL), and the reaction mixture was stirred at room température for 18 hours. After solvents had been removed in vacuo, the residue was suspended in diethyl ether (200 mL) and slowly treated with a halfsaturated aqueous solution of sodium bicarbonate. The suspension was vigorously stirred for 15 minutes, then filtered; the collected solid was washed twice with water and twice with diethyl ether. The solid was then suspended in éthanol (200 mL), concentrated to dryness, resuspended in éthanol (200 mL) and concentrated once more. The residue was similarly treated with diethyl ether and with heptane to afford the racemic product as a white solid. Yield: 12.0 g, 33.6 mmol, 85%. LCMS m/z 358.2 [M+H]+. Ή NMR (400 MHz, DMSO-d6) δ 12.82 (br s, 1H), 8.32-8.36 (m, 1H), 8.10-8.15 (m, 1H), 7.29 (t, JHf=54.2 Hz, 1H), 7.28-7.33 (m, 1H), 7.19-
7.22 (m, 1H), 7.10-7.17 (m, 2H), 2.02 (s, 3H), 1.87 (s, 3H), 1.74 (s, 3H). Séparation of the racemate into its component atropenantiomers was carried out via supercritical fluid chromatography (Column: Chiral Technologies, Chiralpak AS-H, 5 um; Eluent: 85:15 carbon dioxide / methanol). The first-eluting atropenantiomer, obtained as a white solid that exhibited a positive (+) rotation, was designated as Example 16. Yield: 5.22 g, 14.6 mmol, 37%. The second-eluting atropenantiomer, also obtained as a white solid but with a négative (-) rotation, was designated as Example 17. Yield: 5.31 g, 14.8 mmol, 37%. 16: LCMS m/z 358.2 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 8.23-8.27 (m, 1H), 8.08-8.12 (m, 1H), 7.26 (dd, J=7.5, 4.9 Hz, 1H), 7.18-7.20 (m, 1H), 7.12-7.14 (m, 2H), 7.12 (t, JHF=55 Hz, 1H), 2.09 (br d, J=0.4 Hz, 3H), 2.00 (s, 3H), 1.90 (s, 3H). 17: LCMS m/z 358.2 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 8.23-8.27 (m, 1H), 8.08-8.12 (m, 1H), 7.26 (dd, J=7.6, 5.0 Hz, 1H), 7.18-7.20 (m, 1H), 7.12-7.14 (m, 2H), 7.12 (t, JHf=55 Hz, 1H), 2.09 (br d, J=0.5 Hz, 3H), 2.00 (s, 3H), 1.90 (s, 3H).
Example 18
6-{4-[(3-Chloropyridin-2-yl)sulfanyl]-2-methylphenyl}-1,5-dimethylpyrimidine-2,4( 1Η,3Η/dione (18)
Step 1. Synthesis of4-(3,5-dimethyl-2,6-dioxo-1-{[2-(trimethylsilyl)ethoxy]methyl}1,2,3,6-tetrahydropyrimidin-4-yl)-3-methylphenyl trifluoromethanesulfonate (C51).
Trifluoromethanesulfonic anhydride (1.3 g, 4.6 mmol) was slowly added to a 0 °C solution of C5 (600 mg, 1.6 mmol) in pyridine (15 mL), and the reaction mixture was stirred at room température for 3 hours. After solvent had been removed under reduced pressure, the residue was purified by silica gel chromatography (Gradient: 5% to 17% ethyl acetate in petroleum ether) to afford the product as a yellow oil. Yield: 790 mg, 1.55 mmol, 97%. 1H NMR (400 MHz, CDCI3) δ 7.27-7.33 (m, 2H), 7.21-7.25 (m, 1H), 5.50 (AB quartet, JAb=9.2 Hz, Avab=4.1 Hz, 2H), 3.73-3.79 (m, 2H), 3.02 (s, 3H), 2.26 (br s, 3H), 1.63 (s, 3H), 1.00-1.06 (m, 2H), 0.03 (s, 9H).
Step 2. Synthesis ofS-[4-(3,5-dimethyl-2,6-dioxo-1-{[2-(trimethylsilyl)ethoxy]methyl}1,2,3,6-tetrahydropyrimidin-4-yl)-3-methylphenyl] ethanethioate (C52).
Tris(dibenzylideneacetone)dîpalladium(0) (27 mg, 29 pmol) and (fî)-(-)-1-[(SP)-2(dicyclohexylphosphino)ferrocenyljethyldi-tert'-butylphosphine (Josiphos ligand, 33 mg, 60 pmol) were added to a solution of C51 (305 mg, 0.600 mmol) in degassed toluene (7 mL), and the mixture was stirred for 5 minutes at room température. Potassium thioacetate (274 mg, 2.40 mmol) was added and the reaction mixture was heated at 120 °C for 24 hours. It was then filtered through a pad of diatomaceous earth, and the pad was washed with ethyl acetate; the combined filtrâtes were concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 30% ethyl acetate in petroleum ether) provided the product as a brown gum. Yield: 172 mg, 0.396 mmol, 66%. 1H NMR (400 MHz, CDCI3) δ 7.38-7.44 (m, 2H), 7.15 (d, J=7.8 Hz, 1H), 5.48-5.53 (m, 2H), 3.73-3.79 (m, 2H), 3.03 (s, 3H), 2.47 (s, 3H), 2.20 (s, 3H), 1.65 (s, 3H), 1.00-1.06 (m, 2H), 0.03 (s, 9H).
Step 3. Synthesis of 1,5-dimethyl-6-(2-methyl-4-sulfanylphenyl)-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(7H,3H)-dione (C53).
A solution of C52 (300 mg, 0.69 mmol) and potassium hydroxide (168 mg, 2.99 mmol) in a mixture of methanol (10 mL) and water (3 drops) was stirred at room température for 3 hours. After neutralization with 1 M aqueous hydrochloric acid, the mixture was concentrated in vacuo. Préparative thin layer chromatography on silica gel (Eluent: 3:1 petroleum ether / ethyl acetate) afforded the product as a yellow syrup. Yield: 170 mg, 0.433 mmol, 63% yield.
Step 4. Synthesis of 6-{4-[(3-chloropyridin-2-yl)sulfanyl]-2-methylphenyl}-1,5-dimethyl-3{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,3H)-dione (C54).
Compound C53 was reacted with 3-chloro-2-fluoropyridine using the method described for synthesis of C8 in Examples 3 and 4. The product was obtained as a white solid. Yield: 20 mg, 40 pmol, 40%.
Step 5. Synthesis of 6-{4-[(3-chloropyridin-2-yl)sulfanyl]-2-methylphenyl}-1,5dimethylpyrimidine-2,4( 1H, 3H)-dione (18).
A solution of C54 (20 mg, 40 pmol) in trifluoroacetic acid (5 mL) was stirred at room température for 18 hours. The reaction mixture was concentrated in vacuo and the residue was dissolved in methanol (5 mL). Potassium carbonate (69 mg, 0.50 mmol) was added, and the reaction mixture was stirred at room température for 3 hours and filtered; the filtrate was concentrated in vacuo and purified via préparative thin layer chromatography on silica gel (Eluent: 1:2 petroleum ether / ethyl acetate) to provide the product as a white solid. Yield: 7.5 mg, 20 pmol, 50%. LCMS m/z 374.0 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.25 (dd, J=4.7, 1.6 Hz, 1 H), 8.19 (brs, 1H), 7.64 (dd, J=7.9, 1.6 Hz, 1H), 7.55-7.57 (m, 1H), 7.51-7.55 (m, 1H), 7.15 (d, J=7.8 Hz, 1H), 7.06 (dd, J=7.9, 4.6 Hz, 1H), 3.05 (s, 3H), 2.21 (brs, 3H), 1.68 (s, 3H).
Example 19
1,5-Dimethyl-6-(7-{[3-(trifluoromethyl)pyridin-2-yl]oxy}-1iï-indol-4-yl)pyrimidine2,4(1Η,3Η)-0ϊοπθ (19)
Step 1. Synthesis of tert-butyl 7-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)1iï-indole-1 -carboxylate (C55).
To a solution of tert-butyl 4-bromo-7-methoxy-1/7-indole-1-carboxylate (which may be prepared via tert-butoxycarbonyl protection of 4-bromo-7-methoxy-1 /7-indole) (1.0 g, 3.1 mmol) in 1,4-dioxane (20 mL) were added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi-1,3,2-dioxaborolane (1.46 g, 5.75 mmol), potassium acetate (902 mg, 9.19 mmol) and [1,Tbis(diphenylphosphino)ferrocene]dichloropalladium(ll), dichloromethane complex (498 mg, 0.610 mmol). The reaction mixture was stirred for 5 hours at 120 °C, then cooled and filtered; the filtrate was concentrated under reduced pressure and purified via silica gel chromatography (Gradient: 0% to 6% ethyl acetate in petroleum ether) to afford the product as a yellow solid. Yield: 520 mg, 1.4 mmol, 45%. 1H NMR (400 MHz, CDCIg) δ 7.69 (d, <7=8.0 Hz, 1H), 7.55 (d, J=3.5 Hz, 1H), 7.10 (d, <7=3.6 Hz, 1H), 6.81 (d, J=8.0 Hz, 1H), 3.96 (s, 3H), 1.62 (s, 9H), 1.37 (s, 12H).
Step 2. Synthesis of tert-butyl 4-{1-[(benzyloxy)methyl]-3,5-dimethyl-2,6-dioxo-1,2,3,6tetrahydropyrimidin-4-yl}-7-methoxy- 1H-indole-1-carboxylate (C56).
To a solution of C55 (600 mg, 1.6 mmol) in 1,4-dioxane (20 mL) were added C37 (600 mg, 1.8 mmol), tetrakis(triphenylphosphine)palladium(0) (186 mg, 0.161 mmol) and barium hydroxide (830 mg, 4.8 mmol). The reaction mixture was stirred for 18 hours at 60 °C, then cooled and filtered; the filtrate was concentrated in vacuo and subjected to silica gel chromatography (Gradient: 0% to 35% ethyl acetate in petroleum ether), providing the product as a yellow gum. Yield: 310 mg, 0.61 mmol, 38%. 1H NMR (400 MHz, CDCI3) δ 7.60 (d, J=3.6 Hz, 1H), 7.45 (br d, <7=7 Hz, 2H), 7.27-7.39 (m, 3H, assumed; partially obscured by solvent peak), 6.94 (AB quartet, JAB=8.2 Hz, ΔνΑΒ=35.2 Hz, 2H), 6.24 (d, J=3.6 Hz, 1H), 5.63 (AB
Step 3. Synthesis of 6-(7-hydroxy-1iï-indol-4-yl)-1,5-dimethylpyrimidine-2,4(1(-1,3iï)dione (C57).
Boron tribromide (1.5 g, 6.0 mmol) was added drop-wise to a -78 °C solution of C56 (310 mg, 0.61 mmol) in dichloromethane (10 mL), and the reaction mixture was stirred for 18 hours at room température. After addition of methanol (10 mL) and sodium bicarbonate (1 g), the mixture was filtered and the filtrate was concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 4% methanol in dichloromethane) afforded the product as a yellow gum. Yield: 40 mg, 0.15 mmol, 24%. 1H NMR (400 MHz, CD3OD) δ 7.29 (d, J=3.0 Hz, 1H), 6.70 (AB quartet, Jab=7.7 Hz, ΔνΑΒ=41.9 Hz, 2H), 6.18 (d, J=3.1 Hz, 1H), 3.00 (s, 3H), 1.61 (s, 3H).
Step 4. Synthesis of 1,5-dimethyl-6-(7-{[3-(trifluoromethyl)pyridin-2-yl]oxy}-1\-\-indol-4yl)pyrimidine-2,4( 1H,3iï)-dione (19).
2-Chloro-3-(trifluoromethyl)pyridine (133 mg, 0.733 mmol) and césium fluoride (12 mg, 79 pmol) were added to a solution of C57 (20 mg, 74 pmol) in A/,A/-dimethylformamide (5 mL). The reaction mixture was stirred for 18 hours at 100 °C, then cooled and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by préparative thin layer chromatography on silica gel (Eluent: 10:1 dichloromethane / methanol) to provide the product as a white solid. Yield: 9.2 mg, 22 pmol, 30%. LCMS m/z417.0 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 8.20-8.27 (m, 2H), 7.34 (d, J=3.1 Hz, 1H), 7.28 (br dd, J=7, 5 Hz, 1H), 7.01 (AB quartet, JAB=7.9 Hz, ΔνΑΒ=4.4 Hz, 2H), 6.35 (d, J=3.1 Hz, 1H), 3.05 (s, 3H), 1.65 (s, 3H).
Préparations
Préparations P1 and P2 describe préparations of some starting materials or intermediates used for préparation of certain exemplar compounds of the invention.
Préparation P1
6-(4-Hydroxy-2-methylphenyl)-1,5-dimethylpyrazin-2( 1H)-one (P1)
Pd(OAc)2 Bu3SnOMe P(o-tolyl)3
100
Step 1. Synthesis of 1-(4-methoxy-2-methylphenyl)propan-2-one (C58).
This experiment was carried out four times. Tributyl(methoxy)stannane (400 g, 1.24 mol), 1-bromo-4-methoxy-2-methylbenzene (250 g, 1.24 mol), prop-1-en-2-yl acetate (187 g, 1.87 mol), palladium(ll) acetate (7.5 g, 33 mmol) and tri-o-tolylphosphine (10 g, 33 mmol) were stirred together in toluene (2 L) at 100 °C for 18 hours. After it had cooled to room température, the reaction mixture was treated with aqueous potassium fluoride solution (4 M, 400 mL) and stirred for 2 hours at 40 °C. The resulting mixture was diluted with toluene (500 mL) and filtered through diatomaceous earth; the filter pad was thoroughly washed with ethyl acetate (2 x 1.5 L). The organic phase from the combined filtrâtes was dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Gradient: 0% to 5% ethyl acetate in petroleum ether) provided the product as a yellow oil. Combined yield: 602 g, 3.38 mol, 68%. LCMS m/z 179.0 [M+H]+. 1H NMR (400 MHz, CDCIg) δ 7.05 (d, J=8.3 Hz, 1H), 6.706.77 (m, 2H), 3.79 (s, 3H), 3.65 (s, 2H), 2.22 (s, 3H), 2.14 (s, 3H).
Step 2. Synthesis of 1-(4-methoxy-2-methylphenyl)propane-1,2-dione (C59).
Compound C58 (6.00 g, 33.7 mmol) and sélénium dioxide (7.47 g, 67.3 mmol) were suspended in 1,4-dioxane (50 mL) and heated at 100 °C for 18 hours. The reaction mixture was cooled to room température and filtered through diatomaceous earth; the filtrate was concentrated in vacuo. Silica gel chromatography (Eluent: 10% ethyl acetate in heptane) afforded the product as a bright yellow oil. Yield: 2.55 g, 13.3 mmol, 39%. LCMS m/z 193.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 7.66 (d, J=8.6 Hz, 1 H), 6.81 (br d, half of AB quartet, J=2.5 Hz, 1H), 6.78 (br dd, half of ABX pattern, J=8.7, 2.6 Hz, 1H), 3.87 (s, 3H), 2.60 (br s, 3H), 2.51 (s, 3H).
Step 3. Synthesis of 6-(4-methoxy-2-methylphenyl)-5-methylpyrazin-2(1iï)-one (C60).
Compound C59 (4.0 g, 21 mmol) and glycinamide acetate (2.79 g, 20.8 mmol) were dissolved in methanol (40 mL) and cooled to -10°C. Aqueous sodium hydroxide solution (12 N,
3.5 mL, 42 mmol) was added, and the resulting mixture was slowly warmed to room température. After stirring for 3 days, the reaction mixture was concentrated in vacuo. The residue was diluted with water, and 1 M aqueous hydrochloric acid was added until the pH was approximately 7. The aqueous phase was extracted with ethyl acetate, and the combined organic extracts were washed with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was slurried with 3:1 ethyl acetate / heptane, stirred for 5 minutes, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: ethyl acetate) provided the product as a tan solid that contained 15% of an undesired regioisomer; this material was used without further purification. Yield: 2.0 g. LCMS m/z231.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.09 (s, 1H), 7.14 (d, J=8.2 Hz, 1H), 6.82-6.87 (m, 2H), 3.86 (s, 3H), 2.20 (s, 3H), 2.11 (s, 3H).
Step 4. Synthesis of 6-(4-methoxy-2-methylphenyl)-1,5-dimethylpyrazin-2(1\-\)-one (C61)
101
Compound C60 (from the previous step, 1.9 g) was dissolved in A/,A/-dimethylformamide (40 mL). Lithium bromide (0.86 g, 9.9 mmol) and sodium bis(trimethylsilyl)amide (95%, 1.91 g,
9.89 mmol) were added, and the resulting solution was stirred for 30 minutes. Methyl lodide (0.635 mL, 10.2 mmol) was added and stirring was continued at room température for 18 hours. The reaction mixture was then diluted with water and brought to a pH of approximately 7 by slow portion-wise addition of 1 M aqueous hydrochloric acid. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed several times with water, dried over magnésium sulfate, filtered, and concentrated. Silica gel chromatography (Gradient: 75% to 100% ethyl acetate in heptane) afforded the product as a viscous orange oil. Yield: 1.67 g, 6.84 mmol, 33% over two steps. LCMS m/z245.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.17 (s, 1 H), 7.03 (br d, J=8 Hz, 1 H), 6.85-6.90 (m, 2H), 3.86 (s, 3H), 3.18 (s, 3H), 2.08 (br s, 3H), 2.00 (s, 3H).
Step 5. Synthesis of 6-(4-hydroxy-2-methylphenyl)-1,5-dimethylpyrazin-2(1H)-one (P1).
To a -78 °C solution of C61 (1.8 g, 7.4 mmol) in dichloromethane (40 mL) was added a solution of boron tribromide in dichloromethane (1 M, 22 mL, 22 mmol). The cooling bath was removed after 30 minutes, and the reaction mixture was allowed to warm to room température and stir for 18 hours. The reaction was cooled to -78 °C, and methanol (10 mL) was slowly added; the resulting mixture was gradually warmed to room température. After the solvent had been removed in vacuo, methanol (20 mL) was added, and the mixture was again concentrated under reduced pressure. The residue was diluted with ethyl acetate (300 mL) and water (200 mL), the aqueous layer was brought to pH 7 via portion-wise addition of saturated aqueous sodium carbonate solution, and the mixture was extracted with ethyl acetate (3 x 200 mL). The combined organic layers were washed with water and with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated in vacuo to afford the product as a light tan solid. Yield: 1.4 g, 6.0 mmol, 81%. LCMS m/z231.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 8.21 (s, 1H), 6.98 (d, J=8.2 Hz, 1H), 6.87-6.89 (m, 1H), 6.85 (brdd, J=8.2, 2.5 Hz, 1H),
3.22 (s, 3H), 2.06 (br s, 3H), 2.03 (s, 3H).
Préparation P2
6-(4-Hydroxy-2-methylphenyl)-1,5-dimethylpynmidin-2( 1H)-one (P2)
102
cf3so3h ciA/
C63
HO
P2
BBr3
C64
Step 1. Synthesis of 1-(4-hydroxy-2-methylphenyl)propan-1-one (C62).
3-Methylphenol (9.0 g, 83 mmol) was combined with trifluoromethanesulfonic acid (90 mL), cooled to -10 °C, and treated in a drop-wise manner with propanoyl chloride (7.7 g, 83 mmol). The reaction mixture was stirred at -10 °C for 3 hours and then at room température for 18 hours, whereupon it was poured into ice water (600 mL). The resulting solid was collected via filtration and purified by silica gel chromatography (Gradient: 5% to 70% ethyl acetate in petroleum ether) to afford the product as an off-white solid. Yield: 6.7 g, 41 mmol, 49%. 1H NMR (400 MHz, CD3OD) δ 7.75 (d, J=8.5 Hz, 1H), 6.64-6.69 (m, 2H), 2.92 (q, J=7.3 Hz, 2H), 2.45 (s, 3H), 1.13 (t, J=7.3 Hz, 3H).
Step 2. Synthesis of3-(dimethylamino)-1-(4-methoxy-2-methylphenyl)-2-methylprop-2en-1-one (C63).
This experiment was carried out in four batches. A mixture of C62 (1.0 g, 6.1 mmol) and Λ/,/V-dimethylformamide dimethyl acetal (15 mL) was stirred at 130 °C for 30 hours. The four reaction mixtures were combined and concentrated to dryness, providing the product as a dark oil. This was used for the next step without further purification. Yield: 5.0 g, 21 mmol, 86%.
Step 3. Synthesis of 6-(4-methoxy-2-methylphenyl)-1,5-dimethylpyrimidin-2(1\-\)-one (C64).
This experiment was carried out in two batches. A mixture of C63 (from the previous step, 2.5 g, 11 mmol), 1 -methylurea (1.35 g, 18.2 mmol) and p-toluenesulfonic acid (3.13 g, 18.2 mmol) in 1,4-dioxane (100 mL) was heated at reflux for 40 hours, then concentrated under reduced pressure. The residue was mixed with toluene (100 mL), treated with p-toluenesulfonic acid (3.13 g, 18.2 mmol) and heated at reflux for another 20 hours. The two crude products were combined and concentrated in vacuo. Purification via silica gel chromatography (Gradient: 0% to 5% methanol in dichloromethane) afforded the product as a brown solid. Yield: 2.5 g, 10 mmol, 45%. 1H NMR (400 MHz, CDCI3) δ 8.52 (s, 1H), 6.98 (br d, half of AB quartet, J=9 Hz, 1H), 6.86-6.92 (m, 2H), 3.87 (s, 3H), 3.24 (s, 3H), 2.08 (s, 3H), 1.78 (s, 3H).
Step 4. Synthesis of 6-(4-hydroxy-2-methylphenyl)-1,5-dimethylpyrimidin-2(1H)-one (P2).
103
To a -70 °C solution of C64 (2.5 g, 10 mmol) in dichloromethane (100 mL) was added boron tribromide (17.9 g, 71.4 mmol) drop-wise. The reaction mixture was stirred at -60 °C to
-70 °C for 1 hour and then at room température for 18 hours, whereupon it was cooled to -60 °C and quenched with methanol. Water (100 mL) was added, and the mixture was adjusted to a pH of 6 via slow addition of solid sodium bicarbonate. The mixture was extracted with dichloromethane (100 mL) and with ethyl acetate (5 x 100 mL); the combined organic layers were dried, filtered, and concentrated in vacuo. The residue was washed with a mixture of petroleum ether and ethyl acetate (4:1, 40 mL) and the solid was collected by filtration to afford the product as a yellow solid. Yield: 2.2 g, 9.5 mmol, 95%. LCMS m/z 231.0 [M+H]+. 1H NMR (400 MHz, CD3OD) δ 8.65 (s, 1H), 7.05 (d, J=8.3 Hz, 1H), 6.88-6.91 (brs, 1H), 6.87 (brdd, J=8.3, 2.2 Hz, 1H), 3.38 (s, 3H), 2.11 (s, 3H), 1.89 (s, 3H).
Method A
Method A describes a spécifie method for préparations of certain exemplar compounds of the invention.
Préparation of 1,5-dimethyl-6-[2-methyl-4-(substituted pyridin-2-yloxy)phenyl]pyrimidine2,4(1H,3H)-diones
HO'
Step 1. Synthesis of 1,5-dimethyl-6-[2-methyl-4-(substituted pyridin-2-yloxy)phenyl]-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4( 1H,3H)-diones (C65).
A solution of C5 in /V,/V-dimethylformamide (0.33 M, 300 pL, 100 pmol) was combined in a reaction vial with the appropriately substituted 2-chloropyridine or 2-fluoropyridine (100 pmol). Potassium carbonate (300 pmol), copper(l) iodide (10 pmol) and tetrabutylammonium bromide (20 pmol) were added under nitrogen, and the vial was capped and shaken at 130 °C for 16 hours. Solvent was removed using a SpeedVac® concentrator, and the residue was partitioned between ethyl acetate (1 mL) and water (1 mL); the aqueous layer was extracted with ethyl acetate (2 x 1 mL), and the combined organic layers were dried over magnésium sulfate, filtered, and concentrated to provide the crude product, which was used directly in the following step.
Step 2. Synthesis of 1,5-dimethyl-6-[2-methyl-4-(substituted pyridin-2yloxy)phenyl]pyrimidine-2,4(1H,3B )-diones.
The 1,5-dimethyl-6-[2-methyl-4-(substituted pyridin-2-yloxy)phenyl]-3-{[2(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1 A7,3/-/)-dione (C65) from the previous step was
104
dissolved in a mixture of dichloromethane and trifluoroacetic acid (4:1, 1 mL), and the reaction vial was capped and shaken at 30 °C for 16 hours. After removal of solvents, the product was purified by high-performance liquid chromatography using one of the following Systems: a)
DIKMA Diamonsil(2) C18, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile 5 phase B: acetonitrile containing 0.225% formic acid; Gradient: 35% to 70% B; b) Phenomenex
Gemini C18, 8 pm; Mobile phase A: aqueous ammonium hydroxide, pH 10; Mobile phase B: acetonitrile; Gradient: 35% to 75% B.
Table 1 below lists some additional exemplar compounds of invention (Examples 20-81) that were made using methods, intermediates, and préparations describled herein.
Table 1. Examples 20 - 81 (including Method of Synthesis and Physicochemical Data).
Example Number Structure Method of Synthesis: Example Number; Source of Noncommerci al Starting Materials Ή NMR (400 MHz, CDCI3), δ (ppm); LCMS, observed ion m/z [M+H]+ or HPLC rétention time (minutes); LCMS m/z [M+H]+ (unless otherwise indicated)
20 O I Ίι NH Α’Λ Ex 1 & 2; C51 8.25 (br s, 1 H), 8.06 (br d, J=4 Hz, 1 H), 7.62 (brd, J=7 Hz, 1H), 7.02-7.14 (m, 4H), 3.05 (s, 3H), 2.76 (q, J=7.6 Hz, 2H), 2.18 (s, 3H), 1.68 (s, 3H), 1.30 (t, J=7.5 Hz, 3H); 352.2
21 O Ai NH II L i—o 0AJ < ciqA Ex 5; C112 Ή NMR (400 MHz, CD3OD) δ 8.07 (br d, J=4.9 Hz, 1H), 7.98 (br d, J=7.8 Hz, 1H), 7.37 (AB quartet, JAB=8.3 Hz, ΔνΑΒ=35.9 Hz, 4H), 7.18 (dd, J=7.7, 4.8 Hz, 1H), 3.63 (q, J=7.0 Hz, 2H), 1.64 (s, 3H), 1.08 (t, J=7.0 Hz, 3H); 358.0, 360.0
22 I 0 \pNH κΆ^νΑ 1 ο'ίΛν Ex 6; C183 Ή NMR (400 MHz, CD3OD) δ 8.07 (dd, J=4.8, 1.7 Hz, 1H), 7.98 (dd, J=7.8, 1.6 Hz, 1H), 7.37 (br AB quartet, JAB=8.7 Hz, ΔνΑΒ=35.6 Hz, 4H), 7.17 (dd, J=7.8, 4.9 Hz, 1H), 3.05 (s, 3H), 2.11 (q, J=7.4 Hz, 2H), 0.95 (t, J=7.4 Hz, 3H); 357.9
105
23 ο I Al ΝΗ AU 0AJ 1 NC Method A 2.28 minutes4; 363
24 o 1 h NH ιΑΛ-νλο 1 An Y Cl Method A 2.67 minutes4; 372
25 0 1 Al NH 1 H l îAAnA A 1 Cl Method A 2.49 minutes5; 406
26 0 1 Ai NH I H L Αυ'-νΑο AJi 1 F fAn Y Cl Method A 3.00 minutes6; 408
27 0 1 'Ίΐ NH ιΑΑΎο □X 1 X? Method A 2.42 minutes4; 356
28 0 i Ίι NH 0XJ 1 cimAn Y Cl Method A 3.06 minutes6; 392
106
29 0 I H NH ΐίΆ^ίΑο oAJ 1 FÔ Ex 5; C5 Ή NMR (400 MHz, CD3OD) δ 7.95 (br d, J=4.9 Hz, 1H), 7.72 (br dd, J=9.3, 8.9 Hz, 1H), 7.25 (d, J=8.3 Hz, 1H), 7.17- 7.23 (m, 2H), 7.12-7.17 (m, 1H), 3.03 (s, 3H), 2.21 (s, 3H), 1.62 (s, 3H); 341.9
30 0 ι Ίι NH o-M 1 Ô Ex 5; C5 Ή NMR (400 MHz, CD3OD) δ 8.16-8.19 (m, 1H), 7.86-7.92 (m, 1H), 7.25 (d, J=8.3 Hz, 1H), 7.16-7.20 (m, 2H), 7.097.14 (m, 1H), 7.06 (d, J=8.2 Hz, 1H), 3.04 (s, 3H), 2.21 (s, 3H), 1.63 (s, 3H); 323.9
31 0 ι Ίι NH AAA> fx 1 Ex 5; C5 Ή NMR (600 MHz, DMSO-d6) δ 8.00 (d, J=4.8 Hz, 1H), 7.27 (d, J=8.3 Hz, 1H), 7.17-7.22 (m, 2H), 7.11 (br d, J=8 Hz, 1H), 2.87 (s, 3H), 2.44 (s, 3H), 2.14 (s, 3H), 1.48 (s, 3H); 372.1, 374.1
32 0 1 Al NH 1 II L \ AA 1 Y1 1 Ln-A Ex 1 & 2; C57 Ή NMR (400 MHz, CD3OD) δ 7.58 (dd, J=4.8, 1.6 Hz, 1 H), 7.21 (d, J=8.4 Hz, 1H), 7.08-7.13 (m, 2H), 7.01-7.05 (m, 2H), 5.28-5.49 (m, JHf=57.6 Hz, 1H), 4.33 (br ddd, J=19.4, 9.4, 5.8 Hz, 2H), 4.04 (br ddd, J=23.8, 9.3, 3.5 Hz, 2H), 3.02 (s, 3H), 2.19 (br s, 3H), 1.62 (s, 3H); 397.0
33 0 1 A] NH [AAiAo o-V ‘ v-°A Ex 1 & 2; C58 8.12 (brs, 1H), 7.81 (dd, J=4.9, 1.6 Hz, 1H), 7.63 (dd, J=7.8, 1.6 Hz, 1H), 7.06- 7.14 (m, 4H), 3.82-3.88 (m, 1H), 3.03 (s, 3H), 2.17 (s, 3H), 1.66 (s, 3H), 0.85-0.90 (m, 4H); 380.0
34 o 1 Ai NH AA o-V 1 oŸ 1 F C59,1011 8.35 (br s, 1 H), 7.93 (d, J=2.8 Hz, 1 H), 7.08-7.13 (m, 3H), 6.59 (d, J=5.8 Hz, 1H), 3.99 (s, 3H), 3.04 (s, 3H), 2.18 (s, 3H), 1.67 (s, 3H); 372.0
107
35 0 1 h NH □ r71 ζΤ Ex 1 & 2; C57 1H NMR (400 MHz, CD3OD) δ 7.55 (dd, J=4.8, 1.6 Hz, 1H), 7.19 (d, J=8.4 Hz, 1H), 7.09 (dd, J=7.8, 4.8 Hz, 1H), 7.047.06 (m, 1H), 6.97-7.02 (m, 2H), 4.01 (t, J=7.3 Hz, 4H), 3.02 (br s, 3H), 2.34 (quintet, J=7.3 Hz, 2H), 2.18 (s, 3H), 1.62 (s, 3H); 379.1
36 0 1 Al NH 1 H 1 AAnA fi AJ 1 fAy ù Ex 1 & 2; C57 1H NMR (400 MHz, CD3OD) δ 7.63 (dd, J=4.4, 2.1 Hz, 1H), 7.22 (d, J=8.3 Hz, 1H), 7.05-7.15 (m, 4H), 4.37 (t, J=12.0 Hz, 4H), 3.03 (s, 3H), 2.20 (br s, 3H), 1.62 (s, 3H); 415.0
37 0 i Ji NH ΛΛνΛ Ά C51210'11 1H NMR (400 MHz, CD3OD) δ 8.06 (dd, J=4.9, 1.8 Hz, 1 H), 7.80 (dd, J=7.5, 1.8 Hz, 1H), 7.22 (d, 4=8.3 Hz, 1H), 7.18 (dd, J=7.3, 5.0 Hz, 1H), 7.10-7.13 (m, 1H), 7.03-7.08 (m, 1H), 5.25-5.30 (m, 2H), 3.03 (s, 3H), 2.19 (br s, 6H), 1.63 (s, 3H), 364.1
38 0 1 Ai NH 1 J L aana AA Ex 3713 1H NMR (400 MHz, CD3OD), characteristic peaks: δ 7.97 (dd, 4=5.0, 1.9 Hz, 1H), 7.81-7.84 (m, 1H), 7.22 (d, J=8.3 Hz, 1H), 7.17 (dd, J=7.3, 5.1 Hz, 1H), 7.11-7.13 (m, 1H), 7.03-7.07 (m, 1 H), 3.04 (s, 3H), 2.20 (br s, 3H), 1.63 (s, 3H), 1.33 (d, 4=6.9 Hz, 6H); 366.0
39 0 1 Ai NH ΑΛΥ <A 1 A Ex 1 & 2; C514 1H NMR (400 MHz, CD3OD) δ 7.98 (d, J=5.3 Hz, 1H), 7.23 (d, J=8.3 Hz, 1H), 7.14 (brd, 4=2.3 Hz, 1H), 7.08 (brdd, 4=8.2, 2.2 Hz, 1 H), 6.87 (dd, 4=5.4, 1.4 Hz, 1H), 6.78-6.80 (m, 1H), 3.03 (s, 3H), 2.20 (brs, 3H), 1.95-2.03 (m, 1H), 1.62 (s, 3H), 1.12-1.18 (m, 2H), 0.84-0.90 (m, 2H); 364.0
108
40 0 I Ai NH 1 H l AaAo oV 1 An Ex 9 & 10; C2715 Ή NMR (600 MHz, DMSO-d6) δ 9.91 (br s, 1H), 7.64-7.68 (m, 1H), 7.32 (br d, J=7.9 Hz, 1H), 7.21 (d, J=8.3 Hz, 1H), 7.04-7.09 (m, 2H), 7.00 (br d, J=8.3 Hz, 1H), 2.86 (s, 3H), 2.12 (s, 3H), 1.47 (s, 3H); 340.1
41 0 C'A Ex 16 & 17; C4916 7.95 (d, J=4.9 Hz, 1H), 7.14 (br d, J=2.3 Hz, 1H), 7.10 (brdd, J=8.3, 2.2 Hz, 1H), 7.02 (d, J=8.3 Hz, 1H), 6.96 (dq, J=4.9, 0.6 Hz, 1H), 2.48 (br s, 3H), 2.07 (br s, 3H), 2.00 (s, 3H), 1.96 (s, 3H); 356.2, 358.2
42 0 1 H NH (fAANAo OA> 1 fit* fi Ex 5; C5 Ή NMR (400 MHz, CD3OD) δ 7.77-7.79 (m, 1H), 7.22 (d, J=8.3 Hz, 1H), 7.12 (br d, J=2 Hz, 1H), 7.07 (dd, J=8, 2 Hz, 1H), 6.64 (s, 1H), 3.93 (s, 3H), 3.03 (s, 3H), 2.19 (br s, 3H), 2.13 (br s, 3H), 1.62 (s, 3H); 368.0
43 0 Βγ^Αν Ex16& 17; C49 8.13 (dd, J=4.8, 1.7 Hz, 1H), 7.97 (dd, J=7.7, 1.7 Hz, 1H), 7.16 (brd, J=2.2 Hz, 1H), 7.12 (brdd, J=8.2, 2.3 Hz, 1H), 7.03 (br d, J=8.2 Hz, 1H), 6.96 (dd, J=7.7, 4.8 Hz, 1H), 2.09 (s, 3H), 2.00 (s, 3H), 1.96 (s, 3H); 386.1,388.0
44 0 Al NH Aa A 1 CI'^J Ex 5; C317 8.07 (dd, J=4.9, 1.5 Hz, 1H), 7.97 (dd, J=7.7, 1.4 Hz, 1 H), 7.36 (br AB quartet, JAB=8.6 Hz, Avab=28.4 Hz, 4H), 7.17 (dd, J=7.7, 4.8 Hz, 1H), 3.08 (s, 3H), 1.68 (s, 3H); 343.9
45 o I Ai NH AAa> 0A ' F^°^An (+) Ex 5; C27, C3618'19 8.29 (br s, 1H), 8.06 (dd, J=4.8, 1.7 Hz, 1H), 7.63-7.66 (m, 1H), 7.16-7.18 (m, 1H), 7.11-7.15 (m, 2H), 7.11 (dd, J=7.9, 4.8 Hz, 1H), 6.70 (t, JHf=73.5 Hz, 1H), 3.05 (s, 3H), 2.20 (br s, 3H), 1.68 (s, 3H); 390.1
109
46 0 1 '''Il nh Vif Ex16& 17; C4920 10.15 (brs, 1H), 8.02 (dd, 4=5.0, 1.8 Hz, 1H), 7.28-7.32 (m, 1H), 7.11 (br d, 4=2 Hz, 1H), 7.07 (brdd, 4=8, 2 Hz, 1H), 6.98-7.02 (m, 2H), 2.18-2.26 (m, 1H), 2.07 (s, 3H), 1.99 (s, 3H), 1.95 (s, 3H), 1.01-1.07 (m, 2H), 0.74-0.79 (m, 2H); 348.0
47 1 0 1 il NH r, F 1 fAÔ Ex 12; C17, C33 1H NMR (600 MHz, DMSO-ck) δ 8.338.36 (m, 1H), 8.13 (brd, 4=7 Hz, 1H), 7.30-7.34 (m, 2H), 7.28 (t, 4HF=54 Hz, 1H), 7.23-7.25 (m, 1H), 7.17 (brd, 4=8.2 Hz, 1H), 2.83-2.86 (m, 3H), 2.16 (s, 3H), 2.01-2.09 (m, 1H), 1.70-1.78 (m, 1H), 0.82 (t, 4=7.3 Hz, 3H); 388.1
48 0 'Û Ex 16 & 17; C49 10.43 (br s, 1 H), 8.19 (dd, 4=7.6, 1.6 Hz, 1H), 8.14 (dd, 4=4.8, 1.6 Hz, 1H), 7.14- 7.16 (m, 1H), 7.09-7.13 (m, 1H), 7.03 (d, half of AB quartet, 4=8.3 Hz, 1 H), 6.82 (dd, 4=7.6, 4.9 Hz, 1H), 2.09 (br s, 3H), 1.99 (s, 3H), 1.95 (s, 3H); 433.9
49 0 1 '>i NH iVr O-Λ A -b Ex 16 & 17; C4921 10.64 (br s, 1H), 8.12 (br d, 4=5 Hz, 1H), 7.78 (br d, 4=7 Hz, 1H), 7.12 (dd, 4=7, 5 Hz, 1H), 7.06-7.09 (m, 1H), 7.02-7.06 (m, 1H), 7.00 (d, half of AB quartet, 4=8.2 Hz, 1H), 5.11 (dd, 4=8.4, 5.9 Hz, 2H), 4.93 (dd, 4=7.0, 6.3 Hz, 2H), 4.574.67 (m, 1H), 2.07 (s, 3H), 1.99 (s, 3H), 1.95 (s, 3H); 364.0
50 0 1 Ui NH AV° Τώ Ex 9 & 10; C2722 8.21 (brs, 1H), 7.93 (d, 4=5.0 Hz, 1H), 7.15-7.17 (m, 1H), 7.12-7.14 (m, 2H), 6.99 (br d, 4=5.0 Hz, 1 H), 6.75 (t, 4hf=75.3 Hz, 1 H), 3.05 (s, 3H), 2.43 (br s, 3H), 2.20 (br s, 3H), 1.67 (s, 3H); 404.2
110
51 0 I F3C'0^jAjl Ex16& 17; C49 1H NMR (400 MHz, CD3OD) δ 8.11 (br dd, J=4.8,1.3 Hz, 1H), 7.88 (br d, J=7.8 Hz, 1H), 7.24 (dd, J=7.9, 4.9 Hz, 1H), 7.15-7.19 (m, 1H), 7.08-7.15 (m, 2H), 2.09 (s, 3H), 2.01 (s, 3H), 1.90 (s, 3H); 392.1
52 0 i o NH /yAjAq cY ' FC'°U Ex 9 & 10; C27 1H NMR (400 MHz, CD3OD) δ 8.12 (br d, J=5 Hz, 1H), 7.90 (br d, J=8 Hz, 1H), 7.23-7.29 (m, 2H), 7.19-7.22 (m, 1H), 7.15 (brd, J=8 Hz, 1H), 3.03 (s, 3H), 2.22 (s, 3H), 1.63 (s, 3H); 408.1
53 0 Υνη αΛνΛ 0 V ' YU Ex 9 & 10; C36 1H NMR (400 MHz, CD3OD) δ 8.01 (dd, J=4.9, 1.6 Hz, 1H) 7.76 (br d, J=8 Hz, 1 H), 7.36 (br AB quartet, JAb=8-8 Hz, ΔνΑΒ=23.2 Hz, 4H), 7.22 (dd, J=7.9, 4.9 Hz, 1H), 6.97 (t, JHf=73.5 Hz, 1H), 3.08 (s, 3H), 1.68 (s, 3H); 376.0
54 0 I Yl NH I H L AYnY o-V ' clj (.) Ex 9 & 10; C2723 11.45 (brs, 1H), 8.15 (dd, J=4.8, 1.7 Hz, 1H), 8.09 (dd, J=7.8, 1.7 Hz, 1H), 7.29 (d, J=8.3 Hz, 1H), 7.21-7.25 (m, 2H), 7.15 (brdd, J=8.3, 2.2 Hz, 1H), 2.87 (s, 3H), 2.15 (brs, 3H), 1.48 (s, 3H); 358.1, 360.2
55 0 1 Yl NH 1 II L fY/Y οY 1 CI(+) Ex 9 & 10; C2723 1H NMR (400 MHz, DMSO-d6) δ 11.45 (brs, 1H), 8.15 (dd, J=4.8, 1.7 Hz, 1H), 8.09 (dd, J=7.8, 1.7 Hz, 1H), 7.29 (d, J=8.3 Hz, 1H), 7.21-7.25 (m, 2H), 7.15 (br dd, J=8, 2.5 Hz, 1H), 2.87 (s, 3H), 2.15 (brs, 3H), 1.48 (s, 3H); 358.1, 360.2
56 0 CN Y| NH YYYo F Y ' Ex 9 & 10; C2624 2.56 minutes25; 385.1
111
57 0 I Al NH A1 CI^^N Ex16& 17; C49 10.88 (brs, 1H), 8.09 (dd, J=4.8, 1.6 Hz, 1H), 7.81 (dd, J=7.7, 1.6 Hz, 1H), 7.14- 7.17 (m, 1 H), 7.12 (brdd, J=8, 2 Hz, 1H), 7.01-7.06 (m, 2H), 2.09 (s, 3H), 1.99 (s, 3H), 1.95 (s, 3H); 342.2
58 0 I A NH V F Ex 16 & 17; C49, C36 10.32 (brs, 1H), 8.06 (dd, J=4.8, 1.6 Hz, 1H), 7.63 (brd, J=8 Hz, 1H), 7.13-7.16 (m, 1H), 7.07-7.13 (m, 2H), 7.03 (d, J=8.4 Hz, 1H), 6.71 (t, JHf=73.7 Hz, 1H), 2.08 (s, 3H), 1.99 (s, 3H), 1.94 (s, 3H); 374.2
59 0 I Al NH Av ο ο/Σ H2NA) Ex 16 & 17; C4926 1H NMR (400 MHz, CD3OD) δ 8.40 (dd, J=7.5, 1.9 Hz, 1H), 8.24 (dd, J=5.0, 1.9 Hz, 1H), 7.27 (dd, J=7.5, 4.9 Hz, 1H), 7.24-7.26 (m, 1H), 7.14-7.21 (m, 2H), 2.10 (s, 3H), 2.07 (br s, 3H), 1.95 (br s, 3H); 351.1
60 0 I ο ο>Σ Ex 16 & 1727 1H NMR (400 MHz, CD3OD) δ 8.35 (dd, J=7.6, 1.9 Hz, 1H), 8.29 (dd, J=4.9, 2.0 Hz, 1H), 7.24 (dd, J=7.6, 4.9 Hz, 1H), 7.13-7.16 (m, 1 H), 7.11 (d, half of AB quartet, J=8.0 Hz, 1H), 7.08 (brdd, half of ABX pattern, J=8.3, 2.0 Hz, 1H), 3.93 (s, 3H), 2.08 (s, 3H), 2.00 (s, 3H), 1.90 (s, 3H); 366.1
61 0 l Vi NH f I Ex 1 & 2; C492829 10.17 (brs, 1H), 8.02 (brd, J=5 Hz, 1H), 6.94-7.06 (m, 3H), 6.65 (d, J=6.0 Hz, 1H), 3.93 (s, 3H), 2.20 (s, 3H), 2.05 (s, 3H), 1.98 (s, 3H), 1.93 (s, 3H); 352.0
62 0 I ï NH V1 F3Cjj^n Ex 16 & 17; C4930 10.46 (brs, 1H), 8.14 (d, J=4.8 Hz, 1H), 7.10-7.14 (m, 1H), 7.05 (AB quartet, downfield doublet is broadened, JAB=8.5 Hz, ΔνΑΒ=23.3 Hz, 2H), 6.95 (br d, J=5.0 Hz, 1H), 2.56-2.61 (m, 3H), 2.08 (s, 3H), 1.99 (s, 3H), 1.95 (br s, 3H); 390.3
112
63 0 F Εχ16& 17; C4931 10.3 (ν br s, 1 Η), 8.17 (br d, J=6.0 Hz, 1H), 7.17 (t, JHf=53.7 Hz, 1H), 7.12-7.14 (m, 1H), 7.09 (brdd, J=8.2, 2.4 Hz, 1H), 7.01 (d, J=8.3 Hz, 1H), 6.72 (br d, J=6.0 Hz, 1 H), 4.00 (s, 3H), 2.08 (br s, 3H), 1.99 (s, 3H), 1.95 (s, 3H); 388.2
64 0 ι AfrH 0Λα τ faA 1 Εχ 1 & 2; C493233 8.19 (d, J=5.8 Hz, 1H), 7.12 (brd, J=2 Hz, 1 H), 7.08 (br dd, half of ABX pattern, J=8, 2 Hz, 1H), 7.01 (d, half of AB quartet, J=8.4 Hz, 1H), 6.75 (d, J=5.8 Hz, 1H), 4.00 (s, 3H), 2.07 (s, 3H), 2.02 (s, 3H), 1.97 (s, 3H); 406.2
65 0 ι Ύ, F3Cx> () Εχ 1 & 2; C493435 1H NMR (400 MHz, CD3OD) δ 8.32-8.35 (m, 1H), 8.16-8.20 (m, 1H), 7.28 (ddq, J=7.6, 5.0, 0.8 Hz, 1H), 7.17-7.19 (m, 1H), 7.10-7.16 (m, 2H), 2.09 (br s, 3H), 2.00 (s, 3H), 1.90 (s, 3H); 376.236
66 0 1 Ai ΝΗ ιγ Ρ3°'Χα (+) Εχ 1 & 2; C493435 1H NMR (400 MHz, CD3OD) δ 8.32-8.35 (m, 1H), 8.16-8.20 (m, 1H), 7.28 (ddq, J=7.6, 5.0, 0.7 Hz, 1H), 7.17-7.20 (m, 1H), 7.10-7.16 (m, 2H), 2.09 (brs, 3H), 2.00 (s, 3H), 1.90 (s, 3H); 376.236
67 0 Y) ΝΗ ζ* ,ό Εχ 16 & 17 1H NMR (400 MHz, CD3OD) δ 8.03 (br d, J=5.2 Hz, 1H), 7.21-7.29 (m, 4H), 7.017.04 (m, 1H), 6.89-6.90 (m, 1H), 2.40 (br s, 3H), 2.07 (s, 3H), 1.96 (s, 3H); 308.1
68 0 ι Α| ΝΗ Ύ Εχ 18; C53 8.50 (brd, J=5 Hz, 1H), 8.18 (brs, 1H), 7.94 (brd, J=8 Hz, 1H), 7.54-7.56 (m, 1H), 7.51 (brd, J=8 Hz, 1H), 7.21 (brdd, J=8, 5 Hz, 1 H), 7.14 (d, J=7.9 Hz, 1H), 3.04 (s, 3H), 2.21 (br s, 3H), 1.67 (s, 3H); 408.1
113
69 0 ''Ίι NH AJ 1 Ai- Ex 13; C37, 1H NMR (400 MHz, CD3OD) δ 7.95 (dd, J=4.9, 1.9 Hz, 1H), 7.49 (ddd, J=7.6, 1.9, 0.5 Hz, 1H), 7.30 (br AB quartet, JAB=8.8 Hz, ΔνΑΒ=44.0 Hz, 4H), 7.12 (ddd, J=7.5, 4.9, 0.5 Hz, 1H), 3.08 (s, 3H), 2.16-2.24 (m, 1H), 1.68 (s, 3H), 1.00-1.05 (m, 2H), 0.77-0.82 (m, 2H); 350.2
70 0 Â F Ex 1 & 2; C493940 2.80 minutes25; 340.3
71 Ai- Ex 1 & 2; P1, C641 3.13 minutes6; 348
72 Ά χιΑ cl>C^N Cl Ex 1 & 2; P141 3.34 minutes42; 390
73 p AA A· Ex 1 & 2; P141 3.17 minutes6; 356
74 i A At CI^N Ex 1 & 2; P141 3.03 minutes6; 342
114
75 ,'Â .A Ex 1 & 2; P1, C3641 2.97 minutes6; 374
76 0 CI'YXN γ Cl Ex 1 & 2; P241 3.06 minutes42; 390
77 0 FCY Ex 1 & 2; P241 2.95 minutes6; 376
78 0 °Â Ex 1 & 2; P241 2.96 minutes6; 356
79 0 | ΝΛΝ Ex 1 & 2; P2, C641 2.91 minutes6; 348
80 0 Clt? Ex 1 & 2; P241 2.81 minutes6; 342
115
Ex 12;
1H NMR (600 MHz, DMSO-de) δ 8.31 (d,
1H), 8.11 (d, 1H), 7.35 (d, 1H), 7.35 (dd,
1H), 7.30 (t, 1H), 7.17 (d, 1H), 7.11 (br dd, 1H), 2.65 (m, 1H), 2.31 (m, 1H), 2.16 (s, 3H), 1.48 (s, 3H), 0.4-0.6 (m, 4H).
1. In this case, reaction with the chloropyridine was carried out using tris(dîbenzylideneacetone)dipalladium(0), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos) and potassium terï-butoxide in toluene at elevated température.
2. Compound C11 was reacted with (4-hydroxyphenyl)boronic acid, under the conditions described for the synthesis of C12 in Example 5, to provide 1 -ethyl-6-(4-hydroxyphenyl)-5methyl-3-{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1H,3/-/)-dione.
3. Compound C18 was reacted with (4-hydroxyphenyl)boronic acid, under the conditions described for the synthesis of C19 in Example 6, to provide 5-ethyl-6-(4-hydroxyphenyl)-1methyl-3-{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1H,3/4)-dione.
4. Conditions for analytical HPLC. Column: Waters XBridge C18, 2.1 x 50 mm, 5 pm. Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile. Gradient: 0 to 0.5 minutes, 5% B; 0.5 to 3.4 minutes, linear from 5% to 100% B. Flow rate: 0.8 mL/minute.
5. Conditions for analytical HPLC. Column: Waters XBridge C18, 2.1 x 50 mm, 5 pm. Mobile phase A: 0.0375% trifluoroacetic acid in water; Mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile. Gradient: 0 to 0.5 minutes, 25% B; 0.5 to 3.5 minutes, linear from 25% to 100% B.
Flow rate: 0.8 mUminute.
6. Identical to footnote 5, except that the gradient used was: 0 to 0.6 minutes, linear from 1% to 5% B; 0.6 to 4.0 minutes, linear from 5% to 100% B.
7. The requisite 2-chloropyridine was prepared via reaction of 2-chloro-3-iodopyridine with a sait 20 of the appropriate azetidine, using palladium(ll) acetate, 1,1'-binaphthalene-2,2'- diylbis(diphenylphosphane) (BINAP) and césium carbonate in toluene at elevated température.
8. Reaction of 2-chloropyridin-3-ol with bromocyclopropane, in the presence of césium carbonate in Λ/,/V-dimethylacetamide at 150 °C, afforded 2-chloro-3-(cyclopropyloxy)pyridine.
9. Reaction of 2-chloro-5-fluoropyridin-4-ol with iodomethane and silver carbonate provided 225 chloro-5-fluoro-4-methoxypyridine.
10. The reaction between phénol C5 and the chloropyridine was effected via reaction with copper(l) iodide and césium carbonate in pyridine at 120 °C.
11. Deprotection was carried out according to Example 5.
12. Reaction of ethyl 2-chloropyridine-3-carboxylate with méthylmagnésium iodide yielded 2-(230 chloropyridin-3-yl)propan-2-ol.
13. Olefin réduction was effected via hydrogénation using palladium on carbon and N,Ndiisopropylethylamine in methanol.
116 φ 14. In this case, reaction with the chloropyridine was carried out using 4,5bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos) in place of di-terf-butyl[3,4,5,6tetramethyl-2',4',6'-tri(propan-2-yl)biphenyl-2-yl]phosphane.
15.1-(2-Chîoropyridin-3-yl)ethanone was converted to 2-chloro-3-(1,1-difluoroethoxy)pyridine using the method of D. B. Horne et al., Tetrahedron Lett. 2009, 50, 5452-5455. Upon deprotection, the difluoroethoxy group was also cleaved.
16. In this case, césium fluoride was used in place of césium carbonate in the reaction of the chloropyridine with phénol C49.
17. Compound C3 was reacted with (4-hydroxyphenyl)boronic acid, under the conditions described for préparation of C4 in Examples 1 and 2, to afford 6-(4-hydroxyphenyl)-1,5dimethyl-3-{[2-(trimethylsilyl)ethoxy]methyl}pyrimidine-2,4(1/7,3/-/)-dione.
18. In this case, the deprotection was carried out in trifluoroacetic acid at 100 °C.
19. The racemic product was separated into its atropenantiomers via high-performance liquid chromatography (Column: Chiral Technologies, Chiralpak AD-H, 5 pm; Gradient: éthanol in heptane). This Example was the first-eluting atropenantiomer, and exhibited a positive (+) rotation.
20. Compound C49 was reacted with 2-chloro-3-iodopyridine to afford 5-{4-[(3-iodopyridin-2yl)oxy]-2-methylphenyl}-4,6-dimethyl-2-(tetrahydro-2H-pyran-2-yl)pyridazin-3(2/7)-one; subséquent Suzuki reaction with cyclopropylboronic acid provided 5-{4-[(3-cyclopropylpyridin-2- yl)oxy]-2-methylphenyl}-4,6-dimethyl-2-(tetrahydro-2/7-pyran-2-yl)pyridazin-3(2H)-one.
Deprotection in this case was carried out with trifluoroacetic acid rather than hydrochloric acid.
21. The requisite 2-chloro-3-(oxetan-3-yl)pyridine was prepared from (2-chloropyridin-3yl)boronic acid using the method reported by M. A. J. Duncton et al., Org. Lett. 2008, 10, 32593262.
22. 2-Chloro-3-(difluoromethoxy)-4-methylpyridine was prepared from 2-chloro-4-methylpyridin-
3- ol using conditions reported by L. F. Frey et al., Tetrahedron 2003, 59, 6363-6373.
23. The racemic product was separated into its component atropenantiomers using chiral séparation. Conditions for analytical HPLC. Column: Chiralpak AD-H, 20 x 250 mm; Mobile phase A: Heptane; Mobile phase B: Ethanol; Gradient: 5.0% to 95% B, linear over 12 minutes;
Flow rate: 28 mL/minute.The first-eluting atropenantiomer, which exhibited a positive (+) rotation, was designated as Example 55; the second-eluting one, which gave a négative (-) rotation, was designated as Example 54.
24. The requisite 2-[1-(3,4-dimethoxybenzyl)-3,5-dimethyl-2,6-dioxo-1,2,3,6-tetrahydropyrimidin-
4- yl]-5-hydroxybenzonitrile was prepared via reaction of C26 with 5-hydroxy-2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile, mediated by chloro(2-dicyclohexylphosphino2',6'-dimethoxy-1 ,T-biphenyl)[2-(2'-amino-1,1 '-biphenyl)]palladium(ll) and potassium phosphate.
117 φ 25. Conditions for analytical HPLC. Column: Waters Atlantis dC18, 4.6 x 50 mm, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 5.0% to 95% B, linear over 4.0 minutes; Flow rate: 2 mL/minute.
26. Reaction of C49 with methyl 2-chloropyridine-3-carboxylate afforded methyl 2-(4-(3,55 dimethyl-6-oxo-1 -(tetrahydro-2/-/-pyran-2-yl)-1,6-dihydropyridazin-4-yl]-3methylphenoxy}pyridine-3-carboxylate; the ester group was converted to an amide via subjection to ammonium hydroxide in methanol at elevated température, to provide 2-(4-(3,5dimethyl-6-oxo-1-(tetrahydro-2H-pyran-2-yl)-1,6-dihydropyridazin-4-yl]-3methylphenoxy}pyridine-3-carboxamide.
27. Methyl 2-{4-[3,5-dimethyl-6-oxo-1 -(tetrahydro-2H-pyran-2-yl)-1,6-dihydropyridazin-4-yl]-3methylphenoxy}pyridine-3-carboxylate (see footnote 26) was deprotected to afford this Example.
28. 2,4-Dichloro-3-methylpyridine was converted to 2-chloro-4-methoxy-3-methylpyridine via reaction with sodium hydride / methanol.
29. In this case, the deprotection was carried out with trifluoroacetic acid in dichloromethane at room température.
30. The requisite 2-chloro-4-methyl-3-(trifluoromethyl)pyridine was prepared via reaction of 2chloro-3-iodo-4-methylpyridine with methyl difluoro(fluorosulfonyl)acetate and copper(l) iodide in Λ/,/V-dimethylformamide at 90 °C.
31. Reaction of 2-chloro-4-methoxypyridine-3-carbaldehyde with (diethylamino)sulfur trifluoride afforded 2-chloro-3-(difluoromethyl)-4-methoxypyridine.
32. Reaction of 2,4-dichloro-3-iodopyridine with sodium methoxide in methanol provided 2chloro-3-iodo-4-methoxypyridine; this material was converted to 2-chloro-4-methoxy-3(trifluoromethyl)pyridine as described in footnote 30.
33. The final deprotection was carried using hydrogen chloride in methanol, at room température.
34. Deprotection was carried out using the method described in Examples 16 and 17.
35. Séparation of atropenantiomers was carried out via supercritical fluid chromatography (Column: Chiral Technologies, Chiralpak AS-H, 5 pm; Eluent: 85:15 carbon dioxide / methanol).
The first-eluting atropenantiomer exhibited a positive (+) rotation, and was designated as
Example 66. The second-eluting atropenantiomer displayed a négative (-) rotation, and was designated as Example 65.
36. In this case, mass spectrometry data was obtained on the racemate, prior to séparation of the atropenantiomers.
37. Compound C37 was reacted with (4-hydroxyphenyl)boronic acid, using the method described for préparation of C4 in Examples 1 and 2, to afford 3-[(benzyloxy)methyl]-6-(4hydroxyphenyl)-1,5-dimethylpyrimidine-2,4(1 H,3/V)-dione.
118 φ 38. Conditions for reaction of the phénol with the chloropyridine were similar to those used for synthesis of C7 in Examples 1 and 2.
39. After the coupling reaction, the reaction mixture was partitioned between water and ethyl acetate. The organic layer was dried with sodium sulfate and concentrated in vacuo; this material was deprotected with hydrogen chloride in 1,4-dioxane.
40. Purification was effected via reversed phase high-performance liquid chromatography. Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 30% to 50% B.
41. Purification was carried out via reversed phase high-performance liquid chromatography using an appropriate gradient in one of the following Systems: a) Column: Agela Durashell C18, 5 pm; Mobile phase A: ammonium hydroxide in water, pH 10; Mobile phase B: acetonitrile; b) Column: Phenomenex Gemini, 10 pm; Mobile phase A: ammonium hydroxide in water, pH 10; Mobile phase B: acetonitrile; c) Column: Phenomenex Gemini, 8 pm; Mobile phase A: 0.225% formic acid in water; Mobile phase B: acetonitrile; d) Column: YMS C18, 5 pm; Mobile phase A:
ammonium hydroxide in water, pH 10; Mobile phase B: acetonitrile.
42. Conditions for analytical HPLC. Column: Waters XBridge C18, 2.1 x 50 mm, 5 pm. Mobile phase A: 0.0375% trifluoroacetic acid in water; Mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile. Gradient: 0 to 0.5 minutes, 10% B; 0.5 to 4.0 minutes, linear from 10% to 100% B. Flow rate: 0.8 mL/minute.
43. Compound C33 was reacted with 6-bromo-3-(3,4-dimethoxybenzyl)-1-cyclopropyl-5methylpyrimidine-2,4(1H,3H)-dione, using the method described for Example 12, to yield Example 81. The intermediate, 6-bromo-3-(3,4-dimethoxybenzyl)-1-cyclopropyl-5methylpyrimidine-2,4(1H,3H)-dione, was prepared from commercially available 1-cyclopropyl urea following the methods described for the préparation of C10 and C34.
Example AA: Human D1 Receptor Binding Assay and Data
The affinity of the compounds described herein was determined by compétition binding assays similar to those described in Ryman-Rasmussen et al., “Differential activation of adenylate cyclase and receptor internalization by novel dopamine D1 receptor agonists”, Molecular Pharmacology 68(4):1039-1048 (2005). This radioligand binding assay used [3HJ30 SCH23390, a radiolabeled D1 ligand, to evaluate the ability of a test compound to compete with the radioligand when binding to a D1 receptor.
D1 binding assays were performed using over-expressing LTK human cell lines. To détermine basic assay parameters, ligand concentrations were determined from saturation binding studies where the Kd for [3H]-SCH23390 was found to be 1.3 nM. From tissue concentration curve studies, the optimal amount of tissue was determined to be 1.75 mg/mL per well plate using 0.5 nM of [3H]-SCH23390. These ligand and tissue concentrations were used in time course studies to détermine linearity and equilibrium conditions for binding.
Binding was at equilibrium with the specified amount of tissue in 30 minutes at 37 °C. From
119 these parameters, K, values were determined by homogenizing the specified amount of tissue for each species in 50 mM Tris (pH 7.4 at 4 °C) containing 2.0 mM MgCI2 using a Polytron and spun in a centrifuge at 40,000 x g for 10 minutes. The pellet was resuspended in assay buffer [50 mM Tris (pH 7.4@ RT) containing 4 mM MgSO4 and 0.5 mM EDTA]. Incubations were initiated by the addition of 200 pL of tissue to 96-well plates containing test drugs (2.5 pL) and 0.5 nM [3H]-SCH23390 (50 pL) in a final volume of 250 pL. Non-specific binding was determined by radioligand binding in the presence of a saturating concentration of (+)Butaclamol (10 pM), a D1 antagonist. After a 30 minute incubation period at 37 °C, assay samples were rapidly filtered through Unifilter-96 GF/B PEI-coated filter plates and rinsed with 50 mM Tris buffer (pH 7.4 at 4 °C). Membrane bound [3H]-SCH23390 levels were determined by liquid scintillation counting of the filterplates in Ecolume. The IC50 value (concentration at which 50% inhibition of spécifie binding occurs) was calculated by linear régression of the concentration-response data in Microsoft Excel. K, values were calculated according to the Cheng-Prusoff équation:
Ki =____[Cso______
1+ ([L]//Q where [L] = concentration of free radioligand and Kd = dissociation constant of radioligand for D1 receptor (1.3 nM for [3H]-SCH23390).
Example BB: D1 cAMP HTRF Assay and Data
The D1 cAMP (Cyclic Adenosine Monophosphate) HTRF (Homogeneous TimeResolved Fluorescence) Assay used and described herein is a compétitive immunoassay between native cAMP produced by cells and cAMP labeled with XL-665. This assay was used to détermine the ability of a test compound to agonize (including partially agonize) D1. A Mab anti-cAMP labeled Cryptate visualizes the tracer. The maximum signal is achieved if the samples do not contain free cAMP due to the proximity of donor (Eu-cryptate) and acceptor (XL665) entities. The signal, therefore, is inversely proportional to the concentration of cAMP in the sample. A time-resolved and ratiometric measurement (em 665 nm/em 620 nm) minimizes the interférence with medium. cAMP HTRF assays are commercially available, for example, from Cisbio Bioassays, IBA group.
Materials and Methods
Materials: The cAMP Dynamic kit was obtained from Cisbio International (Cisbio
62AM4PEJ). Multidrop Combi (Thermo Scientific) was used for assay additions. An EnVision (PerkinElmer) reader was used to read HTRF.
Cell Cuture: A HEK293T/hD1#1 stable cell line was constructed internally (Pfizer Ann
Arbor). The cells were grown as adhèrent cells in NuncT500 flasks in high glucose DMEM (Invitrogen 11995-065), 10% fêtai bovine sérum dialyzed (Invitrogen 26400-044), 1x MEM
NEAA (Invitrogen 1140, 25 mM HEPES (Invitrogen 15630), 1x Pen/Strep (Invitrogen 15070120
063) and 500 pg/mL Genenticin (Invitrogen 10131-035) at 37 °C and 5% CO2. At 72 or 96 hours post-growth, cells were rinsed with DPBS, and 0.25% Trypsin-EDTA was added to dislodge the cells. Media was then added and cells were centrifuged and media removed. The cell pellets were re-suspended in Cell Culture Freezing Medium (Invitrogen 12648-056) at a density of 4e7 cells/mL. One mL aliquots of the cells were made in Cryo-vials and frozen at -80 °C for future use in the D1 HTRF assay.
D1 cAMP HTRF assay procedure: Frozen cells were quickly thawed, re-suspended in 50 mL warm media and allowed to sit for 5 min prior to centrifugation (1000 rpm) at room température. Media was removed and cell pellet was re-suspended in PBS/0.5 μΜ IBMX generating 2e5 cells/mL. Using a Multidrop Combi, 5 pL cells/well was added to the assay plate (Greiner 784085), which already contained 5 pL of a test compound. Compound controls [5 pM dopamine (final) and 0.5% DMSO (final)] were also included on every plate for data analysis. Cells and compounds were incubated at room température for 30 min. Working solutions of CAMP-D2 and anti-cAMP-cryptate were prepared according to Cisbio instructions. Using Multidrop, 5 pL cAMP-02 working solution was added to the assay plate containing the test compound and cells. Using Multidrop, 5 pL anti-cAMP-cryptate working solutions was added to assay plate containing test compound, cells and CAMP-D2. The assay plate was incubated for 1 hour at room température. The assay plate was read on an EnVision plate reader using Cisbio recommended settings. A cAMP standard curve was generated using cAMP stock solution provided in the Cisbio kit.
Data Analysis: Data analysis was done using computer software. Percent effects were calculated from the compound controls. Ratio EC50 was determined using the raw ratio data from the EnVision reader. The cAMP standard curve was used in an analysis program to détermine cAMP concentrations from raw ratio data. cAMP EC50 was determined using the calculated cAMP data.
Table 2. Biological Data and Compound Name for Examples 1 - 81.
Example Number Human D1 Receptor Binding, Kj (nM); Géométrie mean of 2 - 5 determinati ons (unless otherwise indicated) Compound Name
1 15.3 (+)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-
121
dimethylpyrimidine-2,4(1/7,3/-/)-dione
2 3.11 (-)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3/7)-dione
3 40.6a (-)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3H)-dione
4 118a (+)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/7,3H)-dione
5 58.0a 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1-ethyl-5- methylpyrimidine-2,4(1H,3/-/)-dione
6 33.1a 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-5-ethyl-1- methylpyrimidine-2,4(1H,3/-/)-dione
7 8.54 (-)-1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2- yl]oxy}phenyl)pyrimidine-2,4(1/-/,3H)-dione
8 21.0 (+)-1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2- yl]oxy}phenyl)pyrimidine-2,4(1H,3H)-dione
9 120a (+)-6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3/-/)-dione
10 82.3a (-)-6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/7,3H)-dione
11 10.9a 6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]phenyl}-1,5dimethylpyrimidine-2,4(1/-/,3/-/)-dione
12 55.1 6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1-ethyl- 5-methylpyrimidine-2,4(1H,3H)-dione
13 6.91 (-)-6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-1,5- dimethylpyrimidine-2,4(1H,3/-/)-dione
14 17.5 (-)-6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1,5- dimethylpyrimidine-2,4(1/7,3/-/)-dione
15 54.7 (+)-6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-1,5- dimethylpyrimidine-2,4(1/7,3/-/)-dione
16 44.3 (+)-5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6- dimethylpyridazin-3(2H)-one
17 59.1 (-)-5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6- dimethylpyridazin-3(2H)-one
18 35.7 6-{4-[(3-chloropyridin-2-yl)sulfanyl]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1 H,3H)-dione
19 12.9a 1,5-dimethyl-6-(7-{[3-(trifluoromethyl)pyridin-2-yl]oxy}-1/7-indol-4- yl)pyrimidine-2,4(1 H,3H)-dione
122
20 52.7 6-{4-[(3-ethylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3/-/)-dione
21 314.2a 6-{4-[(3-chloropyridin-2-yl)oxy]phenyl}-1-ethyl-5-methylpyrimidine- 2,4(1H,3H)-dione
22 82. T 6-{4-[(3-chloropyridin-2-yl)oxy]phenyl}-5-ethyl-1-methylpyrimidine- 2,4(1 H,3/-/)-dione
23 45.9a 2-(4-(3,5-dimethyl-2,6-dioxo-1,2,3,6-tetrahydropyrimidin-4-yl)-3- methylphenoxy]-4-methylpyridine-3-carbonitrile
24 92.1a 6-{4-[(5-chloro-3-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3/-f)-dione
25 38.5a 6-(4-((3,5-dichloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1M3H)-dione
26 117a 6-(4-{[5-chloro-3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)- 1,5-dimethylpyrimidine-2,4(1 H,3H)-dione
27 36.2a 6-{4-[(3-fluoro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/7,3/-/)-dione
28 88.9a 6-(4-((3,5-dichloropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/-/,3/-/)-dione
29 489 6-{4-[(3-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/-/,3H)-dione
30 156a 1,5-dimethyl-6-[2-methyl-4-(pyridin-2-yloxy)phenyl]pyrimidine- 2,4(1/7,3H)-dione
31 3.61 6-{4-((3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3/-/)-dione
32 650a 6-(4-{[3-(3-fluoroazetidin-1-yl)pyridin-2-yl]oxy}-2-methylphenyl)- 1,5-dimethylpyrimidine-2,4(1 H,3H)-dione
33 224a 6-(4-{[3-(cyclopropyloxy)pyridin-2-yl]oxy}-2-methylphenyl)-1,5- dimethylpyrimidine-2,4(1H,3/-/)-dione
34 120a 6-{4-[(5-fluoro-4-methoxypyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3H)-dione
35 246a 6-(4-{[3-(azetidin-1-yl)pyridin-2-yl]oxy}-2-methylphenyl)-1,5- dimethylpyrimidine-2,4(1/-/,3H)-dione
36 826a 6-(4-((3-(3,3-difluoroazetidin-1-yl)pyridin-2-yl]oxy}-2methylphenyl)-1,5-dimethylpyrimidine-2,4(1 /-/,3H)-dione
37 8.42a 1,5-dimethyl-6-(2-methyl-4-{[3-(prop-1 -en-2-yl)pyridin-2- yl]oxy}phenyl)pyrimidine-2,4(1/-/,3H)-dione
38 31.1a 1,5-dimethyl-6-(2-methyl-4-{[3-(propan-2-yl)pyridin-2-
123
yl]oxy}phenyl)pyrimidine-2,4(1H,3/-/)-dione
39 15.0a 6-{4-[(4-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/7,3H)-dione
40 82.7a 6-{4-[(3-hydroxypyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/-/,3/7)-dione
41 5.41a 5-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2H)-one
42 187a 6-{4-[(4-methoxy-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1/7,3/-/)-dione
43 9.65 5-{4-[(3-bromopyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2H)-one
44 87.3a 6-{4-[(3-chloropyridin-2-yl)oxy]phenyl}-1,5-dimethylpyrimidine- 2,4(1 /-/,3/-/)-dione
45 18.6 (+)-6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)- 1,5-dimethylpyrimidine-2,4(1 H,3/-/)-dione
46 7.66a 5-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2/-/)-one
47 82.3 6-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-5-ethyl- 1-methylpyrimidine-2,4(1H,3H)-dione
48 0.571 5-{4-[(3-iodopyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2/-/)-one
49 288 4,6-dimethyl-5-(2-methyl-4-{[3-(oxetan-3-yl)pyridin-2- yl]oxy}phenyl)pyridazin-3(2/-/)-one
50 10.1 6-(4-{[3-(difluoromethoxy)-4-methylpyridin-2-yl]oxy}-2- methylphenyl)-1,5-dimethylpyrimidine-2,4(1 /7,3H)-dione
51 94.9 4,6-dimethyl-5-(2-methyl-4-{[3-(trifluoromethoxy)pyridin-2- yl]oxy}phenyl)pyridazin-3(2/9)-one
52 63.1a 1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethoxy)pyridin-2- yl]oxy}phenyl)pyrimidine-2,4(1H,3/-/)-dione
53 109a 6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}phenyl)-1,5- dimethylpyrimidine-2,4(1H,3/-/)-dione
54 9.33 (-)-6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1 H,3H)-dione
55 24.2 (+)-6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidine-2,4(1H,3H)-dione
56 428a 5-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-(3,5-dimethyl-2,6-dioxo- 1,2,3,6-tetrahydropyrimidin-4-yl)benzonitrile
124
57 30.5a 5-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2H)-one
58 86.2 5-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-4,6- dimethylpyridazin-3(2/-f)-one
59 1220a 2-(4-(3,5-dimethyl-6-oxo-1,6-dihydropyridazin-4-yl)-3- methylphenoxy]pyridine-3-carboxamide, hydrochloride sait
60 767a methyl 2-(4-(3,5-dimethyl-6-oxo-1,6-dihydropyridazin-4-yl)-3- methylphenoxy]pyridine-3-carboxylate
61 23.8 5-{4-[(4-methoxy-3-methylpyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2H)-one
62 12.1 4,6-dimethyl-5-(2-methyl-4-{(4-methyl-3-(trifluoromethyl)pyridin-2- yl]oxy}phenyl)pyridazin-3(2H)-one
63 13.4 5-(4-{[3-(difluoromethyl)-4-methoxypyridin-2-yl]oxy}-2- methylphenyl)-4,6-dimethylpyridazin-3(2H)-one
64 4.17 5-(4-{[4-methoxy-3-(trifluoromethyl)pyridin-2-yl]oxy}-2- methylphenyl)-4,6-dimethylpyridazin-3(2/-/)-one
65 34.4 (-)-4,6-dimethyl-5-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2- yl]oxy}phenyl)pyridazin-3(2H)-one
66 26.9 (+)-4,6-dimethyl-5-(2-methyl-4-[[3-(trifluoromethyl)pyridin-2- yl]oxy}phenyl)pyridazin-3(2/^)-one
67 345a 4,6-dimethyl-5-{4-[(4-methylpyridin-2-yl)oxy]phenyl}pyridazin- 3(2H)-one
68 16.9 1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2- yl]sulfanyl}phenyl)pyrimidine-2,4(1/7,3/-/)-dione
69 38.3 6-{4-[(3-cyclopropylpyridin-2-yl)oxy]phenyl}-1,5- dimethylpyrimidine-2,4(1/7,3H)-dione
70 138a 5-{4-[(5-fluoro-3-methylpyridin-2-yl)oxy]-2-methylphenyl}-4,6- dimethylpyridazin-3(2/-/)-one
71 216a 6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrazin-2(1 H)-one
72 227a 6-(4-((3,5-dichloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrazin-2(1 H)-one
73 43.4a 6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrazin-2(1 /-/)-one
125
74 381a 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrazin-2(1 H)-one
75 87.0a 6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-1,5- dimethylpyrazin-2(1 /-/)-one
76 354a 6-{4-[(3,5-dichloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidin-2(1 H)-one
77 337a 1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2- yl]oxy}phenyl)pyrimidin-2(1/-/)-one
78 32.3a 6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidin-2(1 H)-one
79 109a 6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidin-2(1 H)-one
80 349a 6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5- dimethylpyrimidin-2(1 H)-one
81 58a 1-cyclopropyl-6-(4-((3-(difluoromethyl)pyridin-2-yl)oxy)-2- methylphenyl)-5-methylpyrimidine-2,4(1/7,3/-/)-dione
a. Value represents a sing e détermination.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appendant claims. Each reference (including ail patents, 5 patent applications, journal articles, books, and any other publications) cited in the présent application is hereby incorporated by reference in its entirety.

Claims (32)

  1. WHAT IS CLAIMED IS:
    1. A compound of Formula I or IA-1 :
    R1 R3 or a pharmaceutically acceptable sait thereof, wherein:
    each of T1, T2, T3, and T4 is independently selected from the group consisting of H, halogen, -CN, -SFs, -OH, -N(Ra)(Rb), -C(=O)-N(Ra)(Rb), -C(=O)-ORC, -C(=O)-Rd, Cm alkyl, Cve haloalkyl, C2.6 alkenyl, C2.6 alkynyl, C-|.6 alkoxy, C^e haloalkoxy, -S-(Ci-6 alkyl), C3_7 cycioalkyl, 4- to 7-membered heterocycloalkyl, C3.7 cycloalkoxy, 5- or 6-membered heteroaryl, cyclopropylmethyl, and cyclobutylmethyl, wherein each of the Cv6 alkyl, C2.6 alkenyl, C2.6 alkynyl, -3-(0^6 alkyl), and CV6 alkoxy is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, -N(Ra)(Rb), Cm alkoxy, CM haloalkoxy, and -S-(Cm alkyl); and wherein each of the C3.7 cycioalkyl, 4- to 7-membered heterocycloalkyl, C3.7 cycloalkoxy, 5- or 6-membered heteroaryl, cyclopropylmethyl, and cyclobutylmethyl of T1, T2, and T3 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, oxo, -N(Ra)(Rb), -C(=O)OH, -C(=O)-CW alkyl, -C(=O)-O-CM alkyl, -C(=O)-N(Ra)(Rb), CM alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, Cm haloalkoxy, and -S-(Cm alkyl);
    L1 is selected from the group consisting of O, S, NH, N(CM alkyl),
    N(-C!.2 alkyl-C3.4 cycioalkyl), and N(C3.6 cycioalkyl);
    each of Ra and Rb is independently selected from the group consisting of H, Cm alkyl, C3.7 cycioalkyl, and cyclopropylmethyl;
    or Ra and Rb together with the N atom to which they are attached form 4- to 7-membered heterocycloalkyl optionally substituted with one or more substituents each independently
    127 selected from the group consisting of halogen, -OH, -CN, oxo, -NH2, -NH(CW alkyl),
    -N(C14 alkyl)2, -C(=O)OH, -C(=O)-Cm alkyl, -C(=O)-O-CM alkyl, -C(=O)-NH2,
    -C(=0)-NH(Cm alkyl), - C(=0)-N(Cm alkyl)2, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, CM cyanoalkyl, Cm alkoxy, -S-(C14 alkyl), and Cm haloalkoxy;
    each of R° and Rd is independently Cm alkyl, C3.4 cycloalkyl-C^ alkyl-, or C3.4 cycloalkyl; Q1 is selected from the group consisting of Q1b, Q1c Q1d, and Q1e:
    Q1c (R12)t2 (R13 ôl2A
    X1 r13A
    Q1d Q1e, provided (a) that a ring carbon atom of the Q1 ring is attached to the benzene ring of Formula I and (b) that when L1 is NH, then the Q1 ring is substituted with at least one non-H R9, R10, R11, R12, R13, R9A, R1oa, R1ob, R11A, R12A, or R13A;
    each of X1 and X2 is independently O or S;
    each of R1, R2, R3, and R4 is independently selected from the group consisting of H, halogen, -OH, -NO2, -CN, -SF5, Cm alkyl, C..6 haloalkyl, Cm haloalkoxy, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, a 4- to 10-membered heterocycloalkyl, -N(R5)(R6), -N(R7)(C(=O)R8), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -N(R7)(S(=O)2R8), -S(=O)2-N(R5)(R6), -SR8, and -OR8, wherein each of the Cm alkyl C3.7 cycloalkyl, and heterocycloalkyl is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -CN, oxo, -OH, Cm alkyl, Cm alkoxy, Cm haloalkyl, Cm haloalkoxy, C3.6 cycloalkyl, -N(R5)(R6), -N(R7)(C(=O)R8), -C(=O)-OR8, -C(=O)H, -C(=O)R8, -C(=O)N(R5)(R6), -N(R7)(S(=O)2R8), -S(=O)2-N(R5)(R6), -SR8, and -OR8;
    or R2 and R4 together with the two carbon atoms to which they are attached form a fused
    5- or 6-membered heteroaryl, a fused 5- or 6-membered heterocycloalkyl ring, a fused 5- or 6membered cycloalkyl ring, or a fused benzene ring, wherein each of the fused rings is optionally substituted with 1,2, or 3 substituents each independently selected from the group consisting of
    128 halo, -CN, -OH, Cm alkyl, Cm alkoxy, haloalkyl, and Cm haloalkoxy, and wherein the fused heterocycloalkyl ring or fused cycloalkyl ring is further optionally substituted with 1, 2, or 3 oxo;
    R5 is H, Cm alkyl, Cm haloalkyl, or C3.7 cycloalkyl;
    R6 is H or selected from the group consisting of Cm alkyl, Cm haloalkyl, C3.7 cycloalkyl, a 4- to 10-membered heterocycloalkyl, C6-w aryl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-Ci-4 alkyl-, (4- to 10-membered heterocycloalkyl)-Ci.4 alkyl-, (Ce-ίο aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, wherein each of the sélections from the group is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from the group consisting of -OH, -CN, Cm alkyl, C3.7 cycloalkyl, Cm hydroxylalkyl, -S-Cm alkyl, -C(=O)H, -C(=0)-Cm alkyl, -C(=O)-O-Cm alkyl, -C(=O)-NH2, -C(=0)-N(Cm alkyl)2, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy;
    or R5 and R6 together with the N atom to which they are attached form a 4- to 10membered heterocycloalkyl or a 5- to 10-membered heteroaryl, each optionally substituted with 1,2,3, 4, or 5 substituents each independently selected from the group consisting of halogen, -OH, oxo, -C(=O)H, -C(=O)-Cm alkyl, -C(=O)OH, -C(=0)-0-Cm alkyl, -C(=O)-NH2, -C(=0)-N(Cm alkyl)2, -CN, Cm alkyl, Cm alkoxy, Cm hydroxylalkyl, Cm haloalkyl, and Cm haloalkoxy;
    R7 is selected from the group consisting of H, Cm alkyl, and C3.7 cycloalkyl;
    R8 is selected from the group consisting of Cm alkyl, C3.7 cycloalkyl, a 4- to 10membered heterocycloalkyl, C6-ioaryl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (C6_io aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-Ci.4 alkyl-, wherein each of the sélections from the group is optionally substituted with 1, 2, or 3 substituents each independently selected from the group consisting of halogen, -CF3, -CN, -OH, oxo, -S-Cm alkyl, Cm alkyl, Cw haloalkyl, C2.6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, Cm alkoxy, and Cm haloalkoxy;
    each R9 and R12 is independently selected from the group consisting of halogen, -OH, -CN, -SF5, -NO2, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm alkoxy, Cm haloalkoxy, C3.7 cycloalkyl, C2.6 alkenyl, C2.6 alkynyl, a 4- to 10-membered heterocycloalkyl, a 5- to 10membered heteroaryl, (C3.7 cycloalkyi)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (C6-io aryl)-CM alkyl-, (5- to 10-membered heteroaryl)-CM alkyl-, -N(R5)(R6), -N(R7)(C(=O)R8), -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -SR8, and -OR8, wherein each of the Cm alkyl, C3.7 cycloalkyl, 4- to 10-membered heterocycloalkyl, 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-Cv4 alkyl-, (4- to 10-membered heterocycloalkyl)-CV4 alkyl-, (Ce-io aryO-Ci-4 alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- is optionally substituted
    129 with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -NO2, Cm alkyl, Cm hydroxylalkyl, Cm alkoxy, -N(R5)(R6), -S-(Cm alkyl),
    -S(=O)2-(Cm alkyl), C6-i0 aryloxy, [(C6-io aryl)-CM alkyloxy- optionally substituted with 1 or 2
    Cm alkyl], oxo, -C(=O)H, -C(=O)-Cm alkyl, -C(=O)O-Cm alkyl, -C(=O)NH2, -NHC(=O)H,
    5 -NHC(=0)-(Cm alkyl), C3.7 cycloalkyl, a 5- or 6-membered heteroaryl, Cm haloalkyl, and
    Cm haloalkoxy;
    each of R10, R11 and R13 is independently selected from the group consisting of halogen, -OH, -CN, -SF5, -N02, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm alkoxy, Cm haloalkoxy, C3-7 cycloalkyl, C2.6 alkenyl, C2.6 alkynyl, C6-w aryl, a 4- to 10-membered heterocycloalkyl, a 5- to 10 10-membered heteroaryl, (C3.7 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (C6-io aryl)-CM alkyl-, (5- to 10-membered heteroaryl)-CM alkyl-, -N(R5)(R6), -N(R7)(C(=O)R8), -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=0)-Ra, -C(=0)-0R8, -SR8, and -OR8, wherein each of the Cm alkyl, C3.7 cycloalkyl, C6.10 aryl, 4- to 10-membered heterocycloalkyl, 5- to 10-membered heteroaryl,
    15 (C3.7 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-Ci-4 alkyl-, (C6.10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-Ci_4 alkyl- is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -N02, Cm alkyl, Cm hydroxylalkyl, Cm alkoxy, -N(R5)(R6), -S-(Cm alkyl), -S(=0)2-(Cm alkyl), C6.10 aryloxy, [(C6.10 aryl)-CM alkyloxy- optionally substituted with 1 or 2 Cm
    20 alkyl], oxo, -C(=O)H, -C(=O)-Cm alkyl, -C(=O)O-C^ alkyl, -C(=O)NH2, -NHC(=0)H,
    -NHC(=0)-(C1_4 alkyl), C3.7 cycloalkyl, a 5- or 6-membered heteroaryl, Cm haloalkyl, and
    Cm haloalkoxy;
    each of R9A and R10A is independently selected from the group consisting of H, Cm alkyl, Cm hydroxylalkyl, C2.6 alkenyl, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=0)-R8, -C(=O)-OR8,
    25 -SR15, -C(R14)2-OH, -C(R14)2-OS(=O)2H, -C(R14)2-OP(=O)(OH)2, -C(R14)2-OR15,
    -C(R14)2-OC(=O)-R15, -C(R14)2-N(R5)(R6), each of R10B, R11A, R12A, and R13A is independently selected from the group consisting of H, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, C3.7 cycloalkyl, C3.6 alkenyl, C3.6 alkynyl, C6.10 aryl, a 4- to 10-membered heterocycloalkyl, a 5- to 10-membered heteroaryl,
    30 (C3.7 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-Ci.4 alkyl-, (C6_io aryl)-CM alkyl-, (5- to 10-membered heteroaryl)-Ci_4 alkyl-, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, and -C(=0)-0R8, wherein each of the Cm alkyl, C3.7 cycloalkyl, C6_io aryl, 4- to 10-membered heterocycloalkyl, 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-Cv4 alkyl-,
    35 (C6.10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, is optionally substituted with 1, 2, 3, or 4 substituents each independently selected from the group consisting of halogen,
    130
    -OH, -CN, -NO2, Cm alkyl, hydroxylalkyl, CM alkoxy, -N(RS)(R6), -S-(Cm alkyl),
    -S(=0)2-(Cm alkyl), C6-io aryloxy, [(C6.10 aryl)-CM alkyloxy- optionally substituted with 1 or 2
    Cm alkyl], oxo, -C(=O)H, -C(=O)-Cm alkyl, -C(=O)O-Cm alkyl, -C(=O)NH2, -NHC(=O)H, -NHC(=0)-(Cm alkyl), -OC(=O)-Cm alkyl, C3.7 cycloalkyl, a 5- or 6-membered heteroaryl, Cm haloalkyl, and CM haloalkoxy;
    each R14 is independently H or selected from the group consisting of C^o alkyl, C3.14 cycloalkyl, C2.10 alkenyl, C2.10 alkynyl, C6.10 aryl, 4- to 10-membered heterocycloalkyl, 5- to 10-membered heteroaryl, (C3.14 cycloalkyl)-Ci_10 alkyl-, (4- to 14-membered heterocycloalkylj-CMo alkyl-, (C6-io aryl)-Cv10 alkyl-, (5- to 10-membered heteroarylj-CMo alkyl-, wherein each of the sélections of the group is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -NO2, Cm alkyl, Cm hydroxylalkyl, Cm alkoxy, -N(R5)(R6), -N(R7)C(=O)R8, -N(R7)C(=O)OR8, -N(R7)S(=O)2R8, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -SR8, -OR8, -S(=O)2-R8, C6-i0 aryloxy, [(C6.10 aryl)-CM alkyloxyoptionally substituted with 1 or 2 Cm alkyl], oxo, -C(=O)H, -NHC(=O)H, C3.7 cycloalkyl, a 5- or 6membered heteroaryl, CV4 haloalkyl, and Cm haloalkoxy;
    R15 is selected from the group consisting of Ci.2O alkyl, C3.14 cycloalkyl, C2.20 alkenyl, C2.20 alkynyl, C6.10 aryl, 4- to 14-membered heterocycloalkyl, 5- to 10-membered heteroaryl, (C3-14 cycloalkyl)-Cv2o alkyl-, (4- to 10-membered heterocycloalkylj-Cv^ alkyl-, (θε-ίο arylj-CMo alkyl-, (5- to 10-membered heteroarylj-Cvao alkyl-, wherein each of the sélections of the group is optionally substituted with 1,2, 3, or 4 substituents each independently selected from the group consisting of halogen, -OH, -CN, -NO2, Cm alkyl, Cm hydroxylalkyl, CM alkoxy, -N(R5)(R6), -N(R7)C(=O)R8, -N(R7)C(=O)OR8, -N(R7)S(=O)2R8, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -SR8, -OR8, -S(=O)2-R8, C6-io aryloxy, [(C6.10 aryl)-CM alkyloxy- optionally substituted with 1 or 2 Cm alkyl], oxo, -C(=O)H, -NHC(=O)H, C3-7 cycloalkyl, a 5- or 6-membered heteroaryl, Cm haloalkyl, and Cm haloalkoxy;
    t1 is 0, 1, or 2;
    t2 is 0 or 1 ; and t3 is 0, 1, or 2.
  2. 2. The compound of Claim 1, or a pharmaceutically acceptable sait thereof, wherein L1 is O or S.
    131
  3. 3. The compound of Claim 1 or 2, or a pharmaceutically acceptable sait thereof, wherein the compound is a compound of Formula IB-1, IB-2, ΙΒ-3, ΙΒ-4, or IB-5, IB-6, ΙΒ-7, IB-8, IB-9, or
    IB-7
    IB-8
    132
  4. 4. The compound of Claim 1 or 2, or a pharmaceutically acceptable sait thereof, wherein
  5. 5 the compound is a compound of Formula IA-1, IA-2, IA-3, IA-4, IA-5, IA-6, IA-7, IA-8, IA-9, IA10, IC-1, IC-2, IC-3, IC-4, IC-5, IC-6, ID-1, ID-2, ID-3, ID-4, ID-5, ID-6, ID-7, ID-8, ID-9, ID-10,
    IE-1, IE-2, IE-3, IE-4, or IE-5, IE-6, IE-7, IE-8, IE-9, or IE-10:
    133
    IA-10
    IC-1 IC-2
    IC-3
    IC-4
    134
    ID-5
    ID-6
    135
    IE-3
    IE-4
    136
    IE-9
    IE-10.
    5. The compound of any one of Claims 1 to 4, or a pharmaceutically acceptable sait thereof, wherein:
    10 each R9 is independently selected from the group consisting of -CN, Cm alkyl,
    Cm haloalkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R9 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R9 is optionally substituted with 15 one or more substituents each independently selected from the group consisting of halogen,
    -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, CM alkoxy, and
    Ci-4 haloalkoxy
    R9A is selected from the group consisting of H, Cm alkyl, Cm hydroxylalkyl, allyl,
    -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -C(R14)2-OH, -C(R14)2-OS(=O)2H,
    20 -C(R14)2-OP(=O)(OH)2, -C(R14)2-OR15, and -C(R14)2-OC(=O)-R15;
    137
    R10 is selected from the group consisting of -CN, Cm alkyl, C3-4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the C1.4 alkyl of R10 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, C1.4 alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, 5 cyclopropylmethyl, and cyclobutylmethyl of R10 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, CV4 alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy;
    R1oa is selected from the group consisting of H, Cm alkyl, Ci-3 hydroxylalkyl, C2.4 alkenyl, -S(=O)2N(R5)(R6), -C(=O)-N(R5)(R6), -C(=O)-R8, -C(=O)-OR8, -C(R14)2-OH, -C(R14)2-OS(=O)2H, 10 -C(R14)2-OP(=O)(OH)2, -C(R14)2-OR15, and -C(R14)2-OC(=O)-R15; and
    R1ob is selected from the group consisting of Cm alkyl, C3-4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R10B is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and 15 cyclobutylmethyl of R10B is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy.
    each R11 is independently selected from the group consisting of -CN, Cm alkyl,
    C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R11 is
    20 optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R11 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm
    25 haloalkoxy;
    R11A is selected from the group consisting of Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R11A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and 30 cyclobutylmethyl of R11A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, C1.4 haloalkyl, Cm hydroxylalkyl, CM cyanoalkyl, Cm alkoxy, and Cm haloalkoxy;
    each R12 is independently selected from the group consisting of -CN, Cm alkyl,
    CM cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R12 is
    35 optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the
    138
    C3_4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R12 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH,
    -CN, Cm alkyl, CM haloalkyl, C^.4 hydroxylalkyl, C1.4 cyanoalkyl, C^4 alkoxy, and Ci-4 haloalkoxy;
    R12A is selected from the group consisting of Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the C^.4 alkyl of R12A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R12A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy;
    each R13 is independently selected from the group consisting of -CN, Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the Cm alkyl of R13 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Ci.4 haloalkoxy; and wherein each of the C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R13 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy; and
    R13A is selected from the group consisting of Cm alkyl, C3.4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl, wherein the C^4 alkyl of R13A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and Cm haloalkoxy; and wherein each of the C3-4 cycloalkyl, cyclopropylmethyl, and cyclobutylmethyl of R13A is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, C-\.4 haloalkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy.
  6. 6. The compound of any one of Claims 1 to 5, or a pharmaceutically acceptable sait thereof, wherein:
    each R9 is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl;
    each of R10 and R10B is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl;
    each of R11 and R11A is independently selected from the group consisting of Cm alkyl, Cm haloalkyl, and cyclopropyl;
    139 each of R12 and R12A is independently selected from the group consisting of alkyl,
    CV3 haloalkyl, and cyclopropyl; and each of R13 and R13A is independently selected from the group consisting of alkyl,
    Ci_3 haloalkyl, and cyclopropyl.
  7. 7. The compound of any one of claims Claim 1 to 6, or a pharmaceutically acceptable sait thereof, wherein:
    each R9 is independently methyl or ethyl;
    each of R10 and R10B is independently methyl or ethyl;
    each of R11 and R11A is independently methyl or ethyl;
    each of R12 and R12A is independently methyl or ethyl; and each of R13 and R13A is independently methyl or ethyl.
  8. 8. The compound of any one of Claims 1 to 7, or a pharmaceutically acceptable sait thereof, wherein each of R1 and R2 is independently selected from the group consisting of H, halogen, -CN, Cve alkyl, C^e haloalkyl, Ci.6 alkoxy, C^e haloalkoxy, C3.6 cycloalkyl, -C(=O)-(Ci_4 alkyl), -C(=O)OH, and C(=O)-O-(Ci_4 alkyl), wherein each of the C^e alkyl and C3.6 cycloalkyl is optionally substituted with 1,2, 3, 4, or 5 substituents each independently selected from halogen, -OH, -CN, C14 alkyl, C^ haloalkyl, Ο;.4 alkoxy, and Ον4 haloalkoxy.
  9. 9. The compound of any one of Claims 1 to 8, or a pharmaceutically acceptable sait thereof, wherein each of R1 and R2 is independently H, methyl, or halogen.
  10. 10. The compound of any one of Claims 1 to 9, or a pharmaceutically acceptable sait thereof, wherein each of R1 and R2 is H.
  11. 11. The compound of any one of Claims 1 to 10, or a pharmaceutically acceptable sait thereof, wherein each of R3 and R4 is independently selected from the group consisting of H, halogen, -CN, - Ci_4 alkyl, C^ alkoxy, and C3.4 cycloalkyl, wherein each of the C^ alkyl and CM alkoxy of R3 and R4 is optionally substituted with 1,2, 3, 4, or 5 substituents each independently selected from halogen, -OH, Cî-4 alkoxy, and C^ haloalkoxy; and wherein the C3.4 cycloalkyl of R3 and R4 is optionally substituted with 1,2,3, 4, or 5 substituents each independently selected from halogen, -OH, C;.4 alkyl, Ον4 haloalkyl, C^ alkoxy, and Ci.4 haloalkoxy.
  12. 12. The compound of any one of Claims 1 to 11, or a pharmaceutically acceptable sait thereof, wherein R3is H; and R4 is H, halogen, or methyl, wherein the methyl is optionally
    140 substituted with 1,2, or 3 substituents each independently selected from the group consisting of halogen, -OH, and Cmalkoxy.
  13. 13. The compound of any one of Claims 1 to 12, or a pharmaceutically acceptable sait 5 thereof, wherein R3is H and R4 is methyl.
  14. 14. The compound of any one of Claims 1 to 13, or a pharmaceutically acceptable sait thereof, wherein each of T1, T2, T3, and T4 is independently selected from the group consisting of H, halogen, -CN, Cm alkyl, Cm haloalkyl, C2_4 alkenyl, Cm alkoxy, Cm haloalkoxy, and
    10 C3.4 cycloalkyl, wherein each of the Cm alkyl, C2.4 alkenyl, and Cm alkoxy of T1, T2, T3, and T4 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkoxy, and CM haloalkoxy; and wherein the C3.4 cycloalkyl of T1, T2, T3, and T4 is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -OH, -CN, Cm alkyl, Cm haloalkyl,
  15. 15 Cm hydroxylalkyl, Cm cyanoalkyl, Cm alkoxy, and Cm haloalkoxy.
    15. The compound of any one of Claims 1 to 14, or a pharmaceutically acceptable sait thereof, wherein each of T1, T2, T3, and T4 is independently selected from the group consisting of H, halogen, Cm alkyl, Cm hydroxylalkyl, Cm haloalkyl, Cm alkoxy, Cm haloalkoxy,
    20 C3.4 cycloalkyl, and C3.4 halocycloalkyl.
  16. 16. The compound of any one of Claims 1 to 15, or a pharmaceutically acceptable sait thereof, wherein T1 is selected from the group consisting of halogen, cyclopropyl, halocyclopropyl, methyl, C! haloalkyl, methoxy, and C! haloalkoxy; and T4 is H.
  17. 17. A compound of Claim 1 selected from: (-)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine-
    2,4(1 H,3/-/)-dione; (-)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine30 2,4(1 /-/,3/-/)-dione;
    6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-5-ethyl-1-methylpyrimidine-2,4(1H,3H)dione;
    (-)-1,5-dimethyl-6-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2-yl]oxy}phenyl)pyrimidine2,4(1 H,3H)-dione;
    35 (-)-6-{4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine2,4(1H,3H)-dione;
    141
    6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]phenyl}-1,5-dimethylpyrimidine-2,4(1/-/,3H)-dione;
    (-)-6-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy)-2-methylphenyl)-1,5-dimethylpyrimidine-
    2,4(1/7,3/i)-dione;
    (+)-5-(4-{[3-(difluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6-dimethylpyridazin3(2H)-one;
    6-{4-[(3-chloropyridin-2-yl)sulfanyl]-2-methylphenyl}-1,5-dimethylpyrimidine-2,4(1/7,3/-/)dione;
    5-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-4,6-dimethylpyridazin-3(2/-/)one;
    5-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-4,6-dimethylpyridazin-3(2/-/)-one;
    5-{4-[(3-iodopyridin-2-yl)oxy]-2-methylphenyl)-4,6-dimethylpyridazin-3(2/-/)-one;
    (-)-6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidine-2,4(1/-/,3/-/)dione;
    5-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-4,6-dimethylpyridazin-3(2/-/)-one;
    5-(4-{[3-(difluoromethoxy)pyridin-2-yl]oxy}-2-methylphenyl)-4,6-dimethylpyridazin-3(2H)one;
    5- (4-{[4-methoxy-3-(trifluoromethyl)pyridin-2-yl]oxy}-2-methylphenyl)-4,6dimethylpyridazin-3(2/-/)-one;
    (+)-4,6-dimethyl-5-(2-methyl-4-{[3-(trifluoromethyl)pyridin-2-yl]oxy)phenyl)pyridazin3(2H)-one;
    6- {4-[(3-cyclopropylpyridin-2-yl)oxy]phenyl}-1,5-dimethylpyrimidine-2,4(1H,3H)-dione;
    6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrazin-2(1H)-one;
    6-{4-[(3-chloro-4-methylpyridin-2-yl)oxy]-2-methylphenyl}-1,5-dimethylpyrimidin-2(1H)one; and
    1-cyclopropyl-6-(4-((3-(difluoromethyl)pyridin-2-yl)oxy)-2-methylphenyl)-5methylpyrimidine-2,4(1/-/,3/-/)-dione, or a pharmaceutically acceptable sait thereof.
  18. 18. A compound of Claim 1 that is (+)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl)-
    1.5- dimethylpyrimidine-2,4(1H,3/-/)-dione, or a pharmaceutically acceptable sait thereof.
  19. 19. A compound of Claim 1 that is (-)-6-{4-[(3-chloro-5-fluoropyridin-2-yl)oxy]-2-methylphenyl}-
    1.5- dimethylpyrimidine-2,4(1 H,3H)-dione, or a pharmaceutically acceptable sait thereof.
  20. 20. A compound of Claim 1 that is (+)-1,5-dimethyl-6-(2-methyl-4-[[3-(trifluoromethyl)pyridin-2yl]oxy}phenyl)pyrimidine-2,4(1 /-/,3H)-dione, or a pharmaceutically acceptable sait thereof.
    142
  21. 21. A compound of Claim 1 !hat is (-)Ί .S-dimethyl-e^a-msthyl^-Îia^tnfluoromethyOpyridin-ayljoxyJphenyOpyrimidine^^il H,3H)-dione, or a pharmaceutically acceptable sait thereof.
    5
  22. 22. A compound of Claim 1 that is (+)-6-{4-[(3-chlaropyridin-2-yl)oxy]-2-methylphenyl]-1l5dimeÎhylpyrimidine-2l4(1/7,3H)-dione, or a pharmaceutically acceptable sait thereof.
  23. 23. A compound of Claim 1 thatis(-)-6-{4-[(3-chloropyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethyîpyrimidine-2,4(lH,3H)-dione, or a pharmaceutically acceptable sait thereof.
  24. 24. A compound of Claim 1 that is (+)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl}-1,5dimethylpyrirnidine^AflH.aHJ-diane, or a pharmaceutically acceptable sali thereof.
  25. 25. A compound of Claim 1 that Is (-)-6-{4-[(3-cyclopropylpyridin-2-yl)oxy]-2-methylphenyl)-1,5-
    15 dimethylpyrimidine-2,4(1 H,3H)-dione, or a pharmaceutically acceptable sait thereof.
  26. 26. A compound of Claim 1 that is (+)-6-(4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-
    1.5- dimelhylpyrlmidine-2t4(1H,3iO-dÎone, or a pharmaceutically acceptable sait thereof.
    20
  27. 27. A compound of Claim 1 that is (-)-6-(4-[(3-chloro-5-methylpyridin-2-yl)oxy]-2-methylphenyl}-
    1.5- dimethylpyrimidine-2,4(1 H,3H)-dione, or a pharmaceutically acceptable sait thereof.
  28. 28. A compound of Claim 1 that is (+)-6-(4-{[3-(difluoromethoxy)pyrldin-2-yl]oxy}-2methylphenyl)-1,5-dirnethylpyrimidine-2,4(1H,3/7)-dtone, or a pharmaceutically acceptable sait
    25 thereof.
  29. 29. A compound of Claim 1 thaï is (-)-6-(4-([3-(difluoromethoxy)pyridin-2-yl]oxy}-2methylphenyl)-1,5-dimethylpyrimidine-2,4(1 H,3H)-dione, or a pharmaceulically acceptable sali thereof.
  30. 30. A pharmaceutical composition comprising a compound according to any one of Claims 1 to 29 or a pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier.
    L.
    143
  31. 31. Use of a compound according to any one of Claims 1 to 29 or a pharmaceutically acceptable sait thereof for treatment of a disorder that is selected from schizophrenia, cognitive impairment, attention déficit hyperactivity disorder (ADHD), impulsivity, compulsive gambling, overeating, autism spectrum disorder, mild cognitive impairment (MCI), age-related cognitive
    5 décliné, dementia, restless leg syndrome (RLS), Parkinson’s disease, Huntington’s chorea, anxiety, dépréssion, major dépressive disorder (MDD), treatment-resistant dépréssion (TRD), bipolar disorder, chronic apathy, anhedonia, chronic fatigue, post-traumatic stress disorder, seasonal affective disorder, social anxiety disorder, post-partum dépréssion, serotonin syndrome, substance abuse and drug dependence, drug abuse relapse, Tourette’s syndrome, 10 tardive dyskinesia, drowsiness, excessive daytime sleepiness, cachexia, inattention, sexual dysfunction, migraine, systemic lupus erythematosus (SLE), hyperglycemia, atherosclerosis, dislipidemia, obesity, diabètes, sepsis, post-ischemic tubular necrosis, rénal failure, hyponatremia, résistant edema, narcolepsy, hypertension, congestive heart failure, postoperative ocular hypotonia, sleep disorders, and pain.
  32. 32. The use according to claim 31, where the disorder is selected from schizophrenia, cognitive impairment, mild cognitive impairment (MCI), age-related cognitive décliné, dementia, and Parkinson’s disease.
OA1201500511 2013-06-27 2014-06-13 Heteroaromatic compounds and their use as Dopamine DI ligands OA17657A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US61/840,144 2013-06-27

Publications (1)

Publication Number Publication Date
OA17657A true OA17657A (en) 2017-05-15

Family

ID=

Similar Documents

Publication Publication Date Title
US11014909B2 (en) Heteroaromatic compounds and their use as dopamine D1 ligands
EP3134087B1 (en) Heteroaromatic compounds and their use as dopamine d1 ligands
WO2015166370A1 (en) Heteroaromatic compounds and their use as dopamine d1 ligands
AU2015249497A1 (en) Heteroaromatic compounds and their use as dopamine D1 ligands
EP3134405B1 (en) Heteroaromatic compounds and their use as dopamine d1 ligands
EP3137469B1 (en) Heterocyclic compounds and their use as dopamine d1 ligands
US11964961B2 (en) Heteroaromatic compounds and their use as dopamine D1 ligands
OA17657A (en) Heteroaromatic compounds and their use as Dopamine DI ligands
OA18535A (en) Heteroramatic Compounds and their use as Dopamine D1 Ligands.
BR112015030101B1 (en) HETEROAROMATIC COMPOUNDS, PHARMACEUTICAL COMPOSITION AND USE OF SAID COMPOUNDS IN THE TREATMENT OF DISORDERS MEDIATED BY OR ASSOCIATED WITH DOPAMINE D1
OA18107A (en) Heteroaromatic compounds and their use as dopamine D1 ligands