OA18506A - Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment. - Google Patents

Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment. Download PDF

Info

Publication number
OA18506A
OA18506A OA1201700496 OA18506A OA 18506 A OA18506 A OA 18506A OA 1201700496 OA1201700496 OA 1201700496 OA 18506 A OA18506 A OA 18506A
Authority
OA
OAPI
Prior art keywords
alkyl
aryl
aliphatic
membered heteroaryl
compound
Prior art date
Application number
OA1201700496
Inventor
Belew Mekonnen
Jianxing Huang
John A. Butera
Original Assignee
Agenebio, 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 Agenebio, Inc. filed Critical Agenebio, Inc.
Publication of OA18506A publication Critical patent/OA18506A/en

Links

Abstract

This invention relates to benzodiazepine derivatives, compositions comprising therapeutically effective amounts of those benzodiazepine derivatives and methods of using those derivatives or compositions in treating cognitive impairment associated with central nervous system (CNS) disorders. In particular, it relates to the use of a α5-containing GABAΑ receptor agonist (e.g., a α5-containing GABAA receptor positive allosteric modulator) as described herein in treating cognitive impairment associated with central nervous system (CNS) disorders in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer's Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson's disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.

Description

BENZODIAZEPINE DERIVATIVES, COMPOSITIONS, AND METHODS FOR TREATING COGNITIVE IMPAIRMENT
Related Applications |0001 [ This application claims the benefit of and priority from U.S. Provisional Patent
Application 62/182,336, filcd Junc 19, 2015. The foregoing application is incorporated herein by référence in its entircty.
Statement of Govcrnmcnt Support )0001.1] This invention was made with govcrnmcnt support under Grant No.
U01AG041140 awardcd by the National lnstitutcs of Hcalth (NIH), and in particular, its
National Institutc on Aging (NIA) division, an agcncy of the United States Govcmmcnt.The United States Govcrnmcnt has certain rights in the invention.
Field ofthe Invention )0002) The invention relates to compounds, compositions and methods for treating cognitive impairment associated with central nervous system (CNS) disorders in a subject in need of treatment or at risk of said cognitive impairment.
Background of the Invention )0003) Cognitive ability may décline as a normal conséquence of aging or as a conséquence of a central nervous disorder.
(0004) For examplc, a sîgnificant population of eldcrly adults expériences a décline in cognitive ability that exceeds what is typical in normal aging. Such agc-rclated loss of cognitive function is characterized clinically by progressive loss of memory, cognition, rcasoning, and judgment. Mild Cognitive Impairment (MCI), Agc-Associatcd Memory Impairment (AAM1), Agc-Rclatcd Cognitive Décline (ARCD) or similar clinicai groupings are among those related to such agc-rclated loss of cognitive function. According to some estimâtes, there are more than 16 million people with AAMi in the U.S. alone (Barker et al., 1995), and MCI is estimated to affect 5.5 - 7 million in the U.S. over the âge of 65 (Plassman et al., 2008).
)0005) Cognitive impairment is also associated with other central nervous system (CNS) disorders, such as dementia, Alzhcimcr’s Disease (AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, bipolar disorder (in particular, mania), amyotrophie latéral sclcrosîs (ALS), canccr-therapy-relatcd cognitive impairment, mental
retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
[0006] There is, therefore, a need for effective treatment of cognitive impairment associated with central nervous system (CNS) disorders and to improve cognitive function in patients diagnosed with, for example, age-related cognitive impairment, MCI, amnestîc MCI, AAMI, ARCD, dementia, AD, prodromal AD, PTSD, schizophrénie or bipolar disorder (in particular, mania), amyotrophie latéral sclerosis (ALS), cancertherapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction and similar central nervous system (CNS) disorders with cognitive impairment or at risk of developing them.
[0007] GABAa receptors (GABAa R) are pentameric assemblies from a pool of different subunîts (al-6, β1*3, γ1-3, δ, ε, π, 0) that form a CI- permeable channel that is gated by the neurotransmittery-aminobutyric acid (GABA). Various pharmacological 15 effects, including anxiety disorders, epilepsy, insomnia, pre-anesthetic sédation, and muscle relaxation, are mediated by different GABAa subtypes.
[0008] Various studies hâve demonstrated that reduced GABA signaling is linked to various CNS disorders with cognitive impairment. In particular, the a5-containing GABAa Rs, which are relatively sparse in the mammalian brain, play a rôle in modifying 20 leamîng and memory. Previous studies demonstrated a réduction of hippocampal expression ofthe a5 subunit ofthe GABAa receptor in rats with age-related cognitive décliné (see International Patent Publication WO 2007/019312). Such results suggest that upregulation of a5-containing GABAa R function may be effective in the treatment of cognitive impairment associated with said CNS disorders.
[0009] Thus, there is a need for positive allosteric modulators of a5-containing GABAa
R that are useful in therapeutic préparations for the treatment of cognitive impairment associated with said CNS disorders.
Siimmarv of the Invention [0010] The présent invention addresses the aforementioned need by providing a compound of formula I:
I, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
Ais C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(Rk;
EisN,NR7, CR6orC(R6)2;
WisN, NR7, CR6orC(R6)2;
X îs N, NR7,0, CR6 or CfR^;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
Vis Cor CR6, or when Z is C or CR6, V is C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is
-SR8, -(CH2)„OR8, -(CH2)nO(CH2)nR8, -(CH2)PR8 and -(CH2)nN(R”)R10; and wherein R2 is independently substituted with 0-5 R*;
m and n are independently integers selected from 0-4;
p is an integer selected from 2-4;
each occurrence of the bond “ ” is either a single bond or a double bond;
each occurrence of R1, R2, R4, and Rsare each independently selected from: halogen, -R, -OR, -N02, -NCS, -CN, -CF3, -OCF3, -SÎR3, -N(R)2, -SR, -SOR,
-SO2R, -SO2N(R)2, -SO3R, -(CR2)j.3R, -(CR2)|.3-OR, -(CR2)o-3-C(0)NR(CR2)wR, -(CR2)o.3-C(0)NR(CR2)o.3OR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(0)0R, -C(O)C(O)OR, -C(0)C(0)N(R)2, -OC(O)R, -C(0)N(R)2, -OC(0)N(R)2, -C(S)N(R)2, -(CR2)o.3NHC(0)R, -N(R)N(R)C0R, -N(R)N(R)C(0)0R, -N(R)N(R)C0N(R)2, -N(R)S02R, -N(R)SO2N(R)2, -N(R)C(0)0R, -N(R)C(0)R,
-N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R,
-C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
R3 is absent or is selected from:
halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR,
-SO2R,-SO2N(R)2,-SO3R,-(CR2)|.3R, -(CR2)1.3-OR, -(CR2)o.3-C(0)NR(CR2)o-3R, -(CR2)o.3-C(0)NR(CR2)o.3OR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(0)N(R)2, -OC(0)N(R)2, -C(S)N(R)2, -(CR2)o.3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(0)R,
-N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R,
-C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
each R6 is independently -H or -(C 1 -C6)alkyl;
each R7 is independently -H or -(C 1 -C6)alkyl;
each R8 is independently -(Cl*C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to
10- membered heteroaryl, wherein each occurrence of R8 îs independently substituted with 0-5 R’;
each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’;
each R is independently selected from:
H-,
(Cl-C12)-al iphatic-, (C3-C ! 0)-cycloalkyl-, (C3-C10)-cycloalkenyl-, [(C3 -C 10)-cyc loal ky l]-(C ! -C ! 2 )-al iphatic-, [(C3-C10)-cycloalkenyl]-(C ! -C ! 2)-al iphatic-, [(C3-C10)-cycloalkyl]-O-(Cl-C12)-aliphatic-, [(C3-C10)-cycloalkenyl]-O-(C 1 -C 12)-al iphatic-, (C6-C10)-aryl-, (C6-C10)-ary l-(C ! -C 12)a I iphat ic-, (C6-C10)-aryl-O-(C 1 -C 12)a! iphatic-, (C6-C10)-ary l-N(R’ ’ )-(C 1 -C 12)a! iphatic-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyc!yl)-O-(Cl-C!2)aliphatic-, (3- to 10- membered heterocyclyl)-N(R”)-(CI-C12)aliphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl)-(Cl-C12)-al iphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(CI-C12)-al iphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independentiy selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independentiy selected from N, NH, O, and S;
wherein each occurrence of R is independentiy substituted with 0-5 R’;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independentiy selected from N, NH, O, S, SO, and SO2, wherein said ring is optîonally substituted with 0-5 R’, and wherein said ring is optîonally fused to a (Cô-ClO)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independentiy selected from halogen, -R”, -OR”, oxo,
-CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R”)2;
wherein each occurrence of R” is independentiy selected from H,-(C!-C6)-alkyl, -(Cl
C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-CI0)aryl-(Cl-C6)-alkyl-, (5- to 10- membered heteroaryl)-0-(Cl-C6)-alkyl-, and (C6-C10)aryl-O-(CÎ-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR°, oxo, -CH2OR0, -CH2NR0 2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN,
-CF3, -OCF3 and -N(R°)2, wherein each occurrence of R°is independently selected from: -(Cl-C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-C10)-aryl-.
[0011] Some embodiments ofthis application provide a compound of formula I:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
Ais C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6)2;
EisN.NR7, CR6orC(R6)2;
WisN, NR7, CR6orC(R6)2;
X is N, NR7,0, CR6 or C(R6)2;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
V is C or CR6, or when Z is C or CR6, V is C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is
, then R2 is -OR8,
-SR8, -(CH2)nOR8, -(CH2)„O(CH2)nR8, -(CH2)PR8 and -(CH2)„N(R”)R10; and wherein
R2 is independently substituted with 0-5 R’;
m and n are independently întegers selected from 0-4;
p is an integer selected from 2-4;
each occurrence of the bond “ ” is either a single bond or a double bond;
each occurrence of R1, R2, R4, and R5 are each independently selected from: halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR, -SO2R, -SO2N(R)2, -SO3R, -(CR2)i.3R, .(CR2)i.3-OR, -(CR2)o.3-C(0)NR(CR2)o.3R, -(CR2)o.j-C(0)NR(CR2)o.3OR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)q-3NHC(O)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
R3 is absent or is selected from:
halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR, -SO2R, -SO2N(R)2, -SO3R, -(CR2)i-3R, -(CR2)i.3-OR, -(CR2)q-3-C(0)NR(CR2)o.3R, -(CR2)o.3-C(0)NR(CRj)o.3OR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)o.3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
each R6is independently-H or-(Cl-C6)alkyI; each RTis independently-H or-(Cl-C6)alkyl;
each R8 is independently -(Cl-Cô)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-ary 1, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’;
each R10 is independently -(C3-CI0)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6CI0)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R*° is independently substituted with 0-5 R’;
each R is independently selected from:
H-, (CI-CI2)-aliphatic-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloalkenyl-, [(C3-C10)-cycloalky l]-(C 1 -C 12)-a I iphatic-, [(C3-C10)-cycloalkenyl]-(C I -C 12)-al i phatîc-, [(C3-C 10)-cycloalkyl]-O-(C 1 -C 12)-alt phatîc-, [(C3 -C 10)-cycloalkenyl]-O-(C 1 -C 12)-al i phatîc-, (C6-C10)-aryl-, (C6-C10>aryl-(C 1 -C 12)al iphatic-, (C6-C10)-ary 1-O-(C I -C 12)al i phatîc-, (C6-C10)-aryl-N(R”)-(Cl-C 12)al iphatic-.
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-CI2)altphatic-, (3- to 10- membered heterocyclyl)-0-(Cl-C12)aliphatîc-, (3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)aliphatic-,
5- to 10- membered heteroaryl-, .
(5- to 10- membered heteroaryl)-(Cl-C12)-alîphatïc-, (5- to 10- membered heteroaryl)-O-(CI-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-alÎphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SQ2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R is independently substituted with 0-5 R’;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or non aromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCFj and -N(R”)2;
wherein each occurrence of R” îs independently selected from H, -(C l-C6)-alkyl, (C3C6)-cydoalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10)-aryl-, (5-to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10)-ary!-O-(ClC6)-alkyl-.
[0012] Some embodiments of this application provide a compound of formula I:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
Ais C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6)2;
E is N, NR7, CR6 or C(R6)2;
W is N, NR7, CR6 or C(R6)2;
X is N, NR7,0, CR6 or C(R6)2;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
V is C or CR6, or when Z ïs C or CR6, V is C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is
, then R2 is -OR8,
-SR8, or-(CH2)nOR8;
m and n are each independently an integer selected from 0-4;
each occurrence of the bond “ ” is either a single bond or a double bond;
each occurrence of R1, R2, R4, and R5 are each independently selected from: halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR, -SChR, -SO2N(R)2, -SOjR, -(CR2)i.3R, -(CR2)|.3-OR, -(CR2)w-C(0)NR(CR2)o.3R, -(CR2)o.3-C(0)NR(CR2)o.30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R» -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(Rh, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)û.3NHC(O)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SOiR, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(Rh, -P(O)(OR)2, and -P(O)(H)(OR);
R3 is absent or is selected from:
halogen, -R, -OR, -NCfe, -NCS, -CN, -CF3, -OCF3, -SÎRa, -N(R)2, -SR, -SOR, -SO2R, -SQ2N(R)2, -SOjR, -(CR2)|.3R, -(CR2)i-3-OR, -(CR2)o-3-C(0)NR(CR2)o.3R, -(CR2)o.3-C(0)NR(CR2)o.3OR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(Rh -OC(O)N(R)2, -C(S)N(R)2, -(CR2)o.3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R» -OP(O)(OR)2, -P(O)(R)2i -P(O)(OR)2, and -P(O)(H)(OR);
each R6is independently-H or -(C I -C6)alkyl;
each R7 is independently -H or -(C 1 -C6)alkyl;
each Rs is independently -(Cl-C6)alkyl, -(C3-C10)-cycloaIkyl, -(C6-Ci0)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of Rs is independently substituted with 0-5 R’;
each R is independently selected from:
H-, (Cl-C12)-aliphatic-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloalkenyl-, [(C3-C10)-cycloaIkyl]-(Cl-C12)-aliphatic-, [(C3-C10)-cycloalkenyl]-(C 1 -C 12)-aliphatîc-, [(C3 · C10)-cyc loalkyl ]-O-(C 1 -C 12 )-a 1 i phat i c-, [(C3-C10)-cyc I oal kenyl ]-O-(C 1 -C I 2)-a 1 iphat i c-, (C6-C10)-aryl-, (C6-C10)-ary l-(C 1 -C 12)al i ph at i c-, (C6-C 10)-aryl-O-(C 1-C12)al iphati c-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Ci-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-CI2)aliphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl>(CI-C12)-aliphatic-, and (5- to 10- membered heteroaryl)-O-(Cl-CI2)-aliphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, 25 NH, O, and S; ' wherein each occurrence of R is independently substituted with 0-5 R’;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, 30 and SO7, wherein said ring is optionally substituted with 0-5 R\ and wherein said ring is optionally fused to a (C6-CI0)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo,
-CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NCh, -NCS, -CN, -CF3, -OCF3 and-N(R”)2;
wherein each occurrence of R” is independently selected from H, -<Cl-C6)-alkyl, (C3 C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)-ary 1 -, (5 - to 10- membere d hete roary 1)-(C 1 -C6)-alky 1-, (C6-C10)-aryl-(Cl-C6)-alkyl-, (5- to 10- membered heteroaryl)-0-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-, [0013] In another aspect, the présent invention provides a compound of formula II:
II, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, îsomer, or combination thereof, wherein m, R1, R2, R3, R4, R5 and R6 are as defined in formula I.
[0014] In another aspect, the présent invention provides a compound of formula III:
R6
III, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, îsomer, or
combination thereof, wherein m, R1, R2, R3, R4, R8 and R6 are as defined in formula I.
[0015] in another aspect, the présent invention provides a compound of formula IV:
N
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein R2 is -OR8, -SR8, or-(CHiJnOR8, wherein R2 is independently substituted with 0-5 R* and wherein m, n, R1, R3, R4, R8, Rs, and R8 are as defined in formula 1.
[0016] In another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein R2 is -(CH:)nO(CH2)nR8, -(CHîJpR8 or -(CH2)nN(R’’)R10, wherein R2 is independently substituted with 0-5 R’ and wherein m, n, p, R1, R3, R4, R8, R6, R8, R10, and R” are as defined herein.
[0017] The présent invention also provides pharmaceutical compositions that comprise a compound of formulae I, II, III, or IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
[0018] In some embodiments, compounds of formula I are GABAa a5 receptor positive allosteric modulators. In some embodiments, compounds of formula II are GABAa a5 receptor positive allosteric modulators. In some embodiments, compounds of formula III are GABAa <x5 receptor positive allosteric modulators. tn some embodiments, compounds of formula IV are GABAa a5 receptor positive allosteric modulators. Compounds of formula t, Π, III or IV can be used to treat the conditions described herein, such as through activity as GABAa a5 receptor positive allosteric modulators.
[00Î9J In another aspect of the invention, there is provided a method for treating cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment, the method comprising the step of administering to saia subject a therapeutically effective amount of a compound of the invention or a phi rmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof. In some embodiments, the CNS disorder with cognitive impairment includes, hout limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), antnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation.
wi
Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In another aspect of the invention, there is provided a method of preserving or improving cognitive function in a subject in need thereof, the method comprising the step of administering to said subject a therapeutically effective amount ofa compound ofthe invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof. In certain embodiments of the invention, a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof is administered every 12 or 24 hours.
[0020] ln some embodiments, the compounds and compositions of the présent invention are for use as a médicament, ln some embodiments, the compounds and compositions of the présent invention are for use in treating cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenîa, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
[0021] In some embodiments, this application provides the use of a compound or composition described herein in the préparation of a médicament for the treatment of cognitive impairment associated with a CNS disorder în a subject în need of treatment or at risk ofsaid cognitive impairment. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestîc MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenîa, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
Detailed Description ofthe Figures [0022] Figure 1 is a graph depicting the effects of administering methyl 3,5diphenylpyridazine-4-carboxylate on the spatial memory rétention often aged-impaired (AI) rats ïn an eight-arm Radial Arm Maze (RAM) test. The black bars refer to rats treated with vehicle alone; open bars refer to rats treated with methyl 3,5diphenylpyridazine-4-carboxylate at different doses; hatched bar refers to rats treated with the combination of TB2I007 and methyl 3,5-diphenylpyridazine-4-carboxylate.
[0023] Figure 2 is a graph showing the effect of methyl 3,5-diphenylpyridazîne-4carboxylate (administered intravenously) on the binding of RoI54513 in the hippocampus and cerebellum. Methyl 3,5-diphenylpyridazine-4-carboxylate blocked the binding of Rol545I3 in the hippocampus but dîd not affect bindingof Rol54l3 in the cerebellum.
{0024] Figure 3 is a graph showing dose-dependent GABAa a5 receptor occupancy by methyl 3,5-diphenylpyrîdazine-4-carboxylate administered intravenously, with receptor occupancy determined either by the ratio between hippocampus (a région of high GABAa a5 receptor density) exposure ofRO 15-4513 and cerebellum (a région with low GABAa a5 receptor density) exposure of RO 15-4513, or by using the GABAa a5 sélective compound L-655,708 (10 mg/kg, i.v.) to define full occupancy.
[0025] Figure 4 is a graph showing exposure occupancy relationships for methyl 3,5dîphenylpyridazine-4-carboxylate în hippocampus. Methyl 3,5-diphenylpyridazine-4carboxylate occupïes about 32% of GABAA a5 receptors at exposures which are behaviorally active in aged-impaired rats.
[0026] Figures 5 is a graph depictïng the effect of ethyl 3-methoxy-7-methyl-9Hbenzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate on the spatial memory rétention often aged-impaired (AI) rats în an eight-arm Radial Arm Maze (RAM) test. Figure 5 shows the effect ofethyl 3-methoxy-7-methyl-9H15 benzo[f]imidazo[l,5-a][I,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate on the spatial memory rétention of ten aged-impaired (AI) rats in the RAM test, where the vehicle control was tested 3 times, and the different doses of ethyl 3-methoxy-7-methyl-9Hbenzo[f]imidazo[!,5-a][l,2,4]triazolo[4,3-d][l,4]dÎazepine-10-carboxylate were tested twice; In Figure 5, black bars refer to rats treated with vehicle alone and open bars refer to rats treated with ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-
d][l,4]dîazepine-I0-carboxylate at different doses.
[0027] Figure 6 îs a graph showing the effect of ethyl 3-methoxy-7-methyl-9Hbenzo[f]imidazo[I,5-a][l,2,4]triazolo[4,3-d][l,4]diazepÎne-IO-carboxylate (administered intravenously) on the binding of Roi 54513 în the hippocampus and cerebellum. Ethyl 325 methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4.3-d][!,4]diazepine-10carboxylate blocked the binding of Roi54513 în the hippocampus but did not affect binding ofRo!54I3 in the cerebellum.
[0028] Figure 7 is a graph showing dose-dependent GABAa o5 receptor occupancy by ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,330 d][l,4]diazepine-l 0-carboxylate administered intravenously, as calculated by the ratio between hippocampus (a région of high GABAaci5 receptor density) exposure of RO 1518506 • 17
4513 and cerebellum (a région with 1owGABAa«5 receptor density) exposure of RO 154513 to define full occupancy..
[0029] Figure 8(A)-(C) are graphs showing the effect of 6,6 dimethyl-3-(3hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one, as compared to vehicle dimethyl sulfoxtde (DMSO), in aged-impaired rats using a Morris water maze behavioral task. Figure 8(A) shows the escape latency (i.e., the average time in seconds rats took to find the hidden platform in the water pool) during training in rats received 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2benzothiophen-4(5H)-one and rats received vehicle DMSO; Figure 8(B) shows the amount of time spent in target annulus and opposite annulus by rats received 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)one and rats received vehicle DMSO; Figure 8(C) shows number of crossing in target annulus and opposite annulus by rats received 6,6 dimethyl-3-(3-hydroxypropyl)thio-l(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one and rats received vehicle DMSO.
Detailed Description of the Invention
Définitions ]0030] Unless otherwise defined herein, scientific and technical terms used in this application shall hâve the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, chemistry, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurochemistry, virology, immunology, microbiology, pharmacology, genetics and protein and nucleic acid chemistry, described herein, are those well known • and commonly used in the art.
[0031] The methods and techniques of the présent invention are generally performed, unless otherwise indicated, according to conventîonal methods well known in the art and as described in various general and more spécifie référencés that are cited and discussed throughout this spécification. See, e.g. “Principles of Neural Science,” McGraw-Hill Medical, New York, N.Y. (2000); Motulsky, “Intuitive Biostatistics,” Oxford University Press, Inc. (1995); Lodish et al., “Molecular Cell Biology, 4th ed.,” W. H. Freeman &
Co., New York (2000); Griffiths et al., “Introduction to Genetic Analysis, 7th ed.,” W. H.
Freeman & Co., N.Y. (1999); and Gilbert et al., “Developmental Biology, 6th ed.,” Sinauer Associâtes, inc., Sunderland, MA (2000).
[0032] Chemistry terms used herein are used according to conventional usage in the art, as exemplified by “The McGraw-Hill Dictionary of Chemical Terms,” Parker S., Ed., 5 McGraw-Hill, San Francisco, C.A. (1985).
[0033] Ail of the publications, patents and published patent applications referred to in this application are specifically incorporated by reference herein. In case of conflict, the présent spécification, including its spécifie définitions, will control.
[0034] Throughout this spécification, the word “comprise” or variations such as “comprises or “comprising” will be understood to imply the inclusion of a stated integer (or components) or group of integers (or components), but not the exclusion of any other integer (or components) or group of integers (or components).
[0035] The singular forms “a,” “an,” and “the” include the plurals unless the context clearly dictâtes otherwise.
[0036] The term “including” is used to mean “including but not Iimited to”.
“Including” and “including but not Iimited to” are used interchangeably.
[0037] The term “agent” is used herein to dénoté a chemical compound (such as an organic or inorganic compound (including, such as, a compound ofthe présent invention), a mixture of chemical compounds), a biological macromolecule (such as a nucleic acid, 20 an antibody, including parts thereof as well as humanized, chimerîc and human antibodies and monoclonal antibodies, a protein or portion thereof, e.g., a peptide, a lipid, a carbohydrate), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues. Agents include, for example, agents which are known with respect to structure, and those which are not known with 25 respect to structure. The a5-containing GABAa receptor agonist activity of such agents may render them suitable as “therapeutîc agents” in the methods and compositions of this invention.
[0038] A “patient,” “subject,” or “individual” are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animais (including bovine, porcine, etc.), companion animais (e.g., canine, feline, etc.) and rodents (e.g., mice and rats).
[0039] “Cognitive fonction” or “cognitive status” refers to any higher order intellectual brain process or brain state, respectively, involved in leaming and/or memory including, but not limited to, attention, information acquisition, information processing, working memory, short-term memory, long-term memory, anterograde memory, rétrogradé memory, memory retrieval, discrimination leaming, decisîon-making, inhibîtory response control, attentional set-shifting, delayed reinforcement leaming, reversai leaming, the temporal intégration of voluntary behavîor, expressing an interest în one’s surroundings and self-care, speed of processing, reasoning and problem solving and social cognition.
[0040] In humans, cognitive fonction may be measured, for example and without limitation, by the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatrie Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Sélective Remindîng Test (Buschke and Fuld, 1974); the Verbal Paired Associâtes subtest; the Logical Memory subtest; the Visual Reproduction subtest ofthe Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997); the Benton Visual Rétention Test, or the explicit 3-altemative forced choice task, or MATRICS consensus neuropsychological test battery. See Folstein ettA., J Psychiatrie Res 12:189-98,(1975); RobbinsetaL, Dementia 5:266-81, (1994); Rey, L'examen clinique en psychologie, (1964); Kluger et al., J Geriatr Psychiatry Neurol 12:168-79, (1999); Marquis et al., 2002 and Masur et al., 1994. Also see Buchanan, R.W., Keefe, R.S.E., Umbricht, D., Green, M.F., Laughren, T., and Marder, S.R. (2011), The FDA-NIMH-MATRICS guidelines for clinical trial design of cognitive-enhancing drugs: what do we know 5 years later? Schizophr. Bull. 37, 1209-1217.
[0041] In animal model Systems, cognitive fonction may be measured in various conventional ways known in the art, including using a Morris Water Maze (MWM), Bames circular maze, elevated radial arm maze, T maze or any other mazes in which the animais use spatial information. Cognitive fonction can be assessed by reversai leaming, extradimensional set shifting, conditîonal discrimination leaming and assessments of • 20 reward expectancy. Other tests known in the art may also be used to assess cognitive function, such as novel object récognition and odor récognition tasks.
[0042] Cognitive function may also be measured using imaging techniques such as Positron Emission Tomography (PET), functional magnetic résonance imaging (fMRI),
Single Photon Emission Computed Tomography (SPECT), or any other imaging technique that allows one to measure brain function. In animais, cognitive function may also be measured with electrophysiological techniques.
[0043] “Promoting” cognitive function refers to affecting impaired cognitive function so that it more closely resembles the function of a normal, unimpaired subject. Cognitive function may be promoted to any détectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily actïvîties of normal life at a level of proficiency as close as possible to a normal, unimpaired subject or an agematched normal, unimpaired subject.
[0044[ In some cases, “promoting” cognitive function in a subject affected by age15 related cognitive refers to affecting impaired cognitive function so that it more closely resembles the function of an aged-matched normal, unimpaired subject, or the function of a young adult subject. Cognitive function ofthat subject may be promoted to any détectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily activïtîes of normal life at a level of proficiency close as possible to a normal, unimpaired subject or a young adult subject or an age-matched normal unimpaired subject.
[0045] “Preserving” cognitive function refers to affecting normal or impaired cognitive function such that it does not décliné or does not fall below that observed in the subject upon first présentation or diagnosis, or delays such décliné.
[0046] “Improving” cognitive function includes promoting cognitive function and/or preserving cognitive function in a subject.
[0047] “Cognitive impairment” refers to cognitive function in subjects that is not as robust as that expected in a normal, unimpaired subject. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in a normal, unimpaired subject. In some cases, “cognitive impairment” in subjects affected by aged-related cognitive impairment refers to cognitive function in subjects that is not as robust as that expected în an aged-matched normal, unimpaired subject, or the function of a young adult subject (i.e. subjects with mean scores for a given âge in a cognitive test).
[0048] “Age-related cognitive impairment” refers to cognitive impairment in aged subjects, wherein their cognitive function is not as robust as that expected in an agematched normal subject or as that expected in young adult subjects. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in an age-matched normal subject. In some cases, cognitive function is as expected in an age-matched normal subject, but reduced by about 5%, about 10%, about 30%, about 50% or more, compared to cognitive function expected in a young adult subject. Age-related impaired cognitive function may be associated with Mild Cognitive Impairment (MCI) (including amnestic MCI and non-amnestic MCI), Age-Associated Memory Impairment (AAMI), and Age-related Cognitive Décliné (ARCD).
[0049] Cognitive impairment associated with AD or related to AD or in AD refers to cognitive function in subjects that is not as robust as that expected în subjects who hâve not been diagnosed AD using conventional méthodologies and standards.
[0050] “Mild Cognitive Impairment” or “MCI” refers to a condition characterized by isolated memory impairment unaccompanied other cognitive abnormalities and relatively normal functional abilities. One set of criteria for a clinicai characterization of MCI spécifiés the following characteristics: (1) memory complaint (as reported by patient, informant, or physicîan), (2) normal activities of daîly living (ADLs), (3) normal global cognitive function, (4) abnormal memory for âge (defined as scorîng more than 1.5 standard déviations below the mean for a given âge), and (5) absence of indicators of dementia (as defined by DSM-1V guidelînes). Petersen et al., Srch. Neurol. 56:303-308 (1999); Petersen, “Mild cognitive impairment: Aging to Alzheimeris Disease.” Oxford University Press, N.Y. (2003). The cognitive déficit in subjects with MCI may involve any cognîtion area or mental process including memory, language, association, attention, perception, problem solving, executive function and visuospatial skills. See, e.g., Winbald et al., J. Intern. Med. 256:240-240, 2004; Meguro, Acta. Neurol. Taiwan. 15:5518506
57,2008; Ellison et al., CNSSpectr. 13:66-72,2008, Petersen, Semin. Neurol. 27:22-31, 2007. MCI is further subdivided into amnestic MCI (aMCI) and non-amnestic MCI, characterized by the impairment (or lack thereof) of memory in particular. MCI is defined as aMCI if memory is found to be impaired given the âge and éducation level of the subject. If, on the other hand, the memory ofthe subject îs found to be intact for âge and éducation, but other non-memory cognitive domains are impaired, such as language, executive function, or visuospatial skills, MCI is defïnes an non-amnestic MCI. aMCI and non-amnestic MCI can both be further subdivided into single or multiple domain MCI. aMCI-single domain refers to a condition where memory, but not other cognitive areas are impaired. aMCI-multiple domain refers to a condition where memory and at least one other cognitive area are impaired. Non-amnestic MCI îs single domain or multiple domain dépendent on whether nor not more than one non-memory cognitive area is impaired. See, e.g., Peterson and Negash, CNSSpectr. 13:45-53,2008.
[0051] Diagnosis of MCI usually entaîls an objective assessment of cognitive impairment, which can be gamered through the use of well-established neuropsychologîcal tests, including the Mini Mental State Examination (MMSE), the Cambridge Neuropsychological Test Automated Battery (CANTAB) and individual tests such as Rey Auditory Verbal Leaming Test (AVLT), Logical Memory Subtest ofthe revised Wechsler Memory Scale (WMS-R) and the New York University (NYU) Paragraph Recall Test. See Folstein et at, J Psychiatrie Res 12:189-98 (1975); Robbîns étal., Dementia5:266-81 (1994); Khiger et al., J Gériatrie Psychiatry Neurol 12:168-79 (1999).
[0052] Age-Associate Memory Impairment (AAMI) refers to a décliné in memory due to aging. A patient may be considered to hâve AAMI if he or she is at least 50 years old and meets ail ofthe following criteria: a) The patient has noticed a décliné in memory performance, b) The patient performs worse on a standard test of memory compared to young adults, c) Ail other obvious causes of memory décliné, except normal aging, hâve been ruled out (in other words, the memory décliné cannot be attributed to other causes such as a recent heart attack or head injury, dépréssion, adverse reactions to médication, Alzheimer's disease, etc.).
[0053] Age-Related Cognitive Décliné (ARCD) refers to déclinés in memory and cognitive abilities that are a normal conséquence of aging in humans (e.g., Craik & Salthouse, 1992). This is also true in virtually ail mammalian species. Age-Associated Memory Impairment refers to older persons with objective memory déclinés relative to their younger years, but cognitive functioning that is normal relative to their âge peers (Crook et al., 1986). Age-Consistent Memory Décliné is a less péjorative label which emphasizes that these are normal developmental changes (Crook, 1993; Larrabee, 1996), are not pathophysiological (Smith et al., 1991), and rarely progress to overt dementia (Youngjohn & Crook, 1993). The DSM-IV (1994) has codified the diagnostic classification of ARCD.
[0054] “Dementia” refers to a condition characterized by severe cognitive déficit that interfères in normal actîvities of daily living. Subjects with dementia also display other symptoms such as impaired judgment, changes in personality, dîsorientation, confusion, behavior changes, trouble speaking, and motor déficits. There are different types of dementias, such as Alzheïmer’s disease (AD), vascular dementia, dementia with Lewy bodies, and frontotemporal dementia.
[0055] Alzheïmer’s disease (AD) is characterized by memory déficits in its early phase. Later symptoms Include Impaired judgment, disorientation, confusion, behavior changes, trouble speaking, and motor déficits. Histologically, AD is characterized by beta-amyloîd plaques and tangles of protein tau.
[0056] Vascular dementia is caused by strokes. Symptoms overlap with those of AD, but without the focus on memory impairment.
[0057] Dementia with Lewy bodies is characterized by abnormal deposits ofalphasynucletn that form inside neurons in the brain. Cognitive impairment may be similar to AD, including impairments in memory and judgment and behavior changes.
[0058| Frontotemporal dementia is characterized by gliosis, neuronal loss, superfictal spongiform degeneration in the frontal cortex and/or anterior temporal lobes, and Picks’ bodies. Symptoms include changes in personality and behavior, including a décliné in social skîlls and language expression/comprehension.
[0059] “Post traumatic stress disorder (PTSD)” refers to an anxiety disorder characterized by an immédiate or delayed response to a catastrophic event, characterized by re-experiencing the trauma, psychic numbing or avoîdance of stimuli associated with the trauma, and increased arousal. Re-experiencing phenomena include intrusive memories, flashbacks, nightmares, and psychological or physiological distress in response to trauma reminders. Such responses produce anxiety and can hâve significant impact, both chronic and acute, on a patient’s quality of life and physical and emotiona! health. PTSD Is also associated with impaired cognitive performance, and older individuals with PTSD hâve greater décliné in cognitive performance relative to control patients.
[0060] “Schizophrenia” refers to a chronic debilitating disorder, characterized by a spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental représentations (e.g, hallucinations, delusions), négative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g, anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalîties in the brain are proposed to underlic the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely îneffective in treating cognitive impairments in patients.
[0061] Bipolar disorder or BP or manie dépressive disorder or manie dépressive illness refers to a chronic psychological/mood disorder which can be characterized by 20 significant mood changes including periods of dépression and euphorie manie periods. BP may be diagnosed by a skilled physician based on Personal and medical history, interview consultation and physical examinations. The term mania or manie periods or other variants refers to periods where an individual exhibits some or all of the following characteristics: racing thoughts, rapid speech, elevated levels of activity and agitation as 25 well as an înflated sense of self-esteem, euphoria, poor judgment, insomnia, impaired concentration and aggression.
[0062] “Amyotrophie latéral sclerosîs,*’ also known as ALS, refers to a progressive, fatal, neurodegenerative disease characterized by a degeneration of motor neurons, the nerve cells in the central nervous system that control voluntary muscle movement. ALS is also characterized by neuronal degeneration in the entorhinal cortex and hippocampus, memory déficits, and neuronal hyperexcitability in different brain areas such as the cortex.
[0063] “Cancer-therapy-related cognitive impairment” refers to cognitive impairment that develops in subjects that are treated with cancer thérapies such as chemotherapy and radiation. Cytotoxicity and other adverse side-effects on the brain of cancer thérapies resuit in cognitive impairment in such functions as memory, leaming and attention.
[0064] Parkinson’s disease (PD) is a neurological disorder characterized by a decrease of voluntary movements. The afflicted patient has réduction ofmotor activity and slower voluntary movements compared to the normal individual. The patient has characteristic mask face, a tendency to hurry while walking, bent over posture and generalized weakness of the muscles. There is a typical lead-pipe rigïdïty of passive movements. Another important feature ofthe disease is the tremor ofthe extremities occurring at rest and decreasing during movements.
[0065] “Autism, as used herein, refers to an autism spectrum disorder characterized by a neural development disorder leading to impaired social interaction and communication by restricted and répétitive behavior. “Autism Spectrum Disorder refers to a group of developmental disabilities that includes: autism; Asperger syndrome; pervasive developmental disorder not otherwise specified (PDD-NOS or atypical autism); Rett syndrome; and chîldhood disintegrative disorder.
[0066] Mental retardation is a generalized disorder characterized by significantly impaired cognitive function and déficits in adaptive behaviors. Mental retardation is often defined as an Intelligence Quotient (IQ) score of less than 70. Inbom causes are among many underlying causes for mental retardation. The dysfunction in neuronal ' communication is also considered one ofthe underlying causes for mental retardation (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10,207-214).
[0067] In some instances, mental retardation includes, but are not limited to, Down syndrome, velocariofacial syndrome, fêtai alcohol syndrome, Fragile X syndrome, Klinefelter’s syndrome, neurofibromatosis, congénital hypothyroîdism, Williams syndrome, phenylketonuria (PKU), Smith-Lemlî-Opitz syndrome, Prader-Willi syndrome, Phelan-McDermid syndrome, Mowat-Wilson syndrome, ciliopathy, Lowe syndrome and siderium type X-linked mental retardation. Down syndrome is a disorder that includes a combination of birth defects, including some degree of mental retardation, characteristic facial features and, often, heart defects, increased infections, problems with vision and hearing, and other health problems. Fragile X syndrome is a prévalent form of inherited mental retardation, occurring with a frequency of 1 in 4,000 males and 1 in 8,000 females. The syndrome is also characterized by developmental delay, hyperactivity, attention déficit disorder, and autistic-like behavior. There is no effective treatment for fragile X syndrome.
[0068] Obsessive compulsive disorder (OCD) is a mental condition that is most commonly characterized by intrusive, répétitive unwanted thoughts (obsessions) resulting in compulsive behavîors and mental acts that an individual feels driven to perform (compulsion). Current epidemiologîcal data indicates that OCD is the fourth most common mental disorder in the United States. Some studies suggest the prevalence of OCD is between one and three percent, although the prevalence of clinically recognîzed OCD is much lower, suggesting that many indi viduals with the disorder may not be dîagnosed. Patients with OCD are often diagnosed by a psychologîst, psychïatrist, or psychoanalyst according to the Diagnostic and Statistical Manual of Mental Disorders, 4th édition text révision (DSM-IV-TR) (2000) diagnostic criteria that include characteristics of obsessions and compulsions.
[0069] Substance addiction (e.g., drug addiction, alcohol addiction) is a mental disorder. The addiction is not triggered instantaneously upon exposure to substance of abuse. Rather, it involves multiple, complex neural adaptations that develop with different time courses ranging from hours to days to months (Kauer J. A. Nat. Rev. Neurosci. 2007, 8, 844-858). The path to addiction generally begins with the voluntary use of one or more controlled substances, such as narcotics, barbiturates, methamphetamînes, alcohol, nicotine, and any of a variety of other such controlled substances. Over time, with extended use ofthe controlled substance(s), the voluntary ability to abstain from the controlled substance(s) is compromïsed due to the effects of prolonged use on brain function, and thus on behavior. As such, substance addiction generally is characterized by compulsive substance craving, seeking and use that persist even in the face of négative conséquences. The cravings may represent changes in the underlying neurobiology of the patient which likely must be addressed în a meaningful way if recovery is to be obtained. Substance addiction is also characterized in many cases by withdrawal symptoms, which for some substances are life threatening (e.g., alcohol, barbiturates) and in others can resuit in substantial morbidity (which may include nausea, vomiting, fever, dizzîness, and profuse sweating), distress, and decreased ability to obtain recovery. For example, alcoholism, also known as alcohol dependence, is one such substance addiction. Alcoholism is primarily characterized by four symptoms, which include cravings, loss of control, physical dependence and tolérance. These symptoms also may characterize addictions to other controlled substances. The craving for alcohol, as well as other controlled substances, often is as strong as the need for food or water. Thus, an alcoholic may continue to drink despite serious family, health and/or legal ramifications.
[0070] “Treating” a condition or patient refers to taking steps to obtain bénéficiai or desired results, including clinical results. Bénéficiai or desired clinical results include, but are not limited to, preventing or slowing the progression ofthe disease or disorder, or alleviatîon, amelioration, or slowing the progression, of one or more symptoms of cognitive impairment associated with CNS disorders, such as age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenîa, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapyrelated cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In some embodiments, treatment comprises preventing or slowing the progression, of a CNS disorder (such as one as described herein). In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with that CNS disorder. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive déficit. Treating age-related cognitive impairment further comprises slowing the conversion of age-related cognitive impairment (including, but not limited to MCI, ARCD and AAMI) into dementia (e.g., AD).
[0071 ] “Treating cognitive impairment” refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject’s performance in one or more cognitive tests is improved to any détectable degree, or is prevented from further décliné. Preferably, that subject’s cognitive function, after treatment ofcognitive impairment, more closely resembles the function of a normal, unimpaired subject. Treatment of cognitive impairment in humans may improve cognitive function to any détectable degree, but is preferably improved sufficiently to allow the impaired subject to 5 carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject. In some cases, “treating cognitive impairment” refers to taking steps to improve cognitive function în a subject with cognitive impairment so that the subject’s performance in one or more cognitive tests is improved to any détectable degree, or is prevented from further décliné. Preferably, that subject’s cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. In some cases, “treating cognitive impairment” în a subject affecting by age-related cognitive impairment refers to takings steps to improve cognitive function in the subject so that the subject’s cognitive function, after treatment of cognitive impairment, more closely resembles the function of an age-matched normal, unimpaired subject, or the function of a young adult subject.
[0072] Administering or administration of' a substance, a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art. For exemple, a compound or an agent can be administered, întravenously, arterially, intradermally, întramuscularly, intraperitoneally, întravenously, subcutaneously, ocularly, 20 sublingually, orally (by ingestion), intranasally (by inhalation), intraspinally, intracerebrally, and transdermally (by absorption, e.g., through a skin duct). A compound or agent can also appropriately be întroduced by rechargeable or biodégradable polymeric devices or other devices, e.g., patches and pumps, or formulations, which provide for the extended, slow, or controlled release of the compound or agent. Administering can also 25 be performed, for example, once, a plurality of times, and/or over one or more extended periods. In some aspects, the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug. For example, as used herein, a physician who instructs a patient to self-administer a drug, or to hâve the drug administered by another and/or who provides a patient with a 30 prescription for a drug is administering the drug to the patient.
[0073] Appropriate methods of administering a substance, a compound or an agent to a subject will also dépend, for example, on the âge of the subject, whether the subject is active or inactive at the time of administering, whether the subject is cogmtively impaired at the time of administering, the extent of the impairment, and the chemical and biological properties ofthe compound or agent (e.g. solubility, digestibility, bioavailability, stability and toxicity). In some embodiments, a compound or an agent is administered orally, e.g., to a subject by ingestion, or intravenously, e.g., to a subject by injection. In some embodiments, the orally administered compound or agent is in an extended release or slow release formulation, or administered using a device for such slow or extended release.
[0074] As used herein, a “a5-containing GABAa receptor agonîst,” “a5-containing GABAa R agonîst” or a “GABAa «5 receptor agonist” and other variations as used herein refer to a compound that enhances the function of a5-containing GABAa receptor (GABAa R), l e-, a compound that increase GABA-gated Cl currents. In some embodiments, a5-contaînîng GABAa R agonist as used herein refers to a positive allosteric modulator, which potentiates the activity ofGABA. a5-containing GABAa receptor agonists, suitable for use in the présent invention, include the aS-containing GABAa receptor agonists of ail formulas and spécifie aS-containing GABAa receptor agonists described herein, and their hydrates, solvatés, polymorphs, salts (e.g., pharmaceutically acceptable salts), isomers (e.g., stereoisomers, E/Z isomers, and tautomers), and combinations thereof.
|0075] “Antipsychotic”, “antipsychotic agent”, “antipsychotic drug”, or “antipsychotic compound refers to (1) a typical or an atypical antipsychotic; (2) an agent that is selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), Ml muscarinicacétylcholinereceptor(mAChR)positiveallosteric modulators (PAMs), histamine H3 receptor antagonists, AMPA/kainate receptor antagonists, ampakines (CX-516), glutathîone prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, nNOS inhibits, neurosteroids, and neurotrophic factors, alpha-7 agonists or positive allosteric modulators (PAMs)PAMs, serotonin 2C agonists; and/or (3) an agent that is useful in treating one or more signs or symptoms of schîzophrenîa or bipolar disorder (in particular, mania). [0076] ‘Typical an ti psychotic s”, as used herein. refer to conventional antipsychotics, which produce antipsychotic effects as well as movement related adverse effects related to disturbances in the nigrostriatal dopamine system. These extrapyramidal side effects (EPS) include Parkinsonisin. akathisia, tardive dyskinesia and dystonia. See Baldessarînî and Tarazi in Goodman & Gilman's The Pharmacological Basis of Therapeutics 10 Edition. 2001, pp. 485-520.
[0077] Atypîcal antipsychotics”, as used herein. refer to antipsychotic drugs that produce antipsychotic effects with îîttle or no EPS and include. but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasîdone, paliperidone, quetiapine, rispéridone and ziprasidone. Atypical antipsychotics differ from conventional antipsychotics in their pharmacological profiles. While conventional antipsychotics are characterized princîpally by D; dopamine receptor bloc kade, atypical antipsychotics show antagonist effects on multiple receptors including the 5HTa and 5HTC serotonin receptors and varying degrees ofreceptor affinities. Atypical antipsychotic drugs are commonly referred to as serotonin/dopainine antagonists, reflecting the influentia! hypothesîs that greater affinity for the 5HT2 receptor than for the D2 receptor underlies atypical antipsychotic drug action or second génération antipsychotic drugs. However, the atypical antipsychotics often display side effects, including. but not limited to, weight gain, diabètes (e.g., type II diabètes mellitus), hyperlipidemia, QTc interva! prolongation, myocarditis. sexual side effects. extra pyramidal side effects and cataract. Thus, atypical antipsychotics do not represent a homogeneous class, given their différences in the context of both alleviation of clinical symptoms and their potential for inducing side effects such as the ones listed above. Further, the common side effects of the atypical antipsychotics as described above often limit the antipsychotic doses that can be used for these agents.
[0078] Memantine is chemically known as 3,5-dimethy!adamantan-l-amine or 3,5dimethy!tricyclo[3.3.1.13,7]decan-l-amine, which is an uncompetitive N-methy!-Daspartate (NMDA) receptor antagonist with moderate affinity. The proprietary names for memantine include: Axura® and Akatinol® (Merz), Namenda® (Forest Laboratories), Ebixa® and Abixa® (Lundbeck), and Memox® (Unipharm). Memantine is approved for the treatment of moderate to severe Alzheimer's disease (AD) in the United States at a
dose of up to 28 mg/day. Dérivatives or analogs of memantine, which include compounds that structurally or chemically resemble memantine, are also useful in the présent invention. Such dérivatives or analogs of memantine include, but are not limited to those compounds disclosed în U.S. Patents Nos. 3,391,142; 4,122,193; 4,273,774; and
5,061,703; U.S. Patent Application Publication US20040087658, US20050113458,
US20060205822, US20090081259, US20090124659, and US20100227852; EP Patent Application Publication EP2260839A2; EP Patent EPI 682109B1 ; and PCT Application Publication W02005079779, ail of which are incorporated herein by reference. Memantine, as used in the présent invention, includes memantine and its dérivatives and analogs, as well as hydrates, polymorphs, prodrugs, salts, and solvatés thereof. Memantine, as used herein, also includes a composition comprising memantine or a dérivative or an analog or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, or prodrug thereof, wherein the composition optionally further comprises at least one additional therapeutic agent (such as a therapeutic agent useful for treating a
CNS disorder or cognitive impairments associated thereof). In some embodiments, the memantine composition suitable for use in the présent invention comprises memantine and a second therapeutic agent that is donepezil (under the trade name Aricept).
[0079] “Acetylcholinesterase inhibitor” or “AChE-I” as used herein refers to an agent that inhibits the ability of the cholinestérase enzyme to break down the neurotransmitter acétylcholine, thereby increasing the concentration and duration of acétylcholine, maînly in brain synapses or neuromuscular junctions. AChE-Is suitable for use in this application may include, for example, the subcategories of (i) réversible non-competitive inhibitors or réversible compétitive inhibitors, (ii) irréversible, and (iiî) quasi-îrreversible inhibitors. * [0080] The term simultaneous administration, as used herein, means that a a5-containing
GABAa receptor agonist (e.g., a a5-containing GABAa receptor positive allosteric modulator) and a second therapeutic agent (e.g., an antipsychotic, memantine or an AChE-1), or their pharmaceutically acceptable salts, hydrates, solvatés, or polymorphs, are administered with a time séparation of no more than about 15 minutes, and în some embodiments no more than about 10 minutes. When the drugs are administered simultaneously, the a5-containing GABAa receptor agonist (e.g., an a5-containing GABAa receptor positive allosteric modulator) and a second therapeutic agent (e.g., an antipsychotic, memantine or an AChE-I), or their salts, hydrates, solvatés, or polymorphe, may be contained in the same dosage (e.g., a unît dosage form comprising both the a5-containing GABAa receptor agonist (e.g., an a5containing GABAa receptor positive allosteric modulator) and a second therapeutic agent (e.g., an antipsychotic, memantine or an AChE-I) or in discrète dosages (e.g., the a5contaîning GABAa receptor agonist (e.g., an a5-containing GABAa receptor positive allosteric modulator) or its sait, hydrate, solvaté, or polymorph is contained in one dosage form and a second therapeutic agent (e.g., an antipsychotic, memantine or an AChE-I), or its sait, hydrate, solvaté, or polymorph is contained în another dosage form).
[0081] The term sequential administration as used herein means that the a5containing GABAa receptor agonist (e.g., a a5-containing GABAa receptor positive allosteric modulator) and a second therapeutic agent (e.g., an antipsychotic, memantine or an AChE-I), or their pharmaceutically acceptable salts, hydrates, solvatés, polymorphe, are administered with a time séparation of more than about 15 minutes, and ïn some embodiments more than about one hour, or up to 12-24 hours. Either the a5-containing GABAa receptor agonist (e.g., a a5-containing GABAa receptor positive allosteric modulator) or a second therapeutic agent (e.g., an antipsychotic, memantine or an AChEI) may be administered first. The a5-containing GABAa receptor agonist (e.g., a a5containing GABAa receptor positive allosteric modulator) and a second therapeutic agent (e.g., an antipsychotic, memantine or an AChE-I), or their salts, hydrates, solvents, or polymorphs, for sequential administration may be contained in discrète dosage forms, optionally contained in the same container or package.
[0082] A therapeutically effective amount of a drug or agent is an amount of a drug or an agent that, when administered to a subject will hâve the intended therapeutic effect, e.g. improving cognitive function in a subject, e.g, a patient having cognitive impairment associated with a CNS disorder. The full therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a sériés of doses. Thus, a therapeutically effective amount may be administered in one or more administrations. The précisé effective amount needed fora subject will dépend upon, for example, the subject’s size, health and âge, the nature and extent ofthe cognitive impairment or other symptoms ofthe CNS disorder (such as age-related cognitive impairment, Mild Cognitive Impairment (MCI), dementia, Alzheimer’s Dîsease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, bipolar, ALS, cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autîsm spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction), and the therapeutics or combination of therapeutics selected for administration, and the mode ofadministration. The skilled worker can readily détermine the effective amount for a given situation by routine expérimentation.
[0083] The compounds ofthe present invention also include prodrugs, analogs or dérivatives. The term prodrug is art-recognized and is intended to encompass compounds or agents which, under physiological conditions, are converted into a5containing GABAa R positive allosteric modulators. A common method for making a prodrug is to select moieties which are hydrolyzed or metabolized under physiological conditions to provide the desired compound or agent. In other embodiments, the prodrug is converted by an enzymatîc activity ofthe host animal to a GABAa a5 receptor positive allosteric modulator.
[0084] Analog is used herein to refer to a compound which functionally resembles another chemical entity, but does not share the identical chemical structure. For example, an analog is sufficiently similar to a base or parent compound such that it can substitute for the base compound in therapeutic applications, despite minor structural différences.
[0085] Dérivative is used herein to refer to the chemical modification of a compound. Chemical modifications of a compound can înclude, for example, replacement of hydrogen by an alkyl, acyl, or amino group. Many other modifications are also possible.
[0086] The term aliphatic as used herein refers to a straight chained or branched alkyl, alkenyl or alkynyl. It is understood that alkenyl or alkynyl embodiments need at least two carbon atoms in the aliphatic chain. Aliphatic groups typically contains from 1 (or 2) to 12 carbons, such as from I (or 2) to 4 carbons.
[0087] The term aryl as used herein refers to a monocyclic or bicyclic carbocycüc aromatic ring system. Aryl as used herein includes a (C6-C12)-aryl-. For example, aryl as used herein can be a C6-CI0 monocyclic or C8-C12 bicyclic carbocycüc aromatic ring system. In some embodiments, aiyl as used herein can be a (C6-C10)-aryl-. Phenyl (or Ph) is an example of a monocyclic aromatic ring system. Bicyclic aromatic ring Systems include Systems wherein both rings are aromatic, e.g., naphthyl, and Systems wherein only one of the two rings is aromatic, e.g., tetralîn.
[0088] The term “heterocyclic” as used herein refers to a monocyclîc or bïcyclîc non· aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N, NH, S, SO, or SO2 în a chemically stable arrangement. Heterocyclic as used herein includes a 3- to 12- membered heterocyclyl- having 1-4 heteroatoms independently selected from O, N, NH, S, SO, or SO2 For example, heterocyclic as used herein can be a 3- to 10- membered monocyclîc or 8- to 12- membered bicyclic non-aromatic ring system having I to 4 heteroatom or heteroatom groups selected from O, N, NH, S, SO, or SO2 in a chemically stable arrangement. In some embodiments, heterocyclic as used herein can be a 3- to 10- membered heterocyclyl- having 1-4 heteroatoms independently selected from O, N, NH, S, SO, or SO2. In a bicyclic non-aromatic ring system embodiment of heterocyclyl,” one or both rings may contain said heteroatom or heteroatom groups. In another bicyclic heterocyclyl embodiment, one of the two rings may be aromatic. In yet another heterocyclic ring system embodiment, a non-aromatic heterocyclic ring may optionally be fused to an aromatic carbocycle.
[0089] Examples of heterocyclic rings include 3-1 H-benzimidazol-2-one, 3-( 1 -alkyl)benzimidazol-2-one, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3tetrahydrothiophenyl, 2-morpholino, 3-morpholino, 4-morpholino, 2-thîomorpholino, 3thiomorpholino, 4-thiomorpholino, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazînyl, I-pîperîdînyl, 2pjperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5-pyrazolinyl, 1piperîdinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2-thîazolidinyl, 3-thiazolidiny 1,4thiazolidinyl, 1-ïmïdazolidinyl, 2-imidazolidinyl, 4-imidazolidinyl, 5-imidazolidinyl, indolinyl, tetrahydroquinolinyl, tetrahydroîsoquinolinyl, benzothiolane, benzodithiane, and l,3-dihydro-imidazol-2-one.
[0090] The term heteroaryl as used herein refers to a monocyclîc or bicyclic aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N, NH or S in a chemically stable arrangement. Heteroaryl as used herein includes a 5- to 12membered heteroaryl having 1-4 heteroatoms independently selected from O, N, NH or S. In some embodiments, heteroaryl as used herein can be a 5- to 10- membered heteroaryl having 1-4 heteroatoms independently selected from O. N, NH or S. For
example, heteroaryl as used herein can be a 5- to 10- membered monocyclic or 8- to 12membered bicyclic aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N, NH or S în one or both rings in a chemically stable arrangement. In such a bicyclic aromatic ring system embodiment of heteroaryl:
- both rings are aromatic; and
- one or both rings may contain said heteroatom or heteroatom groups.
[0091] Examples of heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2imidazolyl, 4-imidazolyl, 5-imidazolyl, benzîmidazolyl, 3-isoxazolyl, 4-îsoxazolyl, 5îsoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 210 pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-îndolyl), pyrazolyl (e.g., 2-pyrazolyl), Isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl,
1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, purinyl, pyrazînyl, 1,3,5-triazînyl, quinolinyl (e.g., 2-quînolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3isoquinolinyl, or 4-isoquinolînyl).
[0092] The term cycloalkyl or cycloalkenyl refers to a monocyclic or fused or bridged bicyclic carbocyclic ring system that is not aromatic. For example, cycloalkyl or cycloalkenyl as used herein can be a C3-C10 monocyclic or fused or bridged C8-C12 bicyclic carbocyclic ring system that is not aromatic. Cycloalkenyl rings hâve one or more units ofunsaturatîon. Preferred cycloalkyl or cycloalkenyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, norbomyl, adamantyl and decalinyl.
[0093] As used herein, the carbon atom désignations may hâve the indicated integer and any ïntervenîng integer. For example, the number of carbon atoms in a (Cl-C4)-alkyl group is 1,2,3, or 4. It should be understood that these désignation refer to the total number of atoms in the appropriate group. For example, in a (C3-C10)-heterocyclyl the total number of carbon atoms and heteroatoms is 3 (as in aziridine), 4,5,6 (as in morpholine), 7,8, 9, or 10.
[0094] Pharmaceutically acceptable sait is used herein to refer to an agent or a compound according to the invention that is a therapeutically active, non-toxic base and acid sait form of the compounds. The acid addition sait form of a compound that occurs in its free form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succînic, maleic, fumarîc, malic, tartane, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, ptoluenesulfonic, cyclic, salicylic, p- aminosalicylic, pamoic and the like. See, e.g., WO 01/062726.
[0095] Compounds containing acîdic protons may be converted into their therapeutically active, non-toxic base addition sait form, e. g. métal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base sait forms include, for example, ammonium salts, alkali and earth alkaline meta! salts, e. g., lithium, sodium, potassium, magnésium, calcium salts and the like, salts with organic bases, e. g. N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. Conversely, said sait forms can be converted into the free forms by treatment with an appropriate base or acid.
[0096] Compounds and their salts can be in the form of a solvaté, which is included within the scope of the présent invention. Such solvatés include for example hydrates, alcoholates and the like. See, e.g., WO 01/062726.
[0097] As used herein, the term hydrate refers to a combination of water with a compound wherein the water retains its molecular state as water and is either absorbed, adsorbed or contained within a crystal lattice ofthe substrate compound.
[0098] As used herein, the term polymorph refers to different crystalline forms of the same compound and other solid state molecular forms including pseudo-polymorphs, such as hydrates (e.g., bound water présent in the crystalline structure) and solvatés (e.g., bound solvents other than water) of the same compound. Different crystalline polymorphe hâve different crystal structures due to a different packing of the molécules in the lattice. This results in a different crystal symmetry and/or unit cell parameters which directly influences its physical propertîes such the X-ray diffraction charactenstics of crystals or powders. A different polymorph, for exampie, will ln general dîffract at a different set of angles and will give different values for the intensifies. Therefore X-ray powder diffraction can be used to identity different polymorphs, or a solid form that comprises more than one polymorph, in a reproducible and reliable way. Crystalline polymorphie forms are of interest to the pharmaceutical industry and especially to those involved in the development of suitable dosage forms. If the polymorphie form is not held constant during clinical or stability studies, the exact dosage form used or studîed may not be comparable from one lot to another. It is also désirable to hâve processes for producing a compound with the selected polymorphie form in high purity when the compound ïs used in clinical studies or commercial products since Impurities présent may produce undesired toxicologîcal effects. Certain polymorphie forms may exhibit enhanced thermodynamic stability or may be more readily manufactured in high purity în large quantifies, and thus are more suitable for inclusion in pharmaceutical formulations. Certain polymorphs may display other advantageous physical propertîes such as lack of hygroscopic tendencies, improved solubility, and enhanced rates of dissolution due to different lattîce energies.
[0099] This application contemplâtes ail the isomers of the compounds of formulae IIV. “Isomer” as used herein includes optical isomers (such as stereoisomers, e.g., enantiomers and diastereoisomers), Z (zusammen) or E (entgegen) isomers, and tautomers. Many ofthe compounds useful in the methods and compositions ofthis invention hâve at least one stereogenîc center in their structure. This stereogenîc center may be présent in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem. (1976), 45,11-30. The invention also relates to ail stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds or mixtures thereof (including ail possible mixtures of stereoisomers). See, e.g., WO 01/062726. Furthermore, certain compounds which contain alkenyl groups may exist as Z (zusammen) or E (entgegen) isomers. In each instance, the invention includes both mixture and separate individual isomers. Multiple substituents on a pïperïdinyl or the azepanyl ring can also stand în either ci s or trans relatîonship to each other with respect to the plane of the piperidinyl or the azepanyl ring. Some ofthe compounds may also exist in tautomeric forms. Such forms, although not explicitly indicated in the formulae described herein, are intended to be included within the scope ofthe présent invention. With respect to the methods and compositions of the présent invention, reference to a compound or compounds is intended to encompass that compound în each of its possible isomeric forms and mixtures thereof unless the particular isomeric form is referred to specifically. See, e.g., WO 01/062726.
[0100] The compounds of the invention enhance the function of a5-contaïning GABAa R, i.e., they are a5-containing GABAa R agonists (e.g., a5-containing GABAa receptor positive allosteric modulators) and are capable of increasing GABA-gated Cl' currents.
[0101] The invention further provides pharmaceutical compositions comprising one or more compounds ofthe invention together with a pharmaceutically acceptable carrier or excipient. In some embodiments, the pharmaceutical compositions of this application may further comprise a second therapeutic agent, such as an antipsychotic, memantine or an AChE-I.
[0102] The invention further provides methods for treating cognitive impairment associated with said CNS disorders that are responsive to positive allosteric modulators of a5-containing GABAa receptor, e.g., age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenîa, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome,, compulsive behavior, and substance addiction. In certain embodiments, the method is a method of treating the age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenîa, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In certain embodiments, treatment comprises preventing or slowing the progression of a CNS disorder as described herein (such as those described herein). In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with the CNS disorder. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive déficit. In another aspect of the invention, there îs provided a method of preserving or improving cognitive function in a subject in need thereof, the method comprising the step of administering to said subject a therapeutically effective amount of a compound ofthe invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
[0103] The various CNS disorders with cognitive impairment (e.g., age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapyrelated cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome,, compulsive behavior, and substance addiction) may hâve a variety of étiologies. However, the symptom of cognitive impairment in each ofthe above-mentioned disorders may hâve overiapping causes. Thus, a composition or method of treatment that treats cognitive impairment in one CNS disorder may also treat cognitive impairment in another.
Benzodiazépine Dérivatives [0104] The présent invention provides a compound of formula I:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
A is C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6h;
E is N, NR7, CR6 or C(R6)2;
W îs N, NR7, CR6 or C(R6)2;
X is N, NR7,0, CR6 or C(R6)2;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
V îs C or CR6, or when Z is C or CR6, V îs C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is
, then R2 îs -OR8,
-SR8, -(CH2)nOR8, -(CH2)nO(CH2)nR8, -(CH2)PR8 and -(CH2)nN(R”)R10; and wherein
R2 is independently substituted with 0-5 R';
m and n are independently integers selected from 0-4;
p is an integer selected from 2-4;
each occurrence of the bond “ ” îs either a single bond or a double bond;
each occurrence of R1, R2, R4, and Rsare each independently selected from:
halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SÎR3, -N(R)2, -SR, -SOR, -SO^R, -SO2N(R)2, -SOjR, -(CR2)i.3R, -(CR2)|.3-OR, -(CR2^3-C(O)NR(CR2k3R, -(CR^-CtOÏNRfCRiJwOR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(Rh, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)o-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR); RJ is absent or îs selected from:
halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR, -SO2R, -SO2N(R)2, -SOjR, -(CR2)|.3R, -(CR2)i-3-OR, -(CR2)o.3-C(0)NR(CR2)o.3R, -(CR2)o.3-C(0)NR(CR2K3OR, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, • 41
-C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2, -OC(0)N(R)2, -C(S)N(R)2, -(CR2)o.3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R,
-C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
each R6 is independently -H or -(Cl-C6)alkyl;
each R7 is independently -H or -(C 1 -C6)alkyl;
each R8 is independently -<C1-C6)alkyl, -(C3-C10)-cycloalkyl, (C6-CI O)-aryl, or 5- to
10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’;
each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of Rt0 is independently substituted with 0-5 R’;
each R is independently selected from:
H-, (CI-C12)-altphatîc-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloalkenyl-, [(C3-C10)-cydoalky l]-(C 1 -C 12>al iphatic-, [(C3-C10)-cycloalkeny 1]-(C 1 -C 12)-alîphati c-, [(C3-C l0)-cycloalkyl]-O-(C l-C12>aliphattc-, [(C3-C10)-cydoalkeny l]-O-(C I -C 12)-al i phat ic-, (Cô-ClO)-aryl-, .
(C6-C10)-ary 1-(C 1 -C 12)al iphatic-, (C6-C l0)-aiyl-O-(Cl-C12)al iphatic-, (C6-C10)-aryl-N(R”)-(C I -C 12)al i phat ic-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl )-(C 1 -C 12)al iphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-N(R’*)-(CI-C12)alîphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroarylXCl-C12)-al iphatic-, (5- ίο 10- membered heteroaryl)-O-(Cl-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aliphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R îs independently substituted with 0-5 R*;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R*, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3C 10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCFj and -N(R”)2;
wherein each occurrence of R” is independently selected from H, -(Cl-C6)-alkyl, -(ClC6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)aryl-(CI-C6)-alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10)aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR°, -CH2NR°2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN,
-CFj, -OCFj and -N(R°)2, wherein each occurrence of R° is independently selected from: —(C 1 -C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-C10)-aryl-.
[0105] In some embodiments, the présent invention provides a compound of formula I:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
Ais C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6h;
EisN,NR7,CR6orC(R6)2;
WisN, NR7, CR6orC(R6)2;
X is N, NR7,0, CR6 or C(R6h;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot 15 both be N;
Vis Cor CR6, or when Z is C or CR6, V is C, CR6, or N;
NX , « whereîn when the ring formed by X, Y, Z, V and W is N , then Rz is -OR,
-SRs, -(CH2)nORe, -(CH2)nO(CH2)nR8, -(CH2)pR8 and -ÎCH2)nN(R”)Rt0; and wherein
R2 is independently substituted with 0-5 R’;
m and n are independently integers selected from 0-4;
p is an integer selected from 2-4;
each occurrence of the bond “ ” is either a single bond or a double bond;
each occurrence of R1, R2, R4, and Rsare each independently selected from:
halogen, -R, -OR, -N02, -NCS, -CN, -CFj, -OCF3, -S1R3, -N(R)2, -SR, -SOR, -SO2R, -SO2N(R)2, -SO3R, -(CR2)i.3R, -<CR2)i.3-OR, -(CR2)o-3-C(0)NR(CR2)q.3R, -(CR2)o.3-C(0)NR(CR2)o.30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(Rh,
-OC(O)N(R)2, -C(S)N(R)2, -(CR2)o-jNHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR,
-N(R)N(R)C0N(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(ü)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
R3 is absent or is selected from:
halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR, -SO2R, -SO2N(R)2, -SO3R, -(CR^R, -(CR2)i-3-OR, -(CR2)o-3-C(0)NR(CR2)o.3Ri -(CR2)o.ï-C(0)NR(CR2)o.30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2i
-OC(O)N(R)2, -C(S)N(R)2, -(CR2)ojNHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR,
-N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
each R6 is independently -H or -(C 1 -C6)alky 1 ;
each R7 is independently-H or -(C 1-C6)alkyl;
each R8 is independently -(Cl-C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to
10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’;
each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of Rt0 is independently substituted with 0-5 R*;
each R is independently selected from:
H-, (Cl-C12)-aliphatic-, (C3-C10)-cycloalkyl-, (C3-C 10)-cycloalkenyl-, [(C3-C10)-cycloalkyl]-(CI-C12)-aliphatîc-,
[(C3-C10)-cycloalkenyl]-(C I -C 12)-a I iphati c-, [(C3-C10>cycIoalkyI]-O-(C 1 -C 12>al îphat i c-, [(C3-C 10)-cycloalkenyl]-O-(C 1-C 12)-aliphatic-, (C6-CI0)-aryl-, (C6-C10)-aryl-(Cl-C12)aliphatic-, (C6-CI O)-ary l-O-(C 1 -C 12)aliphatic-, (C6-C10)-ary 1-N(R”)-(C 1 -C 12)aliphatic-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)aIiphatic-, (3-to 10-membered heterocyclyl)-O-(C 1-C 12)aliphatic-, (3- to 10- membered heterocyclyI)-N(R”)-(Cl-C12)aliphatic-,
5-to 10-membered heteroaryl-, (5- to 10- membered heteroary!)-(Cl-C12)-aliphatic-, (5- to 10- membered heteroaryI)-O-(Cl-C12)-aIiphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aliphatîc-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R is independently substituted with 0-5 R*;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C325 C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCF3 and -N(R”)s wherein each occurrence of R” is independently selected from H, -(C 1 -C6)-alkyl, (C330 C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (Cô-ClO)-aryl-O-(ClC6)-alkyl-.
[0106] Some embodiments provide a compound of formula I:
W
I, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
Ais C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6)2;
EisN,NR7,CR6orC(R6)2;
W is N, NR7, CR6 or C(R6)2;
X is N, NR7,0, CR6 or C(R6);;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
Vis Cor CR6, or when Z is C or CR6, V is C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is then R2 is -OR8,
-SR8, or -(CH2)nOR8;
m and n are each independently an integer selected from 0-4;
each occurrence of the bond “ ” is either a single bond or a double bond;
each occurrence of R1, R1, R4, and R3 are each independently selected from: halogen, -R, -OR, -NO2, -NCS, -CN, -CFj, -OCF3, -SiR3, -N(R)2, -SR, -SOR,
-SO2R, -SOiNtRh, -SO3R, -(CR2)|.3R, -(CR2)i-3-OR, -(CR2)o.3-C(0)NR(CR2)o.3R. -(CR2)o-3-C(0)NR(CR2)o-30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)o.3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R):, -N(R)SO2R, -N(R)SO2N(R)2i -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
R3 is absent or is selected from:
halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -S1R3, -N(R)2, -SR, -SOR, -SO2R,-SO2N(R)2,-SO3R,-(CR2)I.ÎR, -(CR2)!.3-OR, -(CR2)o.3-C(0)NR(CR2)o.3R, -(CR2)o-3-C(0)NR(CR2)o-30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R, -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R):, -OC(O)N(R)2, -C(S)N(R)2, -<CR2)q.3NHC(O)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R)2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and-P(O)(H)(OR);
each R6 is independently -H or -(Cl-C6)a!kyl;
each R7is independently-H or -(Cl-C6)a!kyl;
each Rg is independently -(Cl-C6)alkyl, -(C3-C10)-cycloa!ky!, (C6-C10)-aryl, or 5- to
10- membered heteroaryl, wherein each occurrence of Rs is independently substituted with 0-5 R*;
each R is independently selected from:
H-, · (C 1 -C 12)-aliphatic-, (C3-C10)-cycloalkyl-, (C3-C 10>-cyc!oalkenyl-, [(C3-C10)-cycloa!kyl]-(C 1 -C 12)-a! iphatic-, [(C3-CI0)-cycloalkenyl]-(Cl-C12)-a!iphatic-, [(C3-C10)-cycloalkyl]-O-(C 1 -C 12)-al iphatic-, [(C3-C10)-cycloalkeny!]-0-(C 1 -C 12)-al iphatic-, (C6-C10)-aryl-,
(C6-C10)-ary 1-(C 1 -C 12) a 1 i phatic-, (C6-C10)-ary 1-O-(C 1 -C 12)al iphat i c-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3-to 10- membered heterocyclyl)O-(Cl -Cl 2)aliphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl>(Cl-C12)-aliphatîc-, and (5- to 10- membered heteroaryl)-0-(Cl-C12)-alÎphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, 10 SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R is independently substituted with 0-5 R’;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or non15 aromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring îs optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, 20 -CH2OR”, -CH2NR”2, -C(O)N(R”h, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCFj and -N(R”)2;
wherein each occurrence of R” is independently selected from H, -(Cl-C6)-alkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)25 alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(Cl-C6)alkyl-.
[0107] The présent invention provides a compound of formula I:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
Ais C, CR6, or N;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6>2;
EisN.NR7, CR6orC(R6)2;
WisN.NR7, CR6orC(R6)2;
X is N, NR7,0, CR6 or C(R6>2;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot 15 bothbeN;
Vis Cor CR6, or when Z is C or CR6, V is C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is
, then R2 is
-(CH2)nOR8 or -(CH2)nO(CH2)nR8; and wherein R2 is independently substituted with
0-5 R’;
m and n are independently integers selected from 0-4;
p is an integer selected from 2-4;
each occurrence of the bond “ ” is either a single bond or a double bond;
each R* îs independently selected from: halogen, -R, and -OR;
R2 is selected from: halogen, -R and -(CR2)|.î-0R;
R3 îs selected from: -R and -CN;
R4 and R3 are each independently -H or -(C I -C6)alkyl;
each R6is independently-H or -(C 1-C6)alkyl;
each R7 is independently -H or -(C 1-C6)alkyl;
each R8 is independently -(Cl-C6)alkyl, -(C3-CI0)-cycloalkyl, (C6-C10)-aryl, or 5· to
10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’;
each R is independently selected from:
H-, (C I -C 12)-al i phatic-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloalkenyl-, [(C3-C10)-cyc loal ky 1] -(C 1 -C 12)-al i phatic-, [(C3-C10)-cycloalkenyl]-(C 1 -C 12)-al iphatic-, [(C3-C10)-cycloalkyl]-O-(C 1 -C 12)-al iphat ic-, [(C3-C10)-cycloalkenyl]-O-(Cl-C12)-aliphatic-, (C6-C10)-aryl-, (C6-C 10)-aryl-(C 1-Cl 2)al iphatic-, (C6-C10)-aryl-O-(C 1 -C 12)al i phatic-, (C6-C10)-ary 1-N(R* >(C 1 -C 12)al i phatic-,
3- to 10- membered heterocyclyl-, (3-to 10- membered heterocyclyl)-(CI-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)aliphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl)-(Cl-C12)-al iphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)-aliphatîc-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aHphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R îs independently substituted with 0-5 R’;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to whîch they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R*, and wherein said ring 5 îs optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCFj and -N(R”h;
wherein each occurrence of R” is independently selected from H, ~(Cl-C6)-alkyl, -{C 1 C6)-alîphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, (C6-C10)-aryl-, (5-to 10- membered heteroaryl)-(Cl-C6)alkyl-, (C6-C10)-ary!-(CI-C6)-alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)alkyl-, and (C6-C10)-ary!-O-(Cl-C6)-alkyl-, wherein each occurrence of R” îs independently substituted with 0-5 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR°, -CH2N(R°)2, -C(O)N(Ro)2, -C(O)OR°, -NO2, -NCS, -CN, -CFj, -OCFj and -N(R°)2, wherein each occurrenceof R°is independently selected from:-(ClC6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-C10)-aryl-, [0108] The présent invention provides a compound of formula I:
I, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
AisC, CR6, or N;
B and F are each independentiy selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7,0, CR6 or C(R6h;
E is N, NR7, CR6 or C(R6)2;
W is N, NR7, CR6 or C(R6)2;
X is N, NR7,0, CR6 or C(R6)2;
Y and Z are each independentiy selected from C, CR6, and N, wherein Y and Z cannot both be N;
Vis Cor CR6, or when Z is C or CR6, V is C, CR6, or N;
wherein when the ring formed by X, Y, Z, V and W is then R2 is
-(CH2)nOR8 or -(CH2)nO(CH2)nR8, wherein each occurrence of R8 is independentiy (Cl-C6)alkyl or (C6-C10)-aryl (e.g., phenyl), and wherein R2 is independentiy substituted with 0-5 R’;
m and n are independentiy integers selected from 0-4 (in some embodiments, m is 1); p îs an integer selected from 2-4;
each occurrence of the bond “ ” îs either a single bond or a double bond;
each R1 is independentiy selected from: -Cl, -F, -OMe, and -C=CH;
R2 is halogen, -(CR2)i-3-OR, wherein each occurrence of R is independentiy selected from -H, -(Cl-C6)alkyl, (C6-C10)-aryl- (e.g., phenyl), and (C6-C10)-aryl-(ClC12)aliphatic- (e.g., phenyl-(Cl-C6)alkyl-), and wherein each occurrence of R is independentiy substituted with 0-5 R’;
R3 is selected from: -CN, -C=CH, -C=C-(Cl-C6)alkyl, -CsC-phenyl, .ΟΝ'
«wherein R3 îs substituted with 0-5 R’;
each occurrence of R4 and Rs is independentiy -H or -(Cl-C6)alkyl;
each R6 îs independentiy -H or -(C 1 -C6)alkyl;
each R7is independentiy-H or-(Cl-C6)alkyl;
wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CHjOR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -N02, -NCS, -CN, -CF3, -OCF3 and -N(R”)2;
wherein each occurrence of R” is independently selected from H,-(Cl-C6)-alkyl, *-(Cl5 C6)-aliphatic, (C3-C6)-cycloaikyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)alkyl-, (C6-C10)-aryl-(CI-C6)-alkyl-, (5- to 10- membered heteroaryl)-0-(Cl-C6)alkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-5 substituents selected from: halogen, -R°, -OR°, 10 oxo, -CH2OR°, -CH2NR° 2, -C(O)N(R°)2, -C(O)OR0, -NO2, -NCS, -CN, -CFj, -OCF3 and -N(R°)2, wherein each occurrence of R° is independently selected from: —(C I C6)-al iphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-C10)-aryl-.
In some of the above embodiments, R3 is selected from:
wherein each occurrence of R is independently selected from -(CI-C6)-alkyl (e.g., linear or branched), -C^CH, phenyl, thiophene, (5- to 10- membered heteroaryl)-(ClC6)-alkyl-, (C6-C10)-aryl-(Cl-C6)-alkyl-, wherein each R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR”, oxo, -CH2OR°, 20 CH2NR°2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R°)2, wherein each occurrence of R°is independently selected from: -(Cl-C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, and (C6-C10)-aryk [0109] In some embodiments ofa compound of formula I, X, Y, Z, V and W together form a 5-membered aromatic or non-aromatic ring having 1-4 nitrogen atoms, wherein said ring is substituted with 0-3 R6 and 0-2 R7. In some embodiments, X, Y, Z, V and W together form a 5-membered aromatic ring having 1-3 nitrogen atoms, wherein said ring is substituted with 0-2 R6 and 0-1 R7.
[OUI] In some embodiments, X, Y, Z, V and W form a ring that is selected from:
< X rNv N'N<^SRe N'N^(CH2)nORe < X N'N<^(CH2)nO(CH2)nRe
X
Γ*1 'n^CCHAR· and
(CH2)nN(R)R10 [0112] In some embodiments ofa compound of formula I, W is N. In some embodiments, W is N, and X, Y, Z, V and W form a ring that is selected from:
[0113] In some embodiments, W is N, and X, Y, Z, V and W form a ring that is selected from:
(CH2)nN(R )R and [0114] In certain embodiments of a compound of formula I, the ring formed by X, Y, Z,
V and W is:
[0115] In certain embodiments of a compound of formula I, the ring formed by X, Y, Z,
[0116] In certain embodiments of a compound of formula I, the ring formed by X, Y, Z,
V and W is selected from:
< X fl \ NxN<^0Re
K x
H n^n<^sr’
K x fl N\ NxN<^(CH2)nOR’ < X fl N\ N^N‘i^(CH2)nO(CH2)nR’ (CHjjpR8 < X a \ N'f/^(CH2)nN(R)R10 and [0117] In certain embodiments of a compound of formula I, the ring formed by X, Y, Z, V and W is selected from:
(CH2)nOR8 . In some
embodiments, the ring formed by X, Y, Z, V and W is: embodiments, the ring formed by X, Y, Z, V and W is:
. In some
(CHzJnOfCHjJnR8
[0118] In some embodiments of a compound of formula I, A, B, D, E and F together form a 5-membered aromatic or non-aromatic ring having 1-4 nitrogen atoms, wherein said ring is substituted with 0-3 R6 and 0-2 R7. In certain embodiments, A, B, D, E and F together form a 5-membered aromatic ring having 1-3 nitrogen atoms, wherein said ring is substituted with 0-2 Rs and 0-1 R7.
[0119] In some embodiments of a compound of formula I, A, B, D, E and F form a ring that is selected from:
[0120] In certain embodiments of a compound of formula I, the ring formed by A, B, D,
F and E is:
[0121] In some embodiments of a compound of formula I, the compound has a structure of formula II:
II.
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein m, R1, R2, R3, R4, Rs and R6 are as defined in formula I.
[0122] In some embodiments of a compound of formula I, the compound has a structure of formula III:
III.
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein m, R1, R2, R3, R4, R3 and R6 are as defined in formula I.
[0123] In some embodiments of a compound of formula I, the compound has a structure of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein R2 îs -OR’, -SR8, or -(CH2)nOR8, wherein R2 is independently substituted with 0-5 R’ and wherein m, n, R*, R3, R4, R3, R6, and R8 are as * a 4 defined in formula I. In some embodiments, R is -OR . In some embodiments, R is -(CH2)nOR8.
[0124] In some embodiments of a compound of formula I, the compound has a structure of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein R2 is -(CH2)nO(CH2)nR8, -(CH2)pR8 or -(CH2)nN(R”)R10, wherein R2 is independently substituted with 0-5 R’ and wherein m, n, p, R1, R3, R4, R3, R6, R8, R10, and R” are as defined herein. In some embodiments, R2 is -(CH2)nO(CH2)nR8.
[0125] In some embodiments of a compound of formula I, II, III, or IV, each occurrence of R1 is selected from: halogen, -R, -OR, -NO:, -CN, -CF3, -OCF3, -N(R)2, and -N(R)SO2R, wherein each occurrence of R is independently substituted with 0-5 R'. In some embodiments, each occurrence of R1 is independently selected from: halogen, -H, 5 (C I-C6)alky 1, -OH, -O((C I -C6)alky 1), -NO2, -CN, -CF3, -OCF3, -NH2,
-N((C I -C6)alkyl)2, -N((C I -C6)alky 1)SOX(C I -C6)alkyl), and -NHSO:((C I -C6)alkyl), wherein said alkyl is independently substituted with 0-5 R’. In certain embodiments, each occurrence of R1 is independently selected from: -H, -F, -Cl, -Br, -OH, -Me, -Et, OMe, -OEt, -NO2, -CN, -CF3, -OCF3, -NH2, -NMe2, -NEt2, -NHSO:Me, and -NHSO2Et.
In certain embodiments of a compound of any one of formulae I-IV, at least one R1 is -OR. In some embodiments, the at least one R* is -O((Cl-C6)alkyl), such as -OMe. [0126] ln some embodiments of a compound of formula I, II or 111, R3 is selected from: halogen, -R, -OR, -NO:, -(CR2)i-3R, -(CR2)i-3-OR, -CN, -CF3, -C(O)NR2, -C(O)OR, and -OCF3, wherein each occurrence of R is independently substituted with 0-5 R*. In some embodiments, R2 îs selected from:
-H, -<C1-C6)alkyl, -CH2-O((CI-C6)alkyl ), -(C((Cl-C6)alky l)2)t.3-O((C 1-C6)alkyl), -OH, -O((CI-C6)alkyl), -NO:, -CN, -CF3, -OCF3, (C3-C10)-cycloalkyl-, -C(O)N((C1-C6)alkyl)2, -C(O)O((CI-C6)alkyl), 3- to 10- membered heterocyclyl-, (C6-C10)aryI-, 5- to 10- membered heteroaiyl-, (C6-C10)ary 1-(C 1 -C 12)aliphatic-, (C6-C10)aryl-O-(C I-C12)aliphatic-, (C6-C10)ary l-N(R”)-(C 1 -CI 2)aliphatic-,(C6-C 10)aryl-(C I -C 12)aliphatic-O-, (5- to 10-membered heteroaryl)-(Cl-C12)-aliphatic-, (5- to 10-membered heteroaryl)-0-(Cl-C12)-aliphatic-, (5-to 10-membered heteroaryl)-N(R”)-(Cl-Cl 2)-aliphatic-, (5- to 10-membered heteroaryl)-(CI-C12)-aliphatic-0-, (3-to 10-membered heterocyclyl)-(CI-CI2)alîphatic-, (3- to 10-membered heterocyclyl)-O-(CI-C12)aliphatic-, (3- to 10-membered heterocydyl)-N(R”)-(CI-C12)aliphatic-, and (3- to 10- membered heterocyclyl)-(CI-C12)aliphatic-0-, wherein R2 is independently substituted with 0-5 R’.
[0127] In some embodiments of a compound of formula I, Il or III, R2 is selected from: -H, -Me, -Et, propyl, isopropyl, butyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -CFj, -C(O)OMe, -C(O)OEt, -OMe, -CH2OMe, -CH2OEt, -CH2OPh, -CH2-pyrrolidine, -CH2-morpholine, -CH2-pyrîdîne, and -CH2Ph, wherein said R2 ts substituted with 0-3 R’. In some embodiments of a compound of formula I, II or III, R2 is -Me substituted with 0-3 R’ selected from -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -CtOïNtR’^, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCF3 and -N(R”)2, wherein R” is independently selected from H, -<CI-C6)-alkyl, (C6-CI0)-aryl-, and (C6-CI0)-aryl-(CIC6)-alkyl-. In some embodiment, R2 is -Me that is independently substituted wîth 0-3 R’ 10 selected from -N(Me)2, -N(Et)2 and -N(Me)(CH2Ph).
[0128] In some embodiments of a compound of formula 1,11 or III, R2 is selected from: -CH2Ph, -CH2CH2Ph, -Ph, -OCH2Ph, -CH2OPh, -OCH2CH2Ph, -CH2CH2OPh, -CH2pyrrolidine, -CH2-morpholine, -CH2-pyridîne, and -CH2Ph wherein said Ph, pyrrolidine, pyridine or morpholine Îs substituted with 0-5 R’. In some embodiments of a compound 15 of formula I, Il or III, R2 is selected from: -CH2Ph, -CH2CH2Ph, -Ph, -OCH2Ph, CH2OPh, -OCH2CH2Ph, -CH2CH2OPh, -CH2-pyrrolidîne, -CH2-morpholine, -CH2pyridine, and -CH2Ph, wherein said Ph, pyrrolidine, pyridine or morpholine îs substituted with 0-5 R’ independently selected from halogen, (CI-C6)-alkyl, -OH, -O((CI-C6)alkyl), -CH2OH, -CH2O(Cl-C6)-alkyl), -CH2N(CI-C6)-alkylh, -C(O)O(CI-C6)-alkyl), 20 -C(O)N(CI-C6)-alkyl)2, -NO2, -CN, -CF3, -OCFj and -N(CI-C6)-alkyl)2. In some ofthe above embodiments, the-Ph, pyrrolidine, pyridine or morpholine of R2 is substituted with 0-5 R’ independently selected from -F, -Cl, -CN, -Me, -Et, -OMe, and -OEt. In some embodiments of a compound of formula I, II or III, R2 is -CH2Ph,-CH2OPh, -CH2pyridtne, -CH2-pyrrolidine, or -CH2-morpholine wherein said -Ph, pyrrolidine, pyridine 25 or morpholine is substituted with 0-3 R’ independently selected from -F, -Cl, -CN, -Me, and -OMe.
[0129] In some embodiments of a compound of formula IV, R2 is -OR8, -SR8,
-(CH2)„OR8, -(CH2)nO(CH2)nR8, -(CH2)PR8 or -(CH2)nN(R”)R10, wherein each R8 îs independently -(C I-C6)alky I, -(C3-CI0)-cycloalkyl, (C6-CI0)-aryl, or 5- to 1030 membered heteroaryl, wherein each occurrence of R8 is independently substituted with 05 R’; n is an integer selected from 0-4; p is an integer selected from 2-4; and each R10 îs independently -(C3-CI0)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or
5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently * B * substituted with 0-5 R’. In some embodiments, R is OR . In some embodiments, R is OR8, wherein R8 is (C6-CI0)-aryl, substituted with 0-5 R*. In some embodiments, R2 is OR8, wherein R8 is (C6-CI0)-aryl, substituted with 0-3 halogen (such as -F). In some embodiments, R2 is -(CH2)nOR8 or -(CH2)nO(CH2)nRs. In some embodiments, R2 is -(CH2)nOR8 or-(CH2)nO(CH2)nR8, whereîn R8 is -(C 1-C6)alkyl, (C6-CI0)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’.
[0130] In some embodiments of a compound of formula I, II, III, or IV, R3 is selected from: halogen, -R, -CN, -CF3, -SCfeR, -C(O)N(R)2, -C(O)R and -C(O)OR, wherein each occurrence of R is independently substituted with 0-5 R’. In some embodiments, R3 is selected from: -F, -Br, -Cl, -(CI-C6)alkyI, -CN, -C=C, -CF3, -SQ2((CI-C6)alkyl), -C(O)N((CI-C6)alkyI)2, -C(O)NH2, -C(O)((CI-C6)alkyl),
-SO2((C6-C10)-aryl), -C(O)O((Cl-C6)aIkyl), -(C2-C6)-alkenyI, <C2-C6)-alkynyl,
-(C6-C10)-aryl, 5- to 10- membered heteroaryl-, and 3- to 10- membered heterocyclyl-, wherein said alkyl, alkenyl, alkynyl, aryl, heteroaryl or heterocyclyl- is independently substituted with 0-5 R’. In some embodiments of a compound of formula I, II, III, or IV, R3 is selected from: -H, -C(O)OMe, -C(O)Et, -C(O)NMe2, -C(O)NH2, -C(O)OEt, -C(O)OCH2(/er/-butyl), -C(O)OCH2CF3, -C(0)0(isopropyI),-C(0)NEt2,-CHF2, -CN,
-C=C, -SO2Me, -SO2Et, -SO2Ph(Me), -CFj, -CHF2, -Me, -Et, -Br, -Cl, -CH2Ph,
wherein R9 is selected from -H, -Me, -Et, -CF3, isopropyl, -OMe, -OEt, -O-isopropyl, -CHîNMeî, -ZerZ-butyl and cyclopropyl.
[0131] In certain embodiments of a compound of formula I, II, III, or IV, R3 is
-C(O)OMe or -C(O)OEt. In certain embodiments of a compound of formula I, II, III, or
IV, R3 is , wherein R9 is selected from -H,
-Me, -Et, -CF3, isopropyl, -OMe, -OEt, -O-isopropyl, -CHjNMeî, -ferf-butyl and cyclopropyl.
[0I32| In some embodiments of a compound of formula I, II, III, or IV, R4 and Rs are 10 each independently selected from -H, halogen and -R, wherein each occurrence of R is independently substituted with 0-5 R’, or R4 and R5 may be taken together with the
carbon atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-3 additional heteroatoms independently selected from N, O, S, SO, and SO2, wherein said ring is substituted with 0-5 R*. In some embodiments, R4 and Rs are each independently selected from -H, -Me, -Et, -F, or R4 and R3 are taken together 5 with the carbon atom to which they are bound to form a 3- to 8-membered aliphatic ring.
In certain embodiments, both R4 and Rsare -H.
[0133] In some embodiments, the présent invention provides a compound of formula II:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3 (e.g., m is 1);
each R1 is independently selected from: -Cl, -F, -OMe, and -C=CH;
R2 is halogen or-(CR2)i-3-OR, wherein each occurrence of R is independently selected from -H, -(Cl-C6)alkyl, (C6-CI0)-aryl- (e.g., phenyl), and (C6-C10)-aryl-(ClC12)aliphatic- (e.g., phenyl-(Cl-C6)alkyl-), and wherein each occurrence of R is independently substituted with 0-5 R’;
R3 is selected from: -CN, -C=CH, -C=C-(Cl-C6)alkyl, -C^C-phenyl, φ iù -Kl·0 X? , and .wherein R3 is substituted with 0-5 R’;
each occurrence of R4 and RJ is independently-H or -(C 1 -C6)alky 1;
each R6 is independently -H or -(Cl-C6)alkyl;
wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo,
-CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2) -NCS, -CN, -CF3, -OCF3 and -N(R”h;
wherein each occurrence ofR” is independently selected from H, -(CI-C6)-alkyl, -(ClC6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl -C6)alkyl-, (C6-CI0)-aryl-(Cl-C6)-alkyl-, (5- to 10- membered heteroaryl )-O-(Cl-C6)alkyl-, or (C6-CI0)-aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-5 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR°, -CH2N(R°)2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R°)2, wherein each occurrence ofR°is independently selected from: —(ClC6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-C10)-aryl-.
*
In some of the above embodiments, R is selected from:
wherein each occurrence ofR” is independently selected from-(CI-C6)-alkyl (e.g., linear or branched), -C=CH, phenyl, thiophene, (5- to 10- membered heteroaryl)-(ClC6)-alkyl-, and (C6-C10)-aryl-(Cl-C6)-alkyl-, wherein each R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR°, oxo, -CH2OR°, CH2N(R°)2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R°)2, wherein each occurrence of R° îs independently selected from: -(Cl-C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, and (C6-CI0)-aryl-.
[0134] In some embodiments, the présent invention provides a compound of formula II:
Π, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independentiy selected from: halogen (e.g.. Cl, F), -H, -(Cl-Cô)alkyl, -OH, -O((CI-C6)alkyl) (e.g., -OMe), -NOj, -CN, -CF3, and -OCF3, wherein R1 is independentiy substituted with 0-5 R’;
R2 is selected from:
-H, halogen, -(Cl-CÔ)alkyl, -OH, -O((Cl-C6)alkyl), -C(O)O((Cl-C6)alkyl), C(O)NR2, (C6-C10)-aryl- (e.g., phenyl), (C6-C10)-ary 1-(C 1 -C 12)aliphatic-, (C 6-C10)-ary 1-O-(C 1 -C 12)al iphatic-, (C6-C 10)-aryl-N(R”)-(C 1 -C 12)aliphatic-, (5- to 10- membered heteroaryl)-(Cl-C12)aliphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)al iphatic-, (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-C12)aliphatic-, and (3-to 10-membered heterocyclyl)-N(R’*)-(Cl-C12)aliphatic-, wherein R2 is independentiy substituted with 0-5 R*;
R3 is selected from:
-(Cl-Cô)alkyl, -(C2-C6)alkenyl (e.g., -CH=CH2), -C^CH, -CN, halogen (e.g., Br), -SO2((C6-C10)-aryl), -SO2((Cl-C6)alkyl), -C(O)N((CI-C6)alkyl)2, -C(O)NH2, -C(O)O((Cl-C6)alkyl), -C(O)((Cl-C6)alkyl), -(Cô-ClO)aiyl, 5- to 10- membered
heteroaryl (e.g., 5-membered heteroaryl such as an optionally substituted and 5- to 10- membered heterocyclyl (e.g., 5-membered heterocyclyl such as an optionally substituted
), wherein R3 is independently substituted with 0-5 R’;
R4 and R3 are each independently selected from -H, halogen and -(Cl-C6)alkyl; R6 is selected from -H and -(Cl-C6)a!kyl;
each R is independently selected from:
H-, (Cl-C12)-aliphatic-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloalkenyl-, [(C3-C10)-cycloalkyll-(C I -C 12)-al iphatic-, [(C3-C10)-cycloalkenyl]-(C 1 -C 12)-al i ph atic-, [(C3-C10)-cycloalky 1]-O-(C I -C 12)-aI iphatic-, [(C3-C10)-cycloalkeny l]-O-(C I -C 12)-a! iphatic-, .(C6-C10)-aryl-, (C6-C10)-ary 1-(C I -C 12)a 1 iphatic-, (C6-C10)-ary 1-O-(C 1 -C 12)a 1 iphatic-, (C6-C10)-aryl-N(R’ ’)-(C 1 -C 12)al i phat î c-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)al iphatic-, (3- to 10- membered heterocyclyl)-O-(CI-C12)aliphatic-, (3- to 10- membered heterocyclyl)-N(R*’)-(Cl-C12)aliphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl)-(Cl-C12)-aliphatic-, (5- to 10- membered heteroaryl )-O-(C 1 -C 12)-al iphatic-; and (5- to 10- membered heteroaryl)-N(R*’)-(Cl-C12)-aliphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S,
SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N,
NH, O, and S;
wherein each occurrence of R is independently substituted with 0-5 R*;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R*, and wherein said ring is optionally fûsed to a (C6-C 10)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R”h;
wherein each occurrence of R” is independently selected from H, -(Cl-C6)-aIkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10-membered heteroaryl-, (C6C10)-aryl-, (5- to 10- membered heteroaryl)-(CI-C6)-alkyl-, (C6-C10)-aryl-(CI-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-a!kyl-, and (C6-CI0)-aryl-O-(ClC6)-alkyl-.
[0135] In some embodiments, the présent invention provides a compound of formula II:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independently selected from: halogen (e.g.. Cl, F), -H, -(Cl-C6)a!kyl, -OH,
-O((Cl-C6)alkyl) (e.g., -OMe), -NÜ2, -CN, -CFj, and -OCF3, wherein R1 is independently substituted with 0-5 R’;
R2 is selected from:
-H, -C(O)NR2, and (C6-C IO)-aryl- (e.g., phenyl);
R3 is selected from:
-(Cl-C6)alkyl, -(C2-C6)alkenyl (e.g., -CH=CH2), -C=CH, -CN, halogen (e.g., Br), -SO2((C6-C10)-aryl), -SO2((C|.C6)alky1), -C(O)N((C1-C6)alkyl)2, -C(O)NH2, -C(O)O((Cl-C6)alkyl), -C(O)((Cl-C6)alkyl), -(C6-C10)aryl, 5- to 10- membered heteroaryl (e.g., 5-membered heteroaryl such as an optionally substituted
and 5- to 10- membered heterocyclyl (e.g., 5-membered heterocyclyl such as an optionally substituted
), wherein R3 is independently substituted with 0-5
R4 and R3 are each -H, halogen and -(Cl-C6)alkyl;
R6 îs selected from -H and -(C 1 -C6)alkyI;
each R is independently selected from:
H-, (Cl-C12)-al iphatic-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloa!kenyl-, [(C3-C10)-cycloalkyl]-(C 1 -C 12)-al i phat ic-, [(C3-C10)-cyc!oalkenyl]-(C I -C 12)-ali phatic-, [(C3-C10)-cycloalkyl]-O-(C 1 -C 12>aliphatic-, [(C3-C10)-cyc!oalkenyl]-O-(Cl-C12)-aliphatic-, (C6-C10)-aryl-, (C6-C10)-aryl-(C 1 -C 12)al iphatic-, (C6-C10)-aryl-O-(C 1 -C 12)a! iphatic-, (C6-CI0)-aryl-N(R”)-(Cl-C12)a!iphatic-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)al iphatic-, (3- to 10- membered heterocyclyl)-O-(CI-C12)aliphatîc-, (3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)al iphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl)-(Cl-CI2)-al iphatic-, (5- to 10- membered heteroaryl)-0-(Cl-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R’*)-(Cl-C12)-al iphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R îs independently substituted with 0-5 R*;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatîc ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring îs optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C310 C10)cycloalkyl, or a 3-to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CHiOR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NOi, -NCS, -CN, -CFj, -OCFj and -N(R”)2;
wherein each occurrence of R” ïs independently selected from H, -<Cl-C6)-alkyl, (C315 C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10>aryl-, (5-to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10>aryl-O-(ClC6)-alkyl-.
[0136[ ln some embodiments, the présent invention provides a compound of formula II:
Π, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independently selected from: halogen (e.g., Cl, F) and -O((Cl-C6)alkyl) (e.g., 18506
OMe), wherein R1 is independently substituted with 0-5 R’;
R is selected from:
-H, -C(O)NR2, and (C6-Cl0)-aryl- (e.g., phenyl); R3 is selected from:
halogen (e.g., Br), 5- to 10- membered heteroaryl (e.g., 5-membered heteroaryl such as an optionally substituted
), and 5- to 10- membered heterocyclyl (e.g., 5-
membered heterocyclyl such as an optionally substituted ' ), wherein R3 is independently substituted with 0-5 R’; R4 and R5 are each -H; R*is-H;
each R is independently selected from:
H-, (CI-C12)-al iphatic-, (C3-C10)-cycloa!kyl-, (C3-C10}-cycloalkenyl-, [(C3-C10)-cycloalkyl]-(Cl-C 12)-aliphatic-, [(C3-C10)-cycloalkenyl]-(C l-C 12)-al iphatic-, [(C3-C10)-cycloalky 1]-O-(C 1 -C 12)-al iph atic-, [(C3-C10)-cycloalkenyl]-O-(C l-C 12)-alîphatic-, (C6-C10)-aryl-, (C6-C10)-aryl-(Cl-C12)al iphatic-, (C6-C10>aryl-O-(C 1 -C 12)al i phatic-, (C6-C10)-aryl-N(R”)-(C l-C 12)al iphatic-,
3- to 10- membered heterocyclyl-, (3- to 10-membered heterocyclyl)-(Cl-CI2)aliphatic-, (3- to 10- membered heterocyclyl)-0-(CI-C12)aliphatic-, (3- to 10- membered heterocyclyl)-N(R*’)-(Cl-Cl2)aliphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroaryl)-(Cl-C12)-aliphatic-, (5- to 10- membered heteroaryl)-0-(Cl-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aliphatic-;
wherein said heterocyclyl has 1*4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1*4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R is independently substituted with 0-5 R’;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring îs optionally fused to a (Cô-ClO)aryl, 5- to 10- membered heteroaryl, (C3C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R”)2;
wherein each occurrence of R” is independently selected from H,-(Cl-C6)-alkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C1 OJ-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-a!kyl-, and (C6-C10)-aryl-O-(ClC6)-alkyl-.
[0137] In some embodiments, the présent invention provides a compound of formula II:
Π, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R* is independently selected from: halogen (e.g.. Cl, F), -H, -(C I -C6)alkyl, -OH, -O((Cl-C6)alkyl) (e.g., -OMe), -NOj, -CN, -CF3,and -OCFj, wherein R’ is independently substituted with 0-5 R’;
R2 is selected from:
-H, <C1-C6)alkyl, -OH, -O((CI-C6)aIkyI), -C(O)O((Cl-C6)aIkyI), -C(O)NR3, (C6C10)-aiyl(C6-C 10)-aryl-(C 1-C12)al iphatic-, (C6-C10)-aryl-O-(C 1 -C 12)al îphat ic-, (C6-C1O)-ary l-N(R’ ’)-(C I -C 12)al iphatic-, (5-to 10- membered heteroaryl)-(CI-C12)aliphatic-, (5- to 10- membered heteroaryl)-0-(Cl-C12)al iphatic-, (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-0-(Cl-CI2)aliphatic-, and (3- to 10- membered heterocyclyl)-N(R”)-(CI-C12)aliphatic-, wherein R2 is independently substituted with 0-5 R’; R3 is selected from:
-(C2-C6)alkenyl (e.g., -CH=CH3) and 5- to 10- membered heterocyclyl (e.g., 5membered heterocyclyl such as an optionally substituted
independently substituted with 0-5 R’;
R4 and R3 are each independently selected from -H, halogen and -(C 1 -C6)alkyt;
R6 is selected from-H and -(Cl-C6)alkyl;
each R is independently selected from:
(CI-C12)-a!iphatic-, (C3-C10)-cycloalkyl-, (C3-C10)-cycloalkeny 1-, [(C3-C10)-cycloalky I ]-(C I -C 12)- al i phatic-, [(C3-C10)-cycloalkenyl]-(C I -C 12)-al i phatic-, [(C 3-C10)-cycloalkyI]-O-(C I -C 12)-aI i phatic-, [(C 3-C10)-cycloalkenyl]-O-(C 1 -C 12)-al i phatic-, (C6-CI0)-ary!-, (C6-C10)-aryl-(C I -C 12)al iphat ic-, (C6-C10)-aryl-O-(C 1 -C 12)al iphatic-, (C6-C10)-aryl-N(R”)-(C 1 -C 12)al i phatî c-,
3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyc!yl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(CÎ-C12)aliphatic-, (3- to 10- membered heterocyc!yl)-N(R”)-(Cl-C12)a!iphatic-,
5- to 10- membered heteroaryl-, (5- to 10- membered heteroary!)-(Cl-CI2)-a!iphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)-alÎphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-a!iphatic-;
wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S;
wherein each occurrence of R is independently substituted with 0-5 R*;
or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or nonaromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3C10)cyc!oa!kyl, or a 3- to 10- membered heterocyclyl;
wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2N(R”)2, -C(O)N(R”)2, -C(O)OR”, -NO2i -NCS, -CN, -CF3, -OCFj and -N(R”)2;
wherein each occurrence of R” is independently selected from H,-(Cl-C6)-alkyl, (C3C6)-cycloa!kyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-a!kyl-, and (C6-C10)-aryl-O-(ClC6)-alkyk [0138] ln some embodiments, the présent invention provides a compound of formula 11:
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independently selected from: halogen (e.g., Cl, F) and -O((CI-C6)alkyl) (e.g., OMe), wherein R* is independently substituted with 0-5 R*;
R2 is selected from:
-H, -(Cl-C6)alkyl, (C6-C10)-aiyl- (e.g., phenyl), and (C6-C10)-aryl-(C 1-C12)aliphatic-, wherein R2 is independently substituted with 0-5 R’;
R3 is selected from:
-(C2-C6)alkenyl (e.g., -CH=CH2) and 5- to 10- membered heterocyclyl (e.g., 5membered heterocyclyl such as an optionally substituted
independently substituted with 0-5 R’; R4 and Rs are each -H;
Rsis-H;
wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2N(R”)2, -C(O)N(R”)2, -C(O)OR”, -NOj, -NCS, -CN, -CF3, -OCFj and -N(R”)2;
wherein each occurrence of R” is independently selected from H,-(Cl-C6)-alkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6· C10)-aiyl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)18506 alkyl-, (5- to 10- membered heteroaryl)-0-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(ClC6)-alkyl-.
[0139] In some embodiments, the présent invention provides a compound of formula II:
II, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independently selected from: halogen, -H, -(Cl-C6)alkyl, -OH, -O((C1C6)alkyl), -NO2, -CN, -CF3, and -OCF3, wherein said alkyl is independently substituted with 0-5 R’;
R2 is selected from: -(Cl-C6)alkyl, -OH, -O((CI-C6)alkyl), -C(O)O((Cl-C6)alkyl), (C6-C10)-aryl-(Cl-C12)a!iphatic-, (C6-C10)-aryl-O-(Cl-C12)aliphatîc-, (C6-C10)-aryl-(Cl-C12)aliphatic-O-, (3- to 10- membered heterocyclyl)-(C ΙΟΙ 2)aliphatic-, (5- to 10- membered heteroaryl)-(CI-C12)-aliphatic-, (5- to 10membered heteroaryl)-0-(Cl-C12)-alîphatic-,and (5- to 10- membered heteroaryl)(Cl-CI2)-aliphatÎc-O-, wherein said alkyl, aryl or heteroaryl is independently substituted with 0-5 R’;
R3 is selected from: -(Cl-C6)alkyl, -SO2((Cl-C6)a!kyl), -C(O)N((C1-C6)alkyl)2, and -C(O)O((Cl-C6)alkyl), wherein said alkyl is independently substituted with 0-5 R*;
R' is as defined herein;
R4 and R5 are each independently selected from -H, halogen and -(Cl-C6)alkyl; and
R6 is selected from -H and -(Cl-C6)alkyl.
[0140] In some of the embodiments of a compound of formula II, m is 0, 1 or 2; when m is 1 or 2, at least one occurrence of R1 is halogen or -O((C 1 -C6)alky 1) (such as
-F and -OMe);
R2 is selected from: -(Cl-C6)alkyl (e.g., -Me), (C6-CI0)-aryl-(Cl-CI2)aliphatic- (e.g., -CH2Ph), (C6-C10)-aryl-O-(CI-C12)aliphatic- (e.g., -CH2OPh) and (3- to 10membered heterocyclyl)-(Cl-CI2)aliphatîc- (e.g., -CH2-pyrrolidine and-CH2morpholine), wherein said aryl (e.g., -Ph) or heterocyclyl (e.g., pyrrolidine or morpholine) ts independently substituted with 0-5 R’ independently selected from -F, -Me, and -OMe, and wherein said alkyl (e.g., -Me) is independently substituted with 0-3 R* selected from -N(Eth and-N(Me)(CH2Ph).
R3 is -C(O)O((Cl-C6)alkyl) (e.g., -COOEt);
R4 and R3 are both -H; and
R6 is -H.
[0141] In some embodiments, the présent invention provides a compound of formula II:
II, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m îs 0-3;
each R1 is independently selected from: halogen, -H, -(Cl-C6)alkyl, -OH, -O((Cl-C6)alkyl), -NO2, -CN, -CFj, and -OCF3, wherein R* is independently substituted with 0-5 R’;
R2 is selected from:
-(C l-C6)alkyl, -OH, -O((Cl-C6)alkyl), -C(O)O((Cl-C6)alkyl), (C6-C10)-aryl-(C 1 -C 12)a I iphat i c-, (C6-C10)-aryl-O-(Cl -Cl 2)al iphatic-, (C6-C10)-aryl-N(R’*)-(C I -C I2)aliphatic-, (5- to 10- membered heteroaryl)-(C!-C12)aliphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)aliphatic-, (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-(C!-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-C!2)aliphatic-, and (3- to 10- membered heterocyclyl)-N(R”)-(C!-C12)alïphatÎc-, wherein R2 is independently substituted with 0-5 R*;
R3 is selected from:
-(C!-C6)alkyl, -C=C, -CN, halogen, -SO2((C6-C!0)-aryl), -SO2((Cl-C6)alkyl), -C(O)N((C1-C6)alkyl)2, -C(O)NH2, -C(O)O((C!-C6)alkyl), -C(OX(Cl-C6)alkyl), (C6-C10)aryl, and 5- to 10- membered heteroaryl, wherein R3 is independently substituted with 0-5 R’;
R4 and Rs are each independently selected from -H, halogen and -(Cl-C6)alkyl;
R6 is selected from -H and -(Cl-C6)alkyl; and
R’ and R” are as defined herein.
[0142] In some embodiments of a compound of formula II:
m is 0, ! or 2;
when m is l or 2, at least one occurrence of R1 is halogen or-O((Cl-C6)alkyl);
R2 is selected from:
-(Cl-C6)alkyl, (C6-C10)-ary!-(Cl-C12)aliphatîc-, (C6-C!0)aryl-O-(ClC12)alîphatic-, (5- to 10- membered heteroaryl)-(C!-C12)aliphatic-, and (3- to 10membered heterocyclyl)-(Cl-C!2)aliphatic-, wherein R2 is independently substituted with 0-3 R’;
R3 is halogen, -CN, -C=C, -C(O)NH2, -(Cl-Cô)alkyl, -C(O)((Cl-C6)alkyl), -C(O)O((C1C6)alkyl),-SO2(Ph(Me)), wherein R3 is independently substituted with 0-3 R’, and wherein R9 is selected from -H, -Me, -Et, -CF3, isopropyl, -OMe, -ferf-butyl, and cyclopropyl;
R4 and Rs are both -H;
Rs is -H; and
R* is as defined herein.
[0143] In some embodiments of a compound of formula II, R3 is:
, wherein R9 is selected from -H, -Me, -Et,
-CF3, isopropyl, -OMe, and -tert-butyl.
[0144] In some embodiments, the présent invention provides a compound of formula
III:
III, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0,1, or 2, and when m is 1 or 2, at least one occurrence of R1 is -O((Cl-C6)alkyl) (such as -OMe);
R2 is selected from: -(Cl-C6)alkyl (e.g., -Me) and (C6-C10)-aryl-(Cl-C12)aliphatic(e.g., -CH2Ph);
R3 is -C(O)O((C 1-C6)alkyl) (e.g., -COOEt);
R4 and Rs are both -H; and
R6 is -H.
[0145] In another aspect, the présent invention provides a compound of formula IV:
R5
or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3 (e.g., m is 1);
each R1 is independently selected from: -Cl, -F, -OMe, and -C^CH;
R2 Is -(CH2)nORe or -(CH2)nO(CH2)nR8, wherein each occurrence of R8 îs independently (Cl-C6)alkyl or (C6-C10)-aryl (e.g., phenyl), and wherein R2 is independently substituted with 0-5 R’;
R3 is selected from: -CN, -C^CH, -C^C-(Cl-C6)alkyl, -OC-phenyl, and .wherein R3 is substituted with 0-5 R’;
each occurrence of R4 and R3 îs independently -H or -(Cl-C6)alkyl;
each R6 is independently -H or -(C 1 -C6)alkyl ;
wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R)2;
wherein each occurrence ofR” is independently selected from H,-<CI-C6)-alkyl, —(Cl C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)alkyl-, (C6-C10)-aryl-(Cl-C6)-alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)alkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-5 substituents selected from: halogen, -R, -OR, oxo, -CH2OR°, -CH2N(Ro)2, -C(O)N(R°)2, -C(O)OR°, -N02, -NCS, -CN, -cf3, -OCF3 and -N(R°)2, wherein each occurrence of R° is independently selected from: -<Cl-C6)-a!iphatic, (C3-C6)-cycloa!kyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-C10)-ary!-.
In some of the above embodiments, R3 is selected from:
wherein each occurrence of R” is independently selected from -(C 1 -C6)-a!kyI (e.g., linear or branched), -C=CH, phenyl, thiophene, (5- to 10- membered heteroaryl)-(ClC6)-alkyl-, and (C6-CI0)-aryl-(Cl-C6)-alkyl-, wherein each R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR°, CH2N(R°)2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R°)2, wherein each occurrence of R° is independently selected from: -(C I -C6)-al iphatic, (C3-C6)-cycloa!kyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, and (C6-CI0)-aryl-.
[0146] In another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independentiy selected from: halogen (e.g., Cl), -H, -(C I-C6)alkyl, -C^CH, OH, -O((Cl-C6)alkyl) (e.g., OMe), -NO2, -CN, -CF3, and -OCF3, wherein R1 is independentiy substituted with 0-5 R*;
R2 is selected from -OR8, -SR8, -(CH2)nOR8 (e.g., -CH2OMe, -CHjOEt, -CHjOisopropyl, -CHiOpyridyl), -(CH2)nO(CH2)nR8, -(CH2)PR8 and -(CH2)nN(R”)R10, wherein n is an integer selected from 0-4; p is an integer selected from 2-4; each R8 îs independentiy (C 1 -C6)alkyl, -(C3-CI0)-cycloa!kyl, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independentiy substituted with 0-5 R’;
each R10 is independentiy -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-ary 1, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independentiy substituted with 0-5 R’; and wherein R2 is independentiy substituted with 0-5 R’;
RJ is selected from:
-H, -CN, halogen (e.g., Br), -(C l-C6)alkyl, -C^CH, -SO2((Cl-C6)alkyl), -C(O)N((C1C6)alkyl)2, ), -C(O)NH((C1-C6)a!îphatic)2 (e.g., -C(O)NH((C2-C6)alkynylh), (C6C10>aiyl-(Cl-C12)al iphatic-, -C(O)((Cl-C6)aIkyl), -C(O)O((Cl-C6)alkyl), 5- or 6membered heterocyclyl- (e.g., optîonally substituted
or optîonally substituted
), and 5- or 6-membered heteroaryl (e.g., optîonally substituted
, optîonally substituted , , wherein
R9 is selected from -Me, -Et, isopropyl, -CF3, -OMe, -OEt, -O-isopropyl, -CH2NMe2, and cyclopropyl; and wherein RJ is independentiy substituted with 0-5 R’;
R4 and Rs are each independentiy selected from -H, halogen and -(C 1-C6)alky I; R6 is selected from -H and -(C I-C6)alkyI;
wherein each occurrence of R’ is independentiy selected from halogen, -R, -OR”, oxo, -CH2OR”, -CH2N(R”)2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS. -CN, -cf3, -ocf3 and -N(R”)2;
wherein each occurrence of R” is independentiy selected from H, —(C 1 -C6)-alkyl, (C3
C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(CI-C6)alkyl-, (5- to 10- membered heteroaryl)-0-(CI-C6)-alkyl-, and (C6-C10)-aryl-O-(CIC6)-alkyl-.
[0147] In another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 1;
R1 is -OCH, optionally substituted with a R’;
R2 is selected from -OR8, -SR8, -(CH2)nOR8 (e.g., -CH2OMe, -CH2OEt, -CH2Oîsopropyl, -CH2Opyridyl), -(CH2)nO(CH2)nR8, -(CH2)PR8 and -(CH2)nN(R”)Rt0, wherein n is an integer selected from 0-4; p is an integer selected from 2-4; each R8 is independently (Cl-C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R*; each Rt0 Is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 îs independently substituted with 0-5 R’; and wherein R2 is independently substituted with 0-5 R’;
R3 is selected from:
-H, -CN, halogen (e.g., Br), -(Cl-C6)alkyl, -C=CH, -SO2((Cl-C6)alkyl), -C(O)N((C1C6)alkyl)2, ), -C(O)NH((C1-C6)aliphatic)2 (e.g., -C(O)NH((CI-C6)alkynyl)2), (C6C10)-aryl-(Cl-CI2)aliphatic-, -C(O)((Cl-C6)alkyl), -C(O)O((Cl-C6)alkyl), 5-or 618506
membered heterocyclyl- (e.g., optionally substituted
or optionally substituted
), and 5- or 6-membered heteroaryl (e.g., optionally substituted
, optionally substituted
wherein R9 is selected from -Me, -Et, isopropyl, -CF3, -OMe, -OEt, -O-isopropyl, -CH2NMe2, and cyclopropyl; and wherein R3 is independently substituted with 0-5 R’;
R4 and R3 are each independently selected from -H, halogen and -(C 1 -C6)alkyl;
R6 is selected from -H and -(C I -C6)alkyl;
wherein each occurrence of R’ îs independently selected from halogen, -R”, -OR**, oxo,
-CH2OR*-CH2NR”2, -C(O)N(R*% -C(O)OR’*, -NO2, -NCS, -CN, -CF3, -OCF3 and -N(R’*)2;
wherein each occurrence of R” is independently selected from H, -(CI-C6)-alkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6· Cl0)-aryl-, (5- to 10- membered heteroaryl)-(CI-C6)-alkyl-, (C6-CI0)-aryl-(Cl-C6)15 alkyl-, (5- to 10-membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-CI0)-aryl-O-(ClC6)-alkyl-.
[0148] In another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is I;
each R* is -C^CH, optionally substituted with a R’;
R2 is -(CH2)nOR8 (e.g., -CH2OMe, -CH2OEt, -CH2Oisopropyl, -CH2Opyridyl),; and wherein R2 is independently substituted with 0-5 R’;
R3 is selected from:
5- or 6-membered heterocyclyl- (e.g., optionally substituted
or optionally substituted
), and 5- or 6-membered heteroaryl (e.g., optionally substituted
, or optionally substituted
); and wherein R3 is independently substituted with 0-5 R’; R4 and R3 are each -H; R6 is -H; and wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO:, -NCS, -CN, -CFj, -OCFj and -N(R”)2;
wherein each occurrence of R” is independently selected from H, -<Cl-C6)-alkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C620 CI0)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(ClC6)-alkyl-, [0149] In another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
when m îs 1 or 2, at least one occurrence of R1 is -halogen or -O((CI-C6)alkyl);
each R* is independently selected from: halogen (e.g., Cl), -H, -(Cl-Cô)alkyl, -C^CH, OH, -O((C I -C6)alkyl) (e.g., OMe), -NO2, -CN, -CF3, and -OCF3, wherein R1 is independently substituted wîth 0-5 R';
R1 is selected from -OR8, -SR8, -(CH2)nOR8 (e.g., -CH2OMe, -CH2OEt, -CH2Oisopropyl, -CH2Opyridyl), -(CH2)nO(CH2)nR8, -(CH2)PR8 and -(CH2)nN(R”)R10, wherein n is an integer selected from 0-4; p is an integer selected from 2-4; each R8 îs independently (C 1 -C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’; each R10 îs independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’; and wherein R2 is independently substituted with 0-5 R’;
R3 is selected from:
-C^CH, -C(O)NH((CI-C6)aliphatic)2 (e.g., -C(O)NH((C1-C6)alkynyl)2), (C6-C10)aryl-(Cl-C12)aliphatic-, 5- or 6-membered heterocyclyl- (e.g., optionally substituted
or optionally substituted
), optionally substituted
R4 and R3 are each independently selected from -H, halogen and -(Cl-C6)alkyl;
R6 is selected from -H and -(Cl-C6)alkyl; and wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”j, -C(O)N(R”)2, -C(O)OR”, -N02, -NCS, -CN, -CFj, -OCF3 and -N(R”)2;
wherein each occurrence of R” is independently selected from H,-(Cl-C6)-alkyl, (C3C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C610 C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-( (C6-C10)-ary!-(Cl-C6)alkyl-, (5- to 10- membered heteroary!)-0-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(ClC6)-alkyl-.
[0150] In another aspect, the present invention provides a compound of formula IV:
R9 or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
each R1 is independently selected from: halogen (e.g., Cl), -C^CH, and -O((Cl-C6)alkyl) (e.g., OMe), wherein R1 is independently substituted with 0-5 R’;
R2 is -(CH2)nOR8 (e.g., -CH2OMe, -CH2OEt, -CH2O-isopropyl, -CH2O-pyridyl), wherein n is an integer selected from 0-4; R8 is -(Cl-C6)alkyl, -(C3-C10)-cycloalkyl, (C6C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’; and wherein R2 îs independently substituted with 0-5 R’;
R3 is selected from:
-ChCH, -C(O)NH((C1-C6)al iphatic): (e.g., -C(O)NH((C1-C6)alkyny!):)), (C6-C10)ary!-(Cl-C 12)a! iphatic-, 5- or 6-membered heterocyclyl- (e.g., optionally substituted
or optionally substituted
), optionally substituted
optionally substituted
; and wherein R3 is independently substituted with 0-5
R’;
R4 and R3 are each -H;
R6 îs -H or -(Cl-C6)a!kyl; and wherein each occurrence of R* is independently selected from halogen, -R”, -OR”, oxo, -CHjOR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO:, -NCS, -CN, -CF3, -OCF3 and -N(R”):;
wherein each occurrence of R” îs independently selected from H, -(Cl-C6)-aIkyI, (C3C6)-cycIoalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6C10)-atyl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryL(Cl-C6)alkyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(ClC6)-alkyl-.
[0151J ln another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or
combination thereof, wherein:
m is 0, l, or 2, and when m îs l or 2, at least oneoccurrence ofR1 is -O((Cl-C6)alkyl) (such as -OMe);
R2 >s OR8, wherein R8 is (Cô-ClO)-aryl (such as phenyl), substituted with 0-3 halogen 5 (such as -F);
R3 is -C(O)O((CI-C6)alkyl) (e.g., -COOEt);
R4 and R8 are both -H; and
R6 is -H.
[0152] In another aspect, the présent invention provides a compound of formula IV:
IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
m is 0-3;
when m >s I or 2, at least one occurrence of R1 îs -halogen or -O((Cl-C6)alkyl); each R1 îs independently selected from: halogen, -H, -(Cl-Cô)alkyl, -OH, -O((Cl-C6)alkyl), -NOj, -CN, -CFj, and -OCFj, wherein R* is independently substituted with 0-5 R’;
R2 is selected from -OR8, -SR8, -(CH2)nOR8, -(CH2)nO(CH2)nR8, -(CH2)PR8 and
-(CH2)nN(R”)R10, wherein n Îs an integer selected from 0-4; p is an integer selected from 2-4; each R8 is independently -(Cl-C6)alkyl, -(C3-C IO)-cycloalkyl, (C6-C10)aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’; each R10 is independently -(C3-C10)cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-CI0)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’;
and wherein R2 is independently substituted with 0-5 R*; R3 is selected from:
-H, -CN, halogen, -(Cl-C6)alkyl, -SO2((CI-C6)alkyl), -C(O)N((C1-C6)alkyl)2, -C(O)((C 1 -C6)alky l), -C(O)O((C1 -C6)alky I),
wherein R9 is selected from -Me, -Et, isopropyl, -CF3, -OMe, -OEt, -O-isopropyl, -CH2NMe2, and cyclopropyl; and wherein R3 is independently substituted with 0-5 R’;
R4 and R5 are each independently selected from -H, halogen and -(Cl-C6)alkyl;
R6 is selected from-H and -(Cl-C6)alkyl; and
R’ and R’’ are as defined herein.
[0153] In some embodiments of a compound of formula IV:
m is 0,1, or 2;
R2 is -OR8, -(CH2)nORs, -(CH2)nO(CH2)nR8, wherein n îs 1, and wherein R8 is -(ClC6)alkyl, (C6-C10)-aryl or 5- to 10-membered heteroaryl, wherein R8 is independently substituted with 0-3 R*;
R3 is halogen, -H, -CN, -(Cl-C6)alkyl, -C(O)((Cl-C6)alkyl), -C(O)O((Cl-C6)alkyl),
, wherein said alkyl is independently substituted with 0-3 R’; R9 is selected from -Me, -Et, isopropyl, and -CF3;
R4 and RJ are both -H;
R6 is -H; and
R’ is as defined herein.
[0154] Examples of particular compounds of the présent application include:
Compound Structure
Compound Structure
1 Λ-COjEt ..Al N
2 fA^CO2Et Al n'N
3 ΓΝ/)-0Ο2ΕΙ cQ f!|. 7ΌΗ3 n'N
4 f^V-COjEt „Alp
5 r >-COzEt cdp 1 vv * N % / N
6 r >-co2Et AÎp F y N'V
Compound Structure
7 rt-CO2Et NV
8 Γ à-CQzEt O J o
9 Λ-002ΕΙ xf Nv% ό
10 Λ-οο,ει XX j Η,ΟΟ-^Χ-/ Ν-'./Λ nx 0 F
11 Λ-CO2Et .xi 0 och3
Compound Structure
12 Λ-COjEt O-CH3 H3C t
44 r >-CO2Et
45 £VCOiEt « n 5 à> F F y__/^ NV
46 Λ-οο,ε. jCu ô NV
47 a fô Ny_/
48 Λ-οο,ει il)o MeO^^N A/N' N'V
Compound Structure
49 -N r>-co2Et Xk?
50 N-/
51 Γ />-COjEt ΓΎ ) O MeO'^^N-Y Q-7
52 r>-co2Ët ..Æfp-
53 -A OÎ O- MeO^^N-^ 7=/ Q-^
Compound Structure
54 \ /V-COjEt JCQJQ • υ J Vi cl
55 r y-cojEt MeoX^:p-0
56 Λ-°θ2ΕΙ MeoÀArV”0F n'N
101 r >-co2Et Ν'/ ΓΎ j O
102 V />-CO2Et
103 fA—COjEt MeOxAxjÿiks^N 1/~Ά N o—
Compound Structure
104 f>N>-CO2Et ô
105 >-CO2Et ZVN \ Jl /Z~R N λν \_f/~ OMa
106 r >-co2Et MeO-^^V-N V-Br
107 Λ-οο2ει °'ÿ n'N
108 (^/V-COjEt jCO O MeCr'''Î^SN-'\ /=\
109 ΛNV\_. o
Compound Structure
110 -N /O'N X
111 Λ Γ/>-οο2η M o N=/
112 -<Λ
113 n'N
114 N'N
115 Ô-CN N- ' N'N
Compound Structure
116 Λ-cn Λί. Ν', Ν
117 ου αΥ NÀI~
118 ,^Ν /°'Ν Μβο^Γ^°Ό ν
119 y- ^^,Ν-y ο ν
120 αΗ ^.Ν-Υΐ) «Ή
121 Ν- ' Ν
Compound Structure
122 Q-cn H'N
123 Μ-ρΟ N'N
124 Λ-/ ιΥν n'N
125 tA_/ N ** ' 'N
126 Λ /°'N 'N
127 rW'î < 'N
Compound Structure
134 fy-'0'7 fi-
135 Xi N- '— 'N
136 il·8' 'fi
137 F^^Sj-N OPh
138 -N /O'N F'iX°Ph
139 ô-O ΑΓ Λ— NV
104
Compound Structure
146 N CCT N
147 AA F
148 α_λ XN^ v
149 Afv NV~
150 λ/Α'A NV
151 N' / N'N
Compound Structure
158 FMrv)ph ”'N
159 (A-Z? f zyN'\ * iiFj ρΧ^ιΓΝ\ zOph ^-n
160 'N
161 κΓ Ν' ' W'N
162 xyVA MeO^^V-N oph N ?—Z n'N
163
107
Compound Structure
164 Metrvÿ!i\-'N OMe N ' W'N
165 Æ/? N ' M'N
166 J^Y-cn MeO^^^V-N N * ”'N
167 -N P'N ιχΓ Av n'N
16S rNy~/'? Χ1Ά
169 rW'î N'y MeO^^Aj-N OMe N v—' 'N
Compound Structure
170 -N P'N n\ O
171 ίχΎΝ~ν^Ν*^^ ΜβΟ'Λ^>Γ-Ν oph J l·-' 'N
172 a/? -pyQ,
173 A-H AA MeO^^X-N OEt I/'
174 xxTa AA
175 ίΑ-/'? χχΡ'Ζζ “ΊΑ CI
109
Compound Structure
176 Γ v n'N N
177 Δ-/1 Γ 'Y-7 X? N C?
178 ΑΛ MeO^XiÿiiX--N /-VO
179 F
180 nX NiN
181 rW'? £ïXN
Compound Structure
182 χΥ'Ρ n'N
183 xcA'
184 ÎKrVy· ΜίΟ^Ά-Ν Ο-^Γ Λ-A Q
185 À, ___ΆγΖ <3 2^Z 0 T
186 fA_A° .oA'V OMe SA
187 (A-Z? ,A^., n'N
Compound Structure
188 F iKxy
189 rA-/''?
190 rA_/·? And
191 O-cn ‘Aro,
192 k/>-CN
193 rrT
112
Compound Structure
194 μ.<ΛΑ-<__o
195 rW'1? xx.Xo'
196 λ-cn “Aw
197 îQcn c|J°^ho
198 P'N jX^ n'N
199 jCO
113
Compound Structure
200 N F
201 (A_Z? ΗβΟ^γΝ 0 1 XT
202 if^B' Cl 0-7¾
203 Λ-Ζ? _.n0 W
204 Cl 0-7¾
205 Ao
115
Compound Structure
217 o ζγ*^Χγζ X 00 o ” /
218 jΠ5 d N o
219 m d MeCT^XX f=/ N-vH N o
220 Γχ j o MeO'^^N'X >=/
221 ZW Z xx Td
222 jA-CN «ΧΧ-Αο-' N''N
223 Λ-cn jCQ k MeO^^N-^ P-Z \ w w n-n
117
Compound Structure
224 ΙΎ ) Me0
225 γ'χ-ον XX ) /=<Br n-'n^
226 Λ-cn “Ύ>-Ο
227 Λ-cn ciJCQ oX
228 Λ-cn a^XyX>r n''N
229 Λ-βγ AJ Q n-n
230 Α-Λ? W)^ x F >-/ NvV O F
119
Compound Structure
237 XX W'N
238 Λ-ύ n'N
239 exx N-7 ci^/TV° N'N
240 Xî^ Q-x N O—
241 0. A «x x
242 b /X z^z—-Ayz >> cAz Έ
Compound Structure
243 Α/°η N'N
244 MA ΓτΦ^ c|X'/TX·0'N'N
245 qXjP Ίθ^ m'N
246 Λ// jCC? N'N
247 xc NV\ N /NH
248 Φ* «•°zJ^Çq nÆ o
121
Compound Structure
249 Δ-ΐ)
250 £/-cn ΜβΟ-^θ^Ν-Ζ 0
251 A-cn ^ajp N' ' ' N'N
252 îQh-GN MeO^^N-/_ZN-/ N'N
253 O-CN V/ _Æfo -V
254 P'N C|A^r ζ>-^Οχ/ n'N
Compound Structure
255 P'N 'N
256 λ— Cl M'N
257 JY P-N ΜβΟ^^ν-Ά N- /
258 P'N NV
259 ci^VÿA. n q Γ 'N
260 (Α-Λι £0 'V ”'N
123
Compound Structure
261 Λ-s n'N
262 rVcN jCq m,° £p-o
263 Α-Λ1 jYV'O
264 nrTNO
265 q-UX)
266
267 ν
124
Compound Structure
268
270
271 AA3
272 λ* /°~N /=x vO
273 jCŒ ΜβΟ^^Ν-Χ /=ξ ΛβΝ>Λ_/
274 'N
275 Cl
276 ΑΓΑ
125
Compound Structure
277 f oA Q-
278 N'N
279 fy-cOiEt xx5 MeCr^^N-C /=\ W Cl'
280 N-n
281 P'N °θγνγ N'N 1
282 N'N
283 a ù ,00 n-n
126
Compound Structure
284
285 n*N
286
287 λχ. JCO N α οΛ
288 Λ-=
289 ΜβΟ'^^Ν-ζ *?
290 w
127
Compound Structure
291 MeO^^N^ V
292 V
293 b
294 py jcA cA^y o, n'N
295 XX > «yr^A n'N
296 Λ-tf IXJ n'N
128
Compound Structure
297 AA
298 A MeP^^X q..
299 AHAO NA
300 Cl FA-/0' NV
301 (A/0! XX > O XAr^0' 'N
302 Λ_/°1 χΓΑ 'N
303 P'N /A α>ΧίΐΓΝ\ /°^CF3 N ' 'N
129
Compound Structure
304 rfSΌ 'N
305 Ύ* —
306 ίΥΖ αΑργ
307 (X, “1 erp tf ο
308 A-COjEI aJ^V^CFs 'N
309 N N—, ÔX 'N
310 λ—b aJCr0r
Compound Structure
311
312 Cr ArV^ 'N
313 n-/ cr n'N
and their pharmaceutically suitable sait, hydrate, solvaté, polymorph, isomer or combination thereof.
[0155] The invention also includes various combinations of R, R and R as described above. These combinations can in tum be combined with any or ail of the values of the other variables described herein. For example, R1 can be-OR or halogen; ; R2 can be (CI-C4)-alkyl-, -OR8, -(CH2)nOR8, or -(CH2)nO(CH2)nR8; and optionally R3 is -C(O)OR, or -C(O)N(R)2. In another example, R1 is -OR or halogen; R2 is (C 1 -C4)-aIkyl-, -OR8, •(CH2)nOR8, or -(CH2)nO(CH2)nR8; and R3 is a 5- or 6-membered heteroaryl, such
. For each of above examples, compounds can hâve the spécifie values ofthe groups described herein.
[0156] Any embodiment described herein is also intended to represent unlabeled forms as well as isotopically labeled forms ofthe compounds, unless otherwise indicated.
Isotopically labeled compounds hâve structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or
* mass number. Examples of isotopes that can be incorporated into compounds ofthe invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2H, 3H, nC, ,3C, UC, 15N, l8F, 31P, 32P, S, 36CI, ,2îI, respectively. The invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3H, l3C, and l4C, are présent. Such isotopically labeled compounds are useful in metabolic studîes (preferably with C), réaction kînetic studîes (with, for example 2H or 3H), détection or imaging techniques, such as positron émission tomography (PET) or single-photon émission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an l8F or labeled compound may be particularly preferred for PET or SPECT studîes. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or tn the examples and préparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled 15 reagent.
[0157] Any ofthe individual embodiments recited herein may define formula I, II, III, or IV individually or be combined to produce a preferred embodiment ofthis invention.
General Synthetic Methodology [0158] The compounds of this invention may be prepared in general by methods known to those skilled in the art. Schemes I-10 below provide general synthetic routes for the préparation ofcompounds of formulae I-IV. Other équivalent schemes, which will be readily apparent to the ordinary skilled organic chemist, may altematively be used to synthesize various portions ofthe molécules as illustrated by the general schemes below.
Scheme 1. General synthesis of a compound of formula I wherein X, Y, Z, V and W form a 1,2,3-triazole ring, or a compound of formula II.
132
trfazoie cllck imldazole formation
1. nitro réduction
LO diazonium, haiide, etc.
2. cycllzatlon
Scheme 2. General synthesis of a compound of formula I or III, wherein X, Y, Z, V and
1. nitro réduction
-
2. cydlzation
imldazole formation -------►
Scheme 3. General synthesis of a compound of formula I, wherein X, Y, Z, V and W form a phenoxy-substituted 1,2,3-triazole ring, or a compound of formula 11.
133
LG diazonium, halide, etc.
1. hydrolysis
2. réduction
Scheme 4. General synthesis of compounds of formula I or II to allow for divergent functionalization on the triazolo-ring formed by X, Y, Z, V and W,
’N
Scheme 5. General synthesis of a compound of formula I wherein X, Y, Z, V and W form an aminomethyl-substituted 1,2,3-triazole ring, or a compound of formula IL
Scheme 6. General synthesis of a compound of formula l wherein X, Y, Z, V and W form an aralkyl-substituted or heteroaralkyl substituted 1,2,3-triazole ring, or a compound of formula IL
135
Scheme 7, General synthesis of a compound of formula I or IV, whereîn X, Y, Z, V and W form a substituted 1,2,4-triazole ring.
t® -OR®, -SR®, -tCHJnOR®, -<CH2)nO(CH2)^s, -tCH2)pRe,or -{CH2)nN(R“)Rio
Scheme 8. General synthesis of a compound of formula I wherein X, Y, Z, V, and W form a methyl-substituted 1,2,3-triazole ring, or a compound of formula II.
Imldazd· famuttûn
136
Scheme 9. General synthesis of a compound of formula I, wherein X, Y, Z, V, and W form a benzyl-substituted 1,2,3-triazole ring, or a compound of formula IL
Scheme 10. General synthesis of a compound of forumla I, H, or IV wherein X, Y, Z, V and W form a substituted triazole ring, such as a 1,2,3-triazole ring or a 1,2,4-triazole ring, and the upper imldazole is substituted with a 1,2,4-oxadizaole ring as illustrated in I0(a) and 10(b).
10(b)
Scheme 10a. General synthesis ofa compound where R3 is an optionally substituted dihydrooxazole or oxazînyl ring is illustrated in scheme 1 Oa.
137
R* = H or Et
Scheme 10b(a) and 10b(b). General synthesis of a compound where R3 îs an optîonally substituted oxazole or îsoxazole is illustrated in schemes 10b(a) and IOb(b).
(b)
PH n-BuLi, conc. H2SO<
Scheme 10c. General synthesis of a compound where R3 is an optîonally substituted alkynyl group îs illustrated in scheme 10c.
[0159] As would be recognized by skilled practitioners, compounds of formulae I-IV with variables other than those depicted above may be prepared by varying chemical reagents or the synthetic routes.
Pharmaceutical Compositions and Modes of Administration [0160] The présent invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formulae 1-IV, or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof.
[0161] The basic nitrogen-containing groups présent în the compounds of the invention may be quatemized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates, such as dimethyl, diethyl, dibutyl and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, 10 bromides and iodides, aralkyl halides, such as benzyl and phenethyl bromides and others.
Water or oil-solubie or dispersible products are thereby obtained.
[0162] It will be appreciated that compounds and agents used in the compositions of this invention preferably should readily penetrate the blood-brain barrier when peripherally administered. Compounds which cannot penetrate the blood-brain barrier, however, can still be effectively administered directly into the central nervous system, e.g., by an intraventricular or other neuro-compatible route.
[0163] In some embodiments of this invention, the a5-containing GABAa R positive allosteric modulator is formulated with a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers that may be used in these compositions include, but 20 are not limited to, ion exchangers, alumina, aluminum stéarate, lecithin, sérum proteins, such as human sérum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloïdal silica, magnésium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose. polyacrylates. waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. In other embodiments, no carrier is used.
For example, the a5-containing GABAa R agonist (e.g., a a5-containïng GABAa receptor positive allosteric modulator) can be administered alone or as a component of a pharmaceutical formulation (therapeutic composition). The a5-containing GABAa R
139 agonist (e.g., a a5-containing GABAa receptor positive allosteric modulator) may be formulated for administration in any convenient way for use in human medicine.
[0164] In some embodiments, the therapeutic methods of the invention include administering the composition of a compound or agent topically, systemically, or locaüy. For example, therapeutic compositions of compounds or agents ofthe invention may be formulated for administration by, for example, injection (e.g., intravenously, subcutaneously, or intramuscularly), inhalation or insufflation (either through the mouth or the nose) or oral, buccal, sublingual, transdermal, nasal, or parentera! administration. The compositions of compounds or agents described herein may be formulated as part of an implant or device, or formulated for slow or extended release. When administered parenterally, the therapeutic composition of compounds or agents for use in this invention is preferably in a pyrogen-free, physîologically acceptable form. Techniques and formulations generally may be found in Remîngton’s Pharmaceutical Sciences, Meade Publishing Co., Easton, PA.
[0165] In certain embodiments, pharmaceutical compositions suitable for parentera! administration may comprise the a5-containing GABAa R positive allosteric modulator in combination with one or more pharmaceutically acceptable stérile isotonie aqueous or non-aqueous solutions, dispersions, suspensions or émulsions, or stérile powders which may be reconstituted into stérile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutés which render the formulation isotonie with the blood of the intended récipient or suspending or thickening agents. Examples of suitable aqueous and non-aqueous carriers which may be employed in the pharmaceutical compositions ofthe invention include water, éthanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance ofthe required particle size in the case of dispersions, and by the use of surfactants.
[0166] A composition comprising a a5-containing GABAa R positive allosteric modulator may also contain adjuvants, such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prévention ofthe action ofmicroorganisms may be ensured
140 by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phénol sorbic acid, and the like. It may also be désirable to include isotonie agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and gelatin.
[0167] In certain embodiments of the invention, compositions comprising a a5containing GABAa R positive allosteric modulator can be administered orally, e.g., in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basîs, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-în-oil liquid émulsion, or as an élixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and the like, each containing a predetermined amount of the α5· containing GABAa R positive allosteric modulator as an active ingrédient.
[0168] In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules, and the like), one or more compositions comprising the a5-containing GABAa R positive allosteric modulator may be mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) bïnders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quatemary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stéarate, magnésium stéarate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
141 [0169] Liquid dosage forms for oral administration include pharmaceutically acceptable émulsions, microemulsions, solutions, suspensions, syrups, and élixirs. In addition to the a5-containing GABAa R positive allosteric modulator, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifîers, such as ethyl alcohol (éthanol), isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
[0170] Suspensions, in addition to the active compounds, may contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol, and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
[0171] As described herein, the compounds, agents, and compositions thereof may be administered for slow, controlled or extended release. The term “extended release” is widely recognized in the art of pharmaceutical sciences and is used herein to refer to a controlled release of an active compound or agent from a dosage form to an environment over (throughout or during) an extended period of time, e.g. greater than or equal to one hour. An extended release dosage form will release drug at substantially constant rate over an extended period of time or a substantially constant amount of drug will be released incrementally over an extended period of time. The term “extended release” used herein includes the terms “controlled release,” “prolonged release,” “sustaîned release,” “delayed release,” or “slow release” as these terms are used în the pharmaceutical sciences. In some embodiments, the extended release dosage is administered in the form of a patch or a pump.
[0172] A person of ordinary skill in the art, such as a physician, is readily able to détermine the requîred amount ofa5-containing GABAa R positive allosteric modulator (s) to treat the subject using the compositions and methods ofthe invention. It is understood that the dosage regimen will be determined for an individual, taking into considération, for example, various factors that modify the action of a5-containing • 142
GABAa R positive allosteric modulator, the severity or stage ofthe disease, route of administration, and characteristics unique to the individual, such as âge, weight, size, and extent of cognitive impairment.
[0173] It is well-known in the art that normalization to body surface area is an appropriate method for extrapolating doses between species. To calculate the human équivalent dose (HED) from a dosage used in the treatment of age-dependent cognitive impairment in rats, the formula HED (mg/kg) = rat dose (mg/kg) x 0.16 may be employed(see Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, December 2002, Center for Biologics Evaluation and Research). For example, using that formula, a dosage of 10 mg/kg în rats îs équivalent to 1.6 mg/kg in humans. This conversion is based on a more general formula HED = animal dose in mg/kg x (animal weight in kg/human weight in kg)033[0174] In certain embodiments of the invention, the dose of the a5-containing GABAa R positive allosteric modulator is between 0.0001 and 100 mg/kg/day (which, given a typical 15 human subject of 70 kg, is between 0.007 and 7000 mg/day).
[0175[ In certain embodiments of the invention, the interval of administration îs once every 12 or 24 hours. Administration at less frequent intervals, such as once every 6 hours, may also be used.
[0176] If administered by an implant, a device or a slow or extended release formulation, the a5-containing GABAa R positive allosteric modulator can be administered one time, or one or more times periodically throughout the lifetime of the patient as necessary. Other administration intervals intermediate to or shorter than these dosage intervals for clinical applications may also be used and may be determined by one skilled in the art following the methods of this invention.
[0177] Desired time of administration can be determined by routine expérimentation by one skilled in the art. For example, the a5-containing GABAa R positive allosteric modulator may be administered for a period of 1-4 weeks, I -3 months, 3-6 months, 6-12 months, I -2 years, or more, up to the lifetime of the patient.
143 [0178] In addition to a5-containing GABAa R positive allosteric modulator, the compositions ofthis invention can also include other therapeutically useful agents. These other therapeutically useful agents may be administered în a single formulation, simultaneously or sequentially with the a5-containing GABAa R positive allosteric modulator according to the methods of the invention.
]0179] It will be understood by one of ordinary skill în the art that the compositions described herein may be adapted and modified as is appropriate for the application being addressed and that the compositions described herein may be employed în other suitable applications. For example, the compositions of this application may further comprise a second therapeutic agent. Such other additions and modifications will not départ from the scope hereof.
Pharmaceutical Compositions with Antipsychotics [0180] The compounds or the compositions of this application may be used in combination with an antipsychotic in treating cognitive impairment associated with schizophrenia or bipolar disorder în a subject having or at risk of said schizophrenia or bipolar disorder (e.g., mania). The antipsychotic or a pharmaceutically acceptable sait, hydrate, solvaté or polymorph thereof that is useful în the methods and compositions of this invention include both typical and atypical antipsychotics. In some embodiments, the compounds or the compositions of the présent invention may be used to treat one or more positive and/or négative symptoms, as well as cognitive impairment, associated with schizophrenia. In some embodiments, the compounds or the compositions ofthe présent invention may be used to treat one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). In some embodiments ofthis invention, the compounds or the compositions of this invention prevent or slow the progression of cognitive impairment of schizophrenia or bipolar disorder (in particular, mania) in said subject.
[0181] In some embodiments, the antipsychotics suitable for use în the présent invention are selected from atypical antipsychotics. Such atypical antipsychotics include, but are not limited to, those disclosed in, for example, U.S. Patents 4,734,416; 5,006,528;
4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500;
144
4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvatés, and polymorphs thereof.
[0182] In some embodiments, atypical antipsychotics suitable for use in the présent invention include, but are not limited to, arîpiprazole, asenapïne, clozapine, iloperidone, olanzapine, lurasîdone, paliperidone, quetiapine, rispéridone and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvatés, and polymorphs thereof. In some embodiments, the antipsychotic suitable for use herein is selected from arîpiprazole (Bristol-Myers Squibb), olanzapine (Lilly) and ziprasidone (Pfizer), and the pharmaceutically acceptable salts, hydrates, solvatés, and polymorphs thereof.
[0183] In some embodiments, the antipsychotics suitable for use in the présent invention are typical antipsychotics, including, but not limited to, acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, dîazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapîne, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pitnozide, pipamperone, pîpotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and the pharmaceutically acceptable salts, hydrates, solvatés, and polymorphs thereof.
[0184] In some embodiments of the présent invention, the antipsychotic or a pharmaceutically acceptable sait, hydrate, solvaté or polymorph thereof may be selected from compounds that are dopaminergic agents (such as dopamine DI receptor antagonists or agonists, dopamine D2 receptor antagonists or partial agonists, dopamine D3 receptor antagonists or partial agonists, dopamine D4 receptor antagonists), glutamatergic agents, N-methyl-D-aspartate (NMDA) receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), Ml muscarinic acétylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, a-amino-3-hydroxy-5-methylisoxazole-4-propionic acid • 145 (AMPA)/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents (such as alpha-2 adrenergic receptor agonists or antagonists and catechol-O-methyl transferase (COMT) inhibitors), serotonin receptor modulators (such as 5-HTîa receptor antagonists, 5-HTia receptor partial agonists, 5-HT2c agonists, and 55 HT6 antagonists, serotonin 2C agonists), cholinergic agents (such as alpha-7 nicotinic receptor agonists or PAMs, alpha4-beta2 nicotinic receptor agonists, allosteric modulators of nicotinic receptors and acetylcholinesterase inhibitors, muscarinic receptor agonists and antagonists), cannabinoîd CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, monoamine oxidase (MAO) B inhibitors, PDE10 inhibitors, neuronal nitric oxide synthase (nNOS) inhibitors, neurosteroids, and neurotrophic factors.
[0185] In some embodiments, an a5-containing GABAa receptor positive allosteric modulator as described herein and an antipsychotic as described herein, or their pharmaceutically acceptable salts, hydrates, solvatés or polymorphs, are administered simultaneously, or sequentially, or in a single formulation, or in separate formulations packaged together. In other embodiments, the a5-containing GABAa receptor positive allosteric modulator and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvatés or polymorphs, are administered via different routes. As used herein, “combination” includes administration by any of these formulations or routes of administration.
Pharmaceutical Compositions with Memantine [0186] The compounds or the compositions of this application may be used in combination with memantine or a dérivative or an analog thereof in treating cognitive impairment associated with central nervous system (CNS) disorders in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI, Age-Associated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia or bipolar disorder, amyotrophie latéral sclerosis (ALS) and cancertherapy-related cognitive impairment.
[0187] Memantine, chemically also known as 3,5-dimethyladamantan-l -amine or 3,5 • 146 dimethyltricyclo[3.3.l.l3,7]decan-l-amine, isanuncompetîtîveN-methyl-D-aspartate (NMDA) receptor antagonist with moderate affinity. The proprietary names for memantine include: Axura® and Akatinol® (Merz), Namenda® (Forest Laboratories), Ebixa® and Abixa® (Lundbeck), and Memox® (Unipharm). Memantine is currently available in the U.S. and in over 42 countries worldwide. It is approved for the treatment of moderate to severe Alzheimefs disease (AD) in the United States at a dose of up to 28 mg/day. Memantine and some of its dérivatives and analogs that are useful in the présent invention are disclosed in U.S. Patents Nos. 3,391,142; 4,122,193; 4,273,774; and 5,061,703, all ofwhich are hereby incorporated by reference. Other memantine dérivatives or analogs that are useful in the présent invention include, but are not limited to, those compounds disclosed in U.S. Patent Application Publication US20040087658, US20050113458, US20060205822, US20090081259, US20090124659, and US20100227852; EP Patent Application Publication EP2260839A2; EP Patent EP1682109BI; and PCT Application Publication WO2005079779, all of which are incorporated herein by reference. Memantine, as used in the présent invention, includes memantine and its dérivatives and analogs, as well as hydrates, polymorphs, prodrugs, salts, and solvatés thereof. Memantine, as used herein, also includes a composition comprising memantine or a dérivative or an analog or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, or prodrug thereof, wherein the composition optionally further comprises at least one additional therapeutic agent (such as a therapeutic agent useful for treating a CNS disorder or cognitive impairments associated thereof). In some embodiments, the memantine composition suitable for use in the présent invention comprises memantine and a second therapeutic agent that is donepezil (under the trade name Arîcept).
[01881 In other embodiments of the invention, the a5-containing GABAa receptor positive allosteric modulator and memantine (or the memantine derivative/analog), or their pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, or prodrugs are administered simultaneously, or sequentially, or in a single formulation or in separate formulations packaged together. In other embodiments, the a5-containing GABAa receptor positive allosteric modulator and memantine (or the memantine derivative/analog), or their pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, or prodrugs are administered via different routes. As used herein,
combination includes administration by any of these formulations or routes of administration.
Pharmaceutical Compositions with Acétylcholine Esterase Inhibitors (AChE-Is) [0189] The compounds or the compositions of this application may be used in combination with an acétylcholine esterase inhibitor in treating cognitive impairment associated with central nervous system (CNS) disorders in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestîc MCI, Age-Assocîated Memory Impairment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer's
Dîsease(AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia or bipolar disorder, amyotrophie latéral sclerosis (ALS) and cancer-therapy-related cognitive impairment.
[0190| AChE-Is known to a person ofordinary skill in the art may belong to the subcategories of (i) réversible non-competîtive inhibitors or réversible compétitive inhibitors, (ii) irréversible, and/or (iii) quasi-irreversible inhibitors.
[0191| In certain embodiment, AChE-Is useful in the présent invention include those described in PCT applications W02014039920 and W02002032412; EP patents Nos. 468187; 481429-A; and U.S. Patents Nos. 4,816,456; 4,895,841; 5,041,455; 5,106,856; 5,602,176; 6,677,330; 7,340,299; 7,635,709; 8,058,268; 8,741,808; and 8,853,219, ail of 20 which are incorporated herein by reference.
[0192] In certain embodiment, typical AChE-Is that may be used in accordance with this invention include, but are not Iimited to, ungeremine, ladostigil, demecarium, echothiophate (Phospholine), edrophonium (Tensilon), tacrine (Cognex), Pralidoxime (2PAM), pyridostigmine (Mestinon), physostîgmine (serine, Antilirium), abmenonium (Mytelase), galantamine (Remînyl, Razadyne), rivastîgmine (Exelon, SZD-ENA-713),
Huperzine A, Icopezil, neostigmine (Prostigmin, Vagostigmin), Arîcept (Donepezîl,
E2020), Lactucopîcrin, monoamine acridines and their dérivatives, piperidine and piperazine dérivatives, N-benzyl-piperidine dérivatives, piperidinyl-alkanoyl heterocyclic compounds, 4-(l-benzyl:piperidyl)-substituted fused quinoline dérivatives and cyclic amide dérivatives. Other typical AChE-Is include carbamates and organophosphonate φ 148 compounds such as Metrifonate (Trichlorfon). Benzazepinols such as galantamine are also useful AChE-Is. In some embodiment, AChE-Is suitable for use in combination with the compounds and compositions ofthis application include: Donepezil (aricept), Galantamine (razadyne), or Rivastigmine (exelon).
[0193] In other embodiments ofthe invention, the aS-containîng GABAa receptor positive allosteric modulator and the AChE-I, or their pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, or prodrugs are administered simultaneously, or sequentially, or in a single formulation or in separate formulations packaged together. In other embodiments, the a5-containing GABAa receptor positive allosteric modulator and the AChE-I, or their pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, or prodrugs are administered via different routes. As used herein, combination includes administration by any of these formulations or routes of administration.
[0194] In some embodiments, the compounds and compositions described herein are for use as a médicament. In some embodiments, the compounds and compositions of the présent invention are for use in treating cognitive impaîrment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impaîrment. In some embodiments, the CNS disorder with cognitive impaîrment includes, without limitation, age-related cognitive impaîrment, Mild Cognitive Impaîrment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impaîrment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-related cognitive impaîrment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavîor, and substance addiction.
[0195] In some embodiments, this application provides the use of a compound or composition described herein in the préparation of a médicament for the treatment of cognitive impaîrment associated with a CNS disorder in a subject in need of treatment or at risk ofsaid cognitive impaîrment. In some embodiments, the CNS disorder with cognitive impaîrment includes, without limitation, age-related cognitive impaîrment, Mild
Cognitive Impaîrment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impaîrment (AAMI), Age Related Cognitive Décliné (ARCD), dementia, Alzheimer’s
149
Disease (AD), prodroma! AD, post traumatic stress disorder (PTSD), schizophrenîa, bipolar disorder, amyotrophie latéral sclerosis (ALS), cancer-therapy-rdated cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
Methods of assessing cognitive impairment [0196] Animal models serve as an important resource for developing and evaluating treatments for cognitive impairment associated with CNS disorders. Features that characterize cognitive impairment in animal models typically extend to cognitive impairment în humans. Efficacy in such animal models is, thus, expected to be prédictive of efficacy in humans. The extent of cognitive impairment in an animal mode! for a CNS disorder, and the efficacy of a method of treatment for said CNS disorder may be tested and confirmed with the use of a variety of cognitive tests.
[0197] A Radial Arm Maze (RAM) behavioral task is one example of a cognitive test, specifically testing spaciai memory (Chappeli et al. Neuropharmacology 37:481-487, 1998). The RAM apparatus consists of, e.g., eïght equîdîstantly spaced arms. A maze arm projects from each facet of a center platform. A food weil is located at the distal end of each arm. Food is used as a reward. Blocks can be positioned to prevent entry to any arm. Numerous extra maze eues surrounding the apparatus may also be provided. After habituation andtraîning phases, spatial memory ofthe subjects may be tested în the RAM under control or test compound-treated conditions. As a part of the test, subjects are pretreated before trials with a vehicle control or one of a range of dosages of the test compound. At the begînnîng of each trial, a subset of the arms of the eîght-arm maze is blocked. Subjects are allowed to obtain food on the unblocked arms to which access is permitted during this initia! “information phase” ofthe trial. Subjects are then removed from the maze for a delay period, e.g., a 60 second delay, a 15 minute delay, a one-hour delay, a two-hour delay, a six hour delay, a 24 hour delay, or longer) between the information phase and the subséquent “rétention test, during which the barriers on the maze are removed, thus allowing access to ail eight arms. After the delay period, subjects are placed back onto the center platform (with the barriers to the previously blocked arms removed) and allowed to obtain the remaining food rewards during this rétention test phase ofthe trial. The identity and configuration of the blocked arms vary across trials.
150
The number of “errors” the subjects make during the rétention test phase is tracked. An error occurs in the trial if the subjects entered an arm from which food had already been retrieved in the pre-delay component of the trial, or if it re-visits an arm in the post-delay session that had already been visited. A fewer number of errors would indicate better spatial memory. The number of errors made by the test subject, under various test compound treatment régimes, can then be compared for efïîcacy ofthe test compound in treating cognitive impairment associated with CNS disorders.
[0198] Another cognitive test that may be used to assess the effects of a test compound on the cognitive impairment of a CNS disorder model animal is the Morris water maze. A water maze is a pool surrounded with a novel set of patterns relative to the maze. The training protocol for the water maze may be based on a modified water maze task that has been shown to be hîppocampal-dependent (de Hoz et al., Eur. J. Neurosci., 22:745-54, 2005; Steele and Morris, Hippocampus 9:118-36, 1999). The subject îs traîned to locate a submerged escape platform hidden undemeath the surface ofthe pool. During the training trial, a subject is released in the maze (pool) from random starting positions around the perimeter ofthe pool. The starting position varies from trial to trial. Ifthe subject does not locate the escape platform within a set time, the expérimenter guides and places the subject on the platform to “teach” the location of the platform. After a delay period following the last training trial, a rétention test in the absence of the escape platform is given to assess spatial memory. The subject’s level of preference for the location ofthe (now absent) escape platform, as measured by, e.g., the time spent in that location or the number of cross in gs ofthat location made by the mouse, indicates better spatial memory, i.e., treatment of cognitive impairment. The preference for the location of the escape platform under different treatment conditions, can then be compared for efïîcacy ofthe test compound in treating cognitive impairment associated with CNS disorders.
[0199] There are various tests known in the art for assessîng cognitive function in humans, for example and without limitation, the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatrie Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Sélective Reminding Test (Buschke and
151
Fuld, 1974); the Verbal Paired Associâtes subtest; the Logical Memory subtest; the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997); the Benton Visual Rétention Test, or MATRICS consensus neuropsychological test battery which includes tests of working memory, speed of processing, attention, verbal leaming, visual leaming, reasoning and problem solving and social cognition. See Folstein et al., J Psychiatrie Res 12:189-98, (1975); Robbins et al., Dementia 5:266-81, (1994); Rey, L’examen clinique en psychologie, (1964); Kluger et al., JGeriatr Psychiatry Neurol 12:168-79, (1999); Marquis et a!., 2002 and Masur et al., 1994. Also see Buchanan, R.W., Keefe, R.S.E., Umbricht, D., Green, M.F., Laughren,
T., and Marder, S.R. (2011)The FDA-NIMH-MATRICS guidelines for clinical trial design ofcognitive-enhancing drugs: what do we know 5 years later? Schizophr. Bull. 37, 1209-1217. Another example of a cognitive test in humans is the explicit 3-altemative forced choice task. In this test, subjects are presented with color photographs ofcommon objects consisting of a mix of three types of image pairs: similar pairs, identical pairs and unrelated foils. The second of the pair of similar objects is referred to as the lure. These image pairs are fully randomized and presented individually as a sériés of images. Subjects are instructed to make a judgment as to whether the objects seen are new, old or similar. A “similar” response to the présentation ofa lure stimulus indicates successful memory retrieval by the subject. By contrast, calling the lure stimulus “old” or “new” indicates that correct memory retrieval did not occur.
[0200] In addition to assessing cognitive performance, the progression of age-related cognitive impairment and dementia, as well as the conversion of age-related cognitive impairment into dementia, may be monitored by assessing surrogate changes in the braîn of the subject. Surrogate changes include, without limitation, changes in régional brain 25 volumes, perforant path dégradation, and changes seen in brain function through resting state fMRI (R-fMRI) and fluorodeoxyglucose positron émission tomography (FDG-PET). Examples of régional brain volumes useful in monîtoring the progression of age-related cognitive impairment and dementia include réduction of hippocampal volume and réduction in volume or thickness of entorhinal cortex. These volumes may be measured in a subject by, for example, MRI. Aisen et al., Alzheimer’s & Dementia 6:239-246 (2010). Perforant path dégradation has been shown to be linked to âge, as well as reduced cognitive function. For example, older adults with more perforant path dégradation tend to perform worse in hippocampus-dependent memory tests. Perforant path dégradation
may be monitored in subjects through ultrahigh-resolution diffusion tensor imaging (DTI). Yassa et al., PNAS 107:12687-12691 (2010). Resting-state fMRI (R-fMRI) involves imaging the brain during rest, and recording large-amplitude spontaneous lowfrequency (<0.I Hz) fluctuations in the fMRI signal that are temporally correlated across functlonally related areas. Seed-based functional connectivity, independent component analyses, and/or frequency-domain analyses of the signais are used to reveal functional connectivity between brain areas, particularly those areas whose connectivity increase or decrease with âge, as well as the extent of cognitive impairment and/or dementia. FDGPET uses the uptake of FDG as a measure of régional metabolic activity in the brain.
Décliné of FDG uptake in régions such as the posterior cingulated cortex, temporoparietal cortex, and prefrontal association cortex has been shown to relate to the extent of cognitive décliné and dementia. Aisen et al., Alzheimer’s & Dementia 6:239-246 (2010), Herholz et al., Neuroimage 17:302-316 (2002).
Age-Related Cognitive Impairment [0201] The invention provides methods and compositions for treating age-related cognitive impairment or the risk thereof using a a5-containing GABAa receptor positive allosteric modulator (i.e., a compound of the invention), such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, ofage-related cognitive impairment. ln certain embodiments, treatment comprises allevïation, amelioration or slowing the progression, of one or more symptoms associated with age-related cognitive impairment. In certain embodiments, treatment of age-related cognitive impairment comprises slowing the conversion of age-related cognitive impairment (including, but not limited to MCI,
ARCD and AAMI) into dementia (e.g., AD). The methods and compositions may be used for human patients in clinical applications in the treating age-related cognitive impairment in conditions such as MCI, ARCD and AAMI or for the risk thereof. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications. In some embodiments ofthe invention, there is provided a method of preserving or improving cognitive function in a subject with age-related cognitive impairmenL the method comprising the step of administering to said subject a therapeutically effective amount of a compound ofthe invention or a
153
pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
[0202] In some embodiments, a subject to be treated by the methods and compositions of this invention exhibits age-related cognitive impairment or is at risk of such impairment. In some embodiments, the age-related cognitive impairment includes, without limitation, Age-Associated Memory Impairment (AAMI), Mild Cognitive Impairment (MCI) and Age-related Cognitive Décliné (ARCD).
[0203] Animal models serve as an important resource for developing and evaluating treatments for such age-related cognitive impairments. Features that characterize ageI0 related cognitive impairment in animal models typically extend to age-related cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be prédictive of efficacy in humans.
J0204] Various animal models of age-related cognitive impairment are known in the art.
For example, extensive behavioral characterization has identified a naturally occurring I5 form of cognitive impairment in an outbred strain of aged Long-Evans rats (Charles River
Laboratories; Gallagher er al., Behav. Neurosci. 107:618-626, (1993)). In a behavioral assessment with the Morris Water Maze (M WM), rats leam and remember the location of an escape platform guided by a configuration of spatial eues surrounding the maze. The cognitive basis of performance is tested in probe trials using measures ofthe animal’s 20 spatial bîas in searching for the location of the escape platform. Aged rats in the study population hâve no difïïculty swimming to a visible platform, but an age-dependent impairment is detected when the platform is camouflaged, requiring the use of spatial information. Performance for individual aged rats in the outbred Long-Evans strain varies greatly. For example, a proportion of those rats perform on a par with young adults. However, approximately 40-50% fall outside the range ofyoung performance.
This variability among aged rats reflects relïablc individual différences. Thus, within the aged population some animais are cognitively impaired and designated aged-impaîred (AI) and other animais are not impaired and are designated aged-unimpaired (AU). See, e.g., Colombo et al., Proc. Natl. Acad. Sci. 94: 14195-14199, (1997); Gallagher and
Burwell, Neurobiol, Aging 10: 691-708, (1989); Gallagher et al. Behav. Neurosci.
107:618-626, (1993); Rapp and Gallagher, Proc. Natl. Acad. Sci. 93: 9926-9930, (1996);
154
Nicolle et al.. Neuroscience 74:741 -756, (1996); Nicolle et ai., J. Neurosci. 19:96049610, (1999); International Patent Publication W02007/019312 and International Patent Publication WO 2004/048551. Such an animal model of age-related cognitive impairment may be used to assay the effectiveness of the methods and compositions this invention in treating age-related cognitive impairment.
[0205[ The efficacy of the methods and compositions of this invention in treating agerelated cognitive impairment may be assessed using a variety of cognitive tests, including the Morris water maze and the radial arm maze, as discussed herein.
Dementia [0206] The invention also provides methods and compositions for treating dementia using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of dementia. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with dementia. In certain embodiments, the symptom to be treated is cognitive impairment. In some embodiments ofthe invention, there is provided a method of preserving or improving cognitive function în a subject with dementia, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof. In certain embodiments, the dementia is Alzheimer’s disease (AD), vascular dementia, dementia with Lewy bodies, or frontotemporal dementia. The methods and compositions may be used for human patients in clinical applications in treating dementia. The dose ofthe composition and dosage interval for the method is, as described herein, one that is safe and effîcacious in those applications.
[0207] Animal models serve as an important resource for developing and evaluating treatments for dementia. Features that characterize dementia in animal models typically extend to dementia in humans. Thus, efficacy in such animal models îs expected to be prédictive ofefficacy in humans. Various animal models ofdementia are known in the art, such as the PDAPP, Tg2576, APP23, TgCRND8. J20, hPS2 Tg, and APP + PSI
155 transgenic mice. Sankaranarayanan, Curr. Top. Médicinal Chem. 6:609-627,2006; Kobayashi et al. Genes Brain Behav. 4: 173-196.2005; Ashe and Zahns, Neuron. 66:
631-45,2010. Such animal models of dementia may be used to assay the effectiveness of the methods and compositions of this invention of the invention în treating dementia.
[0208] The efficacy ofthe methods and compositions ofthis invention în treating dementia, or cognitive impairment associated with dementia, may be assessed in animais models of dementia, as well as human subjects with dementia, using a variety of cognitive tests known in the art, as discussed herein.
Post Traumatic Stress Disorder [0209] The invention also provides methods and compositions for treating post traumatic stress disorder (PTSD) using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, îsomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of PTSD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with PTSD. In certain embodiments, the symptom to be treated is cognitive impairment. In some embodiments ofthe invention, there is provided a method of preserving or improving cognitive function in a subject with PTSD, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications în treating PTSD. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacîous in those applications.
[0210] Patients with PTSD (and, to a lesser degree trauma-exposed patients without
PTSD) hâve smaller hîppocampal volumes (Woon et al., Prog. Neuro-Psychopharm. &
Biological Psych. 34, 1181-1188; Wang et al., Arch. Gen. Psychiatry 67:296-303,2010).
PTSD is also associated with impaired cognitive performance. Older individuals with
PTSD hâve greater déclinés in cognitive performance relative to control patients (Yehuda
étal., Βίο. Psych. 60:714-721,2006) and hâve a greater lïkelïhood of developing dementia (Yaffeer al., Arch. Gen. Psych. 678:608-613,2010).
[0211] Animal models serve as an important resource for developing and evaluating treatments for PTSD. Features that characterize PTSD in animal models typically extend 5 to PTSD in humans. Thus, efficacy in such animal models is expected to be prédictive of efficacy in humans. Various animal models of PTSD are known in the art.
[0212] One rat model of PTSD is Time-dependent sensitization (TDS). TDS involves exposure ofthe animal to a severely stressful event followed by a situational reminder of the prior stress. The following is an example of TDS. Rats are placed in a restrainer, then placed in a swim tank and made to swim for a period of time, e.g., 20 min. Following this, each rat is then immediately exposed to a gaseous anesthetic until loss of consciousness, and finally dried. The animais are left undisturbed for a number of days, e.g., one week. The rats are then exposed to a “restress” session consisting of an initial stressor, e.g., a swimming session in the swim tank (Liberzon et al.,
Psychoneuroendocrinology 22:443-453, 1997; Harvery et al., Psychopharmacology 175:494-502,2004). TDS results in an enhancement of the acoustic startle response (ASR) in the rat, which is comparable to the exaggerated acoustic startle that is a prominent symptom of PTSD (Khan and Liberzon, Psychopharmacology 172:225-229, 2004). Such animal models ofPTSD may be used to assay the effectiveness ofthe methods and compositions of this invention of the invention in treating PTSD.
[0213] The efficacy of the methods and compositions of this invention in treating PTSD, or cognitive impairment associated with PTSD, may also be assessed in animais models of PTSD, as well as human subjects with PTSD, using a variety of cognitive tests known in the art, as discussed herein.
Schizophrénie and Bipolar Disorder [0214] The invention addîtionally provides methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania) using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises φ 157 preventing or slowing the progression of schizophrenia or bipolar disorder (in particular, mania). Schizophrenia is characterized by a wide spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental représentations (e.g., hallucinations, delusions), or dopamine dysregulation-associated symptoms (e.g., hyperdopaminergic responses, hyperdopaminergic behavorial responses, dopaminergic hyperactivity, or hyperlocomotor activity, or psychosis), négative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolitîon), and cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression of one or more positive and/or négative symptoms, as well as cognitive impairment, associated with schizophrenia. Further, there are a number of other psychiatrie diseases such as schizotypical and schizoaffective disorder, other acute- and chronic psychoses and bîpolar disorder (in particular, mania), which hâve an overlapping symptomatology with schizophrenia. In some embodiments, treatment comprises alleviation, amelioration or slowing the progression of one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). In some embodiments ofthe invention, there is provided a method of preserving or improving cognitive function in a subject with schizophrenia or bipolar disorder, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating schizophrenia or bipolar disorder (in particular, mania). The dose ofthe composition and dosage interval for the method is, as described herein, one that is safe and efïicacious in those applications.
[0215] Cognitive impairments are associated with schizophrenia. They précédé the onset of psychosis and are présent in non-affected relatives. The cognitive impairments associated with schizophrenia constitute a good predictor for functional outcome and are a core feature ofthe disorder. Cognitive features în schizophrenia reflect dysfonction in frontal cortical and hippocampal circuits. Patients with schizophrenia also présent hippocampal pathologies such as réductions in hippocampal volume, réductions in neuronal size and dysfunctiona! hyperactivity. An imbalance in excitation and inhibition in these brain régions has also been documented in schizophrénie patients suggesting that drugs targeting inhibitory mechanisms could be therapeutic. See, e.g., Guidotti étal., • 158
Psychopharmacology 180: 191-205,2005; Zierhut, Psych. Res. Neuroimag. 183:187-194, 2010; Wood et al., Neuroimage 52:62-63,2010; Vinkers et al.. Expert Opirt. Investig. Drugs 19:1217-1233, 2009; Younget a/., Pharmacol. Ther. 122:150-202,2009.
[0216] Animal models serve as an important resource for developing and evaluating treatments for schizophrenia. Features that characterize schizophrenia in animal models typically extend to schizophrenia in humans. Thus, efïïcacy in such animal models îs expected to be prédictive ofefïïcacy in humans. Various animal models of schizophrenia are known în the art.
[0217] One animal model of schizophrenia is protracted treatment with méthionine.
Methionine-treated mice exhibit déficient expression of GAD67 in frontal cortex and hippocampus, similar to those reported in the brain of postmortem schizophrenia patients. They also exhibit prepulse inhibition of startle and social interaction déficits (Tremonlizzo et al., PNAS, 99:17095-17100,2002). Another animal model of schizophrenia is methylaoxymethanol acetate (MAM)-treatment in rats. Prégnant female rats are administered MAM (20 mg/kg, intraperitoneal) on gestational day 17. MAMtreatment recapitulate a pathodevelopmental process to schizophrenia-like phenotypes in the offspring, including anatomîcal changes, behavioral déficits and altered neuronal information processing. More specifically, ΜΑΜ-treated rats display a decreased density ofparvalbumin-positive GABAergic intemeurons în portions ofthe prefrontal cortex and hippocampus. In behavioral tests, MAM-treated rats display reduced latent inhibition. Latent inhibition is a behavioral phenomenon where there îs reduced leaming about a stimulus to which there has been prior exposure with any conséquence. Thîs tendency to disregard previously benign stimulî, and reduce the formation of association with such stirtiuli is belïeved to prevent sensory overload. Low latent inhibition is indicative of psychosis. Latent inhibition may be tested in rats în the following manner. Rats are divided into two groups. One group is pre-exposed to a tone over multiple trials. The other group has no tone présentation. Both groups are then exposed to an audîtory fear conditionîng procedure, in which the same tone is presented concurrently with a noxious stimulus, e.g. an electric shock to the foot. Subsequently, both groups are presented with the tone, and the rats* change în locomotor activity during tone présentation îs monitored.
Afïer the fear conditionîng the rats respond to the tone présentation by strongly reducing locomotor activity. However, the group that has been exposed to the tone before the
conditioning period dîsplays robust latent inhibition: the suppression of locomotor activity in response to tone présentation is reduced. MAM-treated rats, by contrast show impaired latent inhibition. That is, exposure to the tone previous to the fear conditioning procedure has no significant effect in suppressing the fear conditioning. (see Lodge et al., 5 J. Neurosci., 29:2344-2354,2009) Such animal models of schîzophrenia may be used to assay the effectîveness of the methods and compositions of the invention in treating schîzophrenia or bipolar disorder (in particular, mania).
[0218] MAM-treated rats display a significantly enhanced locomotor response (or aberrant locomotor activity) to low dose D-amphetamine administration. The MAM10 treated rats also display a significantly greater number of spontaneously firing ventral tegmental dopamine (DA) neurons. These results are believed to be a conséquence of excessive hippocampal activity because in MAM-treated rats, the ventral hippocampus (vHipp) inactivation (e.g., by intra-vHipp administration of a sodium channel blocker, tetrodotoxin (TTX), to MAM rats) completely reversed the elevated DA neuron population activity and also normalized the augmented amphetamîne-induced locomotor behavior. The corrélation of hippocampal dysfunction and the hyper-responsivity of the DA system is believed to underlie the augmented response to amphétamine in MAMtreated animais and psychosis in schîzophrenia patients. See Lodge D. J. et al. Neurobiology of Disease (2007), 27(42), 11424-11430. The use of MAM-treated rats în the above study may be suitable for use to assay the effectiveness of the methods and compositions of the présent invention in treating schîzophrenia or bipolar disorder (in particular, mania). For example, the methods and compositions of this invention maybe evaluated, using MAM-treated animais, for their effects on the central hippocampus (vHipp) régulation, on the elevated DA neuron population activity and on the hyperactive locomotor response to amphétamine in the MAM-treated animais.
[0219] In MAM-treated rats, hippocampal (HPC) dysfunction leads to dopamine system hyperactivity. A benzodiazepine-positive allosteric modulator (PAM), sélective for the a5 subunit of the GABAa receptor, SH-053-2’F-R-CH3, is tested for its effects on the output ofthe hippocampal (HPC). The effect of SH-053-2’F-R-CH3 on the hyperactive 30 locomotor response to amphétamine in MAM-treated animais is also examîned. The aSGABAAR PAM reduces the number of spontaneously active DA neurons in the ventral tegmental area (VTA) of MAM rats to levels observed in saline-treated rats (control
160 group), both when administered systemically and when directly infused into the ventral HPC. Moreover, HPC neurons in both saline-treated and MAM-treated animais show diminished cortical-evoked responses following the a5GABAAR PAM treatment. In addition, the increased locomotor response to amphétamine observed în MAM-treated rats is reduced following the o5GABAaR PAM treatment. See Gill K. M et al. Neuropsychopharmacology (2011), 1-9. The use of MAM-treated rats in the above study may be suitable for use in the présent invention to assay the effectiveness ofthe methods and compositions of the invention in treating schizophrenîa or bipolar disorder (in particular, mania). For example, the methods and compositions of this invention maybe evaluated, using MAM-treated animais, for their effects on the output of the hippocampal (HPC) and on the hyperactive locomotor response to amphétamine in the MAM-treated animais.
[0220] Administration of MAM to prégnant rats on embryonîc day 15 (El 5) severely impairs spatial memory or the ability to leam the spatial location of four items on an eight-arm radial maze în the offspring. In addition, embryonic day 17 (E17) MAMtreated rats are able to reach the level of performance of control rats at the initial stages of traîning, but are unable to process and retrieve spatial information when a 30-min delay is interposed, indicating a significant impainnent in working memory. See Gourevitch R. et al. (2004). Behav. Pharmacol, 15,287-292. Such animal models of schizophrenîa may be used to assay the effectiveness ofthe methods and compositions ofthe invention in treating schizophrenîa or bipolar disorder (in particular, mania).
[0221] Apomorphine-induced climbing (AIC) and stéréotypé (AIS) in mice is another animal model useful in this invention. Agents are administered to mice at a desired dose level (e.g., via intraperitoneal administration). Subsequently, e.g., thirty minutes later, experimental mice are challenges with apomorphine (e.g., with l mg/kg sc). Five minutes after the apomorphine injection, the snïffing-licking-gnawing syndrome (stereotyped behavior) and climbing behavior induced by apomorphine are scored and recorded for each animal. Readings can be repeated every 5 min during a 30-mïn test session. Scores for each animal are totaled over the 30-min test session for each syndrome (stereotyped behavior and climbing). If an effect reached at least of 50% inhibition, and ID$o value (95% confidence interval) is calculated using a nonlinear least squares calculation with inverse prédiction. Mean climbing and stéréotypé scores can be
161 expressed as a percent of control values observed in vehicle treated (e.g., saline-treated) mice that receive apomorphine. See Grauer S. M. et al. Psychopharmacology (2009) 204, 37-48. This mouse model may be used to assay the effectiveness ofthe methods and compositions ofthe invention in treating schizophrenia or bipolar disorder (în particular, mania).
[0222] In another well-established preclinical model of schizophrenia, rats exposed chronically to ketamine, an uncompetitive N-methy!-D-aspartate (NMDA) receptor antagonist, produces positive and négative psychotic symptoms and cognitive impairment. Long-Evans male rats are injected intraperitoneally with ketamine (30 mg/kg, twice a day) for two weeks during adolescence (2 month-old). Rats are behaviorally tested when they reach adulthood (approximately 4-5 month-old) for the behavioral symptoms to ketamine exposure and for the efficacy of treatment to alleviate those symptoms. See, e.g., Enomoto et al. Progress in Neuro-Psychopharmacology & Biological Psychiatry 33 (2009) 668-675.
[0223] The efficacy ofthe methods and compositions ofthis invention în treating schizophrenia or cognitive impairment associated therewith may also be assessed in animal models of schizophrenia or bipolar disorder (in particular, mania), as well as human subjects with schizophrenia, using a variety of cognitive tests known in the art, as dîscussed herein.
Amyotrophie Latéral Sclerosls (ALS) [0224] The invention addîtionally provides methods and compositions for treating ALS using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of ALS. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated wîth ALS. In certain embodiments, the symptom to be treated is cognitive impairment. In some embodiments ofthe invention, there is provided a method of preserving or improving cognitive fonction in a subject with ALS, the method comprising the step of administering to said subject a therapeutically effective amount of a compound ofthe invention or a pharmaceutically acceptable sait.
162 hydrate, solvaté, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating ALS. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efïïcacious în those applications.
[0225] In addition to the degeneration of motor neurons, ALS is characterîzed by neuronal degeneration in the entorhînal cortex and hippocampus, memory déficits, and neuronal hyperexcitability in different brain areas such as the cortex.
[0226] The efficacy of the methods and compositions of this invention in treating ALS, or cognitive impairment associated with ALS, may also be assessed in animal models of ALS, as well as human subjects with ALS, using a variety of cognitive tests known in the art, as discussed herein.
Cancer therapy-related cognitive impairment [0227] The invention additionally provides methods and compositions for treating cancer therapy-related cognitive impairment using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, ofcancer therapy-related cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with cancer therapy-related cognitive impairment. In some embodiments ofthe invention, there is provided a method of preserving or improving cognitive function in a subject with cancer therapy-related cognitive impairment, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating cancer therapy-related cognitive impairment. The dose ofthe composition and dosage interval for the method is, as described herein, one that is safe and efïïcacious in those applications.
φ 163 [0228] Thérapies that are used in cancer treatment, including chemotherapy, radiation, or combinations thereof, can cause cognitive impaîrment in patients, în such functions as memory, leaming and attention. Cytotoxicity and other adverse side-effects on the brain of cancer thérapies are the basis for this form of cognitive impaîrment, which can persist for décades. (Dietrich et al., Oncologist 13:1285-95,2008; Soussain étal., Lancet 374:1639-51,2009).
[0229] Cognitive impaîrment following cancer thérapies reflects dysfonction in frontal cortical and hippocampal circuits that are essential for normal cognition. In animal modeis, exposure to either chemotherapy or radiation adversely affects performance on tests of cognition specifically dépendent on these brain Systems, especially the hippocampus (Kim et al., J. Radiat. Res. 49:517-526,2008; Yang et al., Neurobiol. Leaming and Mem. 93:487-494,2010). Thus, drugs targeting these cortical and hippocampal Systems could be neuroprotective in patients receiving cancer thérapies and efficacious in treating symptoms of cognitive impaîrment that may last beyond the interventions used as cancer thérapies.
[0230] Animal modeis serve as an important resource for developîng and evaluating treatments for cancer therapy-related cognitive impaîrment. Features that characterize cancer therapy-related cognitive impaîrment in animal modeis typically extend to cancer therapy-related cognitive impaîrment în humans. Thus, efficacy in such animal modeis is expected to be prédictive ofefficacy in humans. Various animal modeis of cancer therapy-related cognitive impaîrment are known in the art.
[0231] Examples ofanimal modeis ofcancer therapy-related cognitive impaîrment include treating animais with anti-neoplastic agents such as cyclophosphamide (CYP) or with radiation, e.g., ^Co gamma-rays. (Kim et al., J. Radiat. Res. 49:517-526,2008;
Yang ci or/., Neurobiol. Learning andMem. 93:487-494,2010). The cognitive fonction of animal modeis of cancer therapy-related cognitive impaîrment may then be tested with cognitive tests to assay the efTectiveness of the methods and compositions ofthe invention in treating cancer therapy-related cognitive impaîrment. The efficacy of the methods and compositions ofthis invention in treating cancer therapy-related cognitive impaîrment, as well as human subjects with cancer therapy-related cognitive impaîrment, using a variety of cognitive tests known in the art, as discussed herein.
164
Parkinson’s disease (PD) [0232] Parkinson’s disease (PD) is a neurological disorder characterized by a decrease of voluntary movements. The afïlicted patient has réduction of motor activity and slower voluntary movements compared to the normal individual. The patient has characteristic mask face, a tendency to hurry while walking, bent over posture and generalized weakness of the muscles. There îs a typical lead-pipe” rigidity of passive movements. Another important feature ofthe disease îs the tremor ofthe extremities occurring at rest and decreasing during movements.
|0233] Parkinson’s disease, the etiology of whïch is unknown, belongs to a group of the most common movement disorders named parkinsonism, which affects approximately one person per one thousand. These other disorders grouped under the name of parkinsonism may resuit from viral infection, syphilis, arteriosclerosis and trauma and exposure to toxic chemicals and narcotics. Nonetheless, it is believed that the inappropriate loss of synaptic stability may lead to the disruption of neuronal circuits and to brain diseases. Whether as the resuit ofgenetics, drug use, the aging process, viral infections, or other various causes, dysfunction in neuronal communication is considered the underlying cause for many neurologie diseases, such as PD (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10,207-214).
[0234] Regardless of the cause of the disease, the main pathologie feature is degeneration of dopaminergic cells in basal ganglia, especially in substantia nigra. Due lo prématuré death ofthe dopamine containing neurons in substantia nigra, the largest structure of the basal ganglia, the striatum, will hâve reduced input from substantia nigra resulting in decreased dopamine release. The understanding of the underlying pathology led to the introduction ofthe first successful treatment which can allevîate Parkinson’s disease. Virtually ail approaches to the therapy ofthe disease are based on dopamine replacement. Drugs currently used in the treatment can be converted into dopamine after crossing the blood brain barrier, or they can boost the synthesis of dopamine and reduce its breakdown. Unfortunately, the main pathologie event, degeneration ofthe cells în substantia nigra, is not helped. The disease continues to progress and frequently after a certain length of time, dopamine replacement treatment will lose its efiectiveness.
10235] The invention provides methods and compositions for treating PD using a a5containing GABAa receptor positive allosteric modulator, such as one selected from the
compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphe, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of PD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more 5 symptoms associated with PD. In certain embodiments, the symptom to be treated is cognitive impairment. For example, methods and compositions of the disclosure can be used to improve the motor/cognîtîve impairments symptomatîc of Parkinson’s disease. Moreover, methods and compositions ofthe disclosure may be useful for treating the memory impairment symptomatîc of Parkinson’s disease. In some embodiments of the 10 invention, there is provided a method of preserving or improving cognitive function in a subject with PD, the method comprising the step of administering to said subject a therapeutically effective amount of a compound ofthe invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
[0236] There are a number of animal models for PD. Exemplary animal models for PD 15 include the reserpine model, the methamphetamine model, the 6-hydroxydopamine (6OHDA) model, the l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) model, the paraquat (PQ)-Maneb model, the rotenone model, the 3-nitrotyrosine model and genetic models using transgenic mice. Transgenic models include mice that over express asynuclein, express human mutant forms of a -synudein, or mice that express LRKK2 20 mutations. See review of these models by Ranjita B. et al. (Ranjita B. et al. BioEssays
2002,24,308-318). Additional information regardîng these animal models is readily available from Jackson Laboratories (see also http://research.jax.org/grs/parkinsons.html), as well as in numerous publications disclosing the use of these validated models.
[0237] The efficacy ofthe methods and compositions of this invention in treating PD, or 25 cognitive impairment associated with PD, may be assessed in any of the above animal models of PD, as well as human subjects with PD, using a variety of cognitive tests known in the art, as discussed herein.
Autism [0238] Autism is a neurodevelopmental disorder characterized by dysfonction in three core behavioral dimensions: répétitive behaviors, social déficits, and cognitive déficits.
The répétitive behavior domain involves compulsive behaviors, unusual attachments to objects, rigid adhérence to routines or rituals, and répétitive motor mannerisms such as
166 stereotypies and self- stimulatory behaviors. The social déficit dimension involves déficits in reciprocal social interactions, lack of eye contact, diminished ability to carry on conversation, and impaired daily interaction skjlls. The cognitive déficits can include language abnormalities. Autism is a disabling neurological disorder that affects thousands of Americans and encompasses a number of subtypes, with various putative causes and few documented ameliorative treatments. The disorders of the autistic spectrum may be présent at birth, or may hâve later onset, for example, at âges two or three. There are no clear eut biological markers for autism. Diagnosis of the disorder is made by consîdering the degree to which the child matches the behavioral syndrome, 10 which is characterized by poor communicative abilities, peculiarities in social and cognitive capacities, and maladaptive behavioral patterns. The dysfunction în neuronal communication is considered one of the underlyîng causes for autism (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10,207-214). Recent studîes hâve shown that there is a GABAa a5 déficit in autism spectrum disorder 15 (ASD) and support further investigations of the GABA system in this disorder (Mendez
MA, et al. Neuropharmacology. 2013,68:195-201).
[0239] The invention also provides methods and compositions for treating autism using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, 20 isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of autism. In certain embodiments, treatment comprises allevîation, amelioration, or slowing the progression of one or more symptoms associated with autism. In certain embodiments, the symptom to be treated is cognitive impaîrment or cognitive déficit. For example, methods and compositions of the 25 disclosure can be used to improve the motor/cognitive déficits symptomatic of autism.
In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with autism, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or 30 combination thereof.
[0240] The valproic acid (VPA) rat model of autism using in vitro electrophysiological techniques, established by Rodier et al. (Rodier, P. M. et al. Reprod. Toxicol. 1997,11, 417-422) is one ofthe most exhaustively established insult-based animal models of
167 autism and is based on the observation that prégnant women treated with VPA in the 1960s, during a circumscribed time window of embryogenesis, had a much higher risk of giving birth to an autistic chiid than the normal population. Oflfspring of VPA-exposed prégnant rats show several anatomical and behavioral symptoms typical of autism, such as diminished number of cerebellar Purkinje neurons, impaired social interaction, répétitive behaviors as well as other symptoms of autism, including enhanced fear memory processing. See, Rinaldi T. et al. Frontiers in Neural Circuits, 2008,2,1-7. Another mouse model, BTBR T+tf7J (BTBR) mice, an established model with robust behavioral phenotypes relevant to the three diagnostic behavioral symptoms of autism— unusua! social interactions, impaired communication, and répétitive behaviors—was used to probe the eflficacy ofa sélective négative allosteric modulator ofthe mGluR5 receptor, GRN-529. See, e.g., Silverman J. L. et al. Sci Transi. Med. 2012,4,131.The eflficacy of the methods and compositions of this invention in treating autism, or cognitive déficits associated with autism, may be assessed in the VPA-treated rat model of autism or the BTBR T+tf7J (BTBR) mouse model, as well as human subjects with autism, using a variety of cognitive tests known in the art, as discussed herein.
Mental retardation [0241] Mental retardation is a generalized disorder characterized by significantly impaired cognitive function and déficits in adaptive behaviors. Mental retardation is often defined as an Intelligence Quotient (1Q) score of less than 70. Inbom causes are among many underlying causes for mental retardation. The dysfonction in neuronal communication is also considered one of lhe underlying causes for mental retardation (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10,207-214).
[0242] In some instances, mental retardation includes, but are not limited lo, Down syndrome, velocariofacial syndrome, fêtai alcohol syndrome, Fragile X syndrome, Klinefelter’s syndrome, neurofibromatosis, congénital hypothyroidism, Williams syndrome, phenylketonuria (PKU), Smith-Lemli-Opitz syndrome, Prader-Willi syndrome, Phelan-Mc Dermid syndrome, Mowat-Wilson syndrome, ciliopathy, Lowe syndrome and siderium type X-linked mental retardation. Down syndrome is a disorder that includes a combination of birth defects, including some degree of mental retardation, characteristic facial features and, often, heart defects, increased infections, problems with
vision and hearing, and other health problems. Fragile X syndrome is a prévalent form of inherited mental retardation, occurring with a frequency of ! in 4,000 males and 1 in 8,000 females. The syndrome is also characterized by developmental delay, hyperactivity, attention déficit disorder, and autistic-like behavior. There is no effective treatment for fragile X syndrome.
[0243] The présent invention contemplâtes the treatment of mild mental retardation, moderate mental retardation, severe mental retardation, profound mental retardation, and mental retardation severity unspecified. Such mental retardation may be, but is not required to be, associated with chromosomal changes, (for example Down Syndrome due 10 to trisomy 21), heredity, pregnancy and périnatal problems, and other severe mental disorders. This invention provides methods and compositions for treating mental retardation using a a5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of mental retardation. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with mental retardation. In certain embodiments, the symptom to be treated is cognitive defïcit/impairment For example, methods and compositions of the disclosure can be used to improve the motor/cognitive impairments symptomatic of mental retardation. In some embodiments ofthe invention, there îs provided a method of preserving or improving cognitive function in a subject with mental retardation, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
[0244] Several animal models hâve been developed for mental retardation. For example, a knockout mouse model has been developed for Fragile X syndrome. Fragile X syndrome îs a common form of mental retardation caused by the absence of the FMRI protein, FMRP. Two homologs of FMRP hâve been identified, FXR1P and FXR2P.
FXR2P shows high expression in brain and testis, like FMRP. Both Fxr2 and Fmrl knockout mice, and Fmrl/Fxr2 double knockout mice are believed to be useful models for mental retardation such as Fragile X syndrome. See, Bontekoe C. J. M. et al. Hum.
169
Mol. Genet. 2002, 11 (5): 487-498. The efficacy ofthe methods and compositions ofthis invention in treating mental retardation, or cognitive deficit/impairment associated with mental retardation, may be assessed in the these mouse models and other animal models deveioped for mental retardation, as well as human subjects with mental retardation, using a variety of cognitive tests known in the art, as discussed herein.
Compulsive behavior (obsessive-compulsive disorder) [0245] Obsessive compulsive disorder (OCD) is a mental condition that is most commonly characterized by intrusive, répétitive unwanted thoughts (obsessions) resulting in compulsive behaviors and mental acts that an individual feels driven to perform (compulsion). Current epidemiological data indicates that OCD is the fourth most common mental disorder in the United States. Some studies suggest the prevalence of OCD is between one and three percent, although the prevalence of cl inically recognïzed OCD is much lower, suggesting that many indîviduals with the disorder may not be diagnosed. Patients with OCD are often diagnosed by a psychologist, psychiatrist, or psychoanalyst according to the Diagnostic and Statîstîcal Manual of Mental Disorders, 4th édition text révision (DSM-IV-TR) (2000) diagnostic criteria that include characteristicsofobsessionsand compulsions. Characteristicsofobsession include: (!) récurrent and persistent thoughts, impulses, or images that are experienced as intrusive and that cause marked anxiety or dîstress; (2) the thoughts, impulses, or images are not simply excessive worries about real-life problems; and (3) the person attempts to ignore or suppress such thoughts, impulses, or images, or to neutralîze them with some other thought or action. The person recognizes that the obsessîonal thoughts, impulses, or images are a product of his or her own mind, and are not based în reality. Characteristics of compulsion include: (i) répétitive behaviors or mental acts that the person feels driven to perform in response to an obsession, or according to rules that must be applied rigidly;
(2) the behaviors or mental acts are aimed at preventing or reducing dîstress or preventing some dreaded event or situation; however, these behaviors or mental acts are not actually connected to the issue, or they are excessive.
[0246] Indîviduals with OCD typically perform tasks (or compulsion) to seek relief from obsession-related anxiety. Répétitive behaviors such as handwashing, countïng, checking, or cleaning are often performed with the hope of preventing obsessive thoughts or making them go away. Performîng these rituals, however, only provides temporary
170 relief. People with OCD may also be diagnosed with a spectrum of other mental disorders, such as generalized anxiety disorder, anorexia nervosa, panic attack, or schizophrenia.
[0247] The dysfunction in neuronal communication is considered one of the underlying 5 causes for obsession disorder (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr.
Neurol. Neurosci. Rep. 2010, 10, 207-214). Studies suggest that OCD may be related to abnormal levels of a neurotransmitter called serotonin. The first-lîne treatment ofOCD consists of behavioral therapy, cognitive therapy, and médications. Médications for treatment include serotonin reuptake inhibitors (SRIs) such as paroxetine (Seroxat™,
Paxil®, Xetanor™, ParoMerck™, Rexetin™), sertraline (Zoloft®, Stimuloton™), fluoxetine (Prozac®, Bîoxetin™), escitalopram (Lexapro®), and fluvoxamine (Luvox®) as well as the tricyclic antidepressants, în particular clomipramîne (Anafranil®). Benzodiazépines are also used in treatment. As much as 40 to 60% of the patients, however, fait to adequately respond to the SRI therapy and an even greater proportion of patients fail to expérience complété remission of their symptoms.
[0248] The invention provides methods and compositions for treating OCD using a a5contaîning GABAa receptor agonist (e.g., a a5-containîng GABAa receptor positive allosteric modulator), such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression ofOCD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with OCD. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive déficit. For example, methods and compositions of the disclosure can be used to treat the cognitive déficits in OCD, and/or to improve cognitive function in patients with OCD. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with OCD, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or 30 combination thereof.
[0249] A quinpiroîe-sensitized rat mode! has been developed for OCD. The compulsive checking behavior ofthe quinpiroîe-sensitized rats is subject to interruption, which is an
171 attribute characteristic of OCD compulsions. In addition, a schedule-induced polydipsia (SIP) rodent model of obsessîve-compulsive disorder was used to evaluate the effects of the novel 5-HT2C receptor agonist WAY-163909. See, e.g., Rosenzweig-Lipson S. et al. Psychopharmacôlogy (Berl) 2007, 192,159-70. The efficacy of the methods and compositions of this invention in treating OCD, or cognitive impairment or cognitive déficits associated with OCD, may be assessed in the above animal models and other animal models developed for OCD, as well as human subjects with OCD, using a variety of cognitive tests known in the art, as discussed herein.
I
Substance addiction [0250] Substance addiction (e.g., drug substance addiction, alcohol substance addiction) is a mental disorder. The substance addiction is not triggered înstantaneously upon exposure to substance of abuse. Rather, it involves multiple, complex neural adaptations that develop with different time courses ranging from hours to days to months (Kauer J. A. Nat. Rev. Neurosci. 2007,8,844-858). The path to substance addiction generally ! 5 begins with the voluntary use of one or more controlled substances, such as narcotîcs, barbiturates, methamphetamînes, alcohol, nicotine, and any of a variety of other such controlled substances. Over time, with extended use ofthe controlled substance(s), the voluntary ability to abstain from the controlled substance(s) is compromised due to the effects of prolonged use on brain function, and thus on behavior. As such, substance addiction generally is characterized by compulsive substance craving, seeking and use that persist even in the face of négative conséquences. The cravings may represent changes în the und^rlying neurobiology ofthe patient which likely must be addressed in a meaningful way if recovery is to be obtained. Substance addiction is also characterized in many cases by withdrawal symptoms, which for some substances are life threatening (e.g., alcohol, barbiturates) and in others can resuit in substantial morbidity (which may include nausea, vomiting, fever, dizziness, and profuse sweating), distress, and decreased ability to obtain recovery. For example, alcoholism, also known as alcohol dependence, îs one such substance addiction. Alcoholism is primarily characterized by four symptoms, which include cravings, loss of control, physical dependence and tolérance. These symptoms also may characterize substance addictions to other controlled substances. The craving for alcohol, as well as other controlled substances, often is as strong as the need for food or water. Thus, an alcoholic may continue to drink despîte serîous family, health
172 and/or legal ramifications.
[0251] Recent work exploring the effects of abusing alcohol, central stimulants, and opiates on the central nervous system (CNS) hâve demonstrated a variety of adverse effects related to mental health, including substance-induced impairments in cognition.
See, Nyberg F. Cognitive Impairments in Drug Addicts, Chapter 9. In several laboratories and clintcs substantial damages of brain function are seen to resuit from these drugs. Among the harmful effects of the abusing drugs on brain are those contributing to accelerated obsolescence. An observation that has received spécial attention during recent years is that chronic drug users display pronounced impairment in brain areas associated with executive and memory function. A remarked neuroadaptation caused by addictive drugs, such as alcohol, central stimulants and opiates involves dimtnished neurogenesis în the subgranular zone (SGZ) ofthe hippocampus. Indeed, it has been proposed that decreased adult neurogenesis in the SGZ could modify the hippocampal function in such a way that it contributes to relapse and a maintained addictive behavior. It also raises the possibility that decreased neurogenesis may contribute to cognitive déficits elicîted by these abusing drugs.
[0252] The invention provides methods and compositions for treating substance addiction using aa5-containing GABAa receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvatés, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or siowing the progression of substance addiction. In certain embodiments, treatment comprises alleviation, amelioration, or siowing the progression ofone or more symptoms associated with substance addiction. In certain embodiments, the symptom to be treated is cognitive impairment. For example, methods and compositions of the disclosure can be used to treat the cognitive impairment and/or to improve cognitive function in patients with substance addiction. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with substance addiction, the method comprising the step of administering to said subject a therapeutically effective amount of a compound ofthe invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
φ 173 [0253] Several animal models hâve been developed to study substance addiction. For example, a genetically selected Marchigïan Sardinian alcohol-preferring (msP) rat models was developed to study the neurobiology of alcoholism. See, Ciccocioppo R. et al. Substance addiction Biology 2006,11,339-355. The efficacy of the methods and compositions of this invention in treating substance addiction, or cognitive impairment associated with substance addiction, may also be assessed ln animal models of substance addiction, as well as human subjects with substance addiction, using a variety of cognitive tests known in the art, as discussed herein.
Research Domain Criteria (RDoC) [0254] The invention further provides methods and compositions for treating impairment in neurological disorders and neuropsychiatrie conditions using a a5containing GABAa R positive allosteric modulator or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof as described herein. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with such impairment. In another aspect ofthe invention, there is provided methods and compositions for preserving or improving cognitive function in a subject in need thereof using a compound of the invention or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
[0255] Research Domain Criteria (RDoC) are expected to augment clinical criteria, such as DSM and 1CD, for diagnosis of disease and disorders affecting the nervous system (see, e.g., Am. J. Psychiatry 167:7 (2010)). The RDoC is intended to provide classification based on discoveries in genomics and neuroscience as well as clinical observation. The high expression of a5-containing GABAa receptors in spécifie neural circuits in the nervous system could be therapeutic targets for neural circuit dysfunction identified under RDoC.
Assays for GABAa uS subunlt binding and receptor positive allosteric modulator activity [0256] The affinity of test compounds for a GABAa receptor comprising the GABAa a5 30 subunit may be determined using receptor binding assays that are known in the art. See,
174
e.g., U.S. Patent 7,642,267 and U.S. Patent 6,743,789, which are incorporated herein by reference.
[0257] The activity of the test compounds as a a5-containing GABAa R positive allosteric modulator may be tested by electrophysiological methods known in the art. See, e.g., U.S. Patent 7,642,267 and Guidotti étal., Psychopharmacology 180:191-205, 2005, Positive allosteric modulator activity may be tested, for examples, by assaying GABA-induced chloride ion conductance of GABAa receptors comprising the GABAa a5 subunit. Cells expressing such receptors may be exposed to an effective amount of a compound of the invention. Such cells may be contacted in vivo with compounds of the invention through contact with a body fluid containing the compound, for example through contact with cerebrospinal fluid. In vitro tests may be done by contacting cells with a compound of the invention in the presence of GABA. Increased GABA-induced chloride conductance în cells expressing GABAa receptors comprising the GABAa ct5 subunit in the presence ofthe test compound would indicate positive allosteric modulator activity of said compound. Such changes in conductance may be detected by, e.g., using a voltage-clamp assay performed on Xenopus oocytes injected with GABAa receptor subunit mRNA (including GABAa «5 subunit RNA), HEK 293 cells transfected with plasmîds encoding GABAa receptor subunits, or in vivo, ex vivo, or cultured neurons.
[0258] It will be understood by one of ordinary skill în the art that the methods described herein may be adapted and modified as is appropriate for the application being addressed and that the methods described herein may be employed in other suitable applications, and that such other additions and modifications will not départ from the scope hereof.
[0259] · Thîs invention will be better understood from the Examples which follow. However, one skilled în the art will readily appreciate that the spécifie methods and results discussed are merely illustrative ofthe invention as described more fully în the embodiments which follow thereafter.
175
Example 1: Synthesis of Compound 1
Scheme 11.
NiNOj
HCl (cône ) HjO
R’ - OMe, H. F
R1 -OMe U pTfOH-H/J p-xylene 140 ’C ~
COjEI
R1-0M· 14
H,, Pd/C
ÊÔH 1
R1«0Me15
ElgN, EIOH
1X4-tritt0ie
PQCh, FPraNEt CHjCN '
R1 OMe
It
R1 OMe
R1 OMe-20
CN^COjEI
KO-F0U. DMF
t
I
R1 F. compound 110
R1 “ OCHj- compound 1*7
OMe compound 1
F: compound 2
H-compound 3 · {0260] To a stirred mixture of 5-methoxy-2-nttroani!ine (5g, 29.7 mmol) in HCl (conc. 39 mL) at 0°C was added drop wise a solution ofNaNCh (2.05 g, 29.7 mmol) in H2O (19 mL). The internai température was kept below IO°C. After addition, the mixture was stirred at room température for I h. The diazonium sait was collected by filtration, and was used in the next step. To the diazonium sait in a crystallization dish under fast stirring at room température was added drop wise a solution ofNaNj (1.93 g, 29.6 mmol) in H2O (7 mL). After gas évolution stopped (3 h), it was filtered. The collected solid was re-crystallized from MeOH to give 4.342 g (yield 75% for 2 steps) of the product 13 as a !
yellow solid. To a mixture ofthe phenylazide 13 (1.94 g, 10 mmol) and diethyl 1,318506 φ 176 acetone-diacrboxylate (2.20 mL, 12 mmol) in EtOH (40 mL) at room température was added EtjN (1.67 mL, 12 mmol). After the mixture was stirred at room température for 60 h, the initial suspension tumed into a clear yellow solution. The solution was concentrated under vacuum and the residue was purified by chromatography (RediSep 24 g silica-gel column, 10% to 40% EtOAc în hexanes) to give 2.905 g of triazole 14 as a yellow solid. MS: [M+1] = 379.
[0261] The above triazole 14 (2.95 g, 7.66 mmol) în EtOH (50 mL) with Pd/C (10 wt%, 407 mg, 0.38 mmol) was stirred under H2 (balloon) for 24 h. It was filtered through Celite. The filtrate was concentrated and the residue was purified by chromatography (RediSep 24 g silica-gel column, 10% to 50% EtOAc în hexanes) to give 2.453g of aniline 15 as a white solid. (70% yield for two steps.) MS: [M+1] = 349.
[0262] Compound 15 (2.45 g, 7.03 mmol) and catalytic amount ofp-TsOH· H2O (24 mg) in p-xylene (30 mL) were heated in a 140°C oil bath ovemight. The mixture was cooled and filtered. The solid was washed with cold EtOAc. After drying, it gave 1.88 g 15 (88% yield) ofthe lactam 16. MS: [M+1] = 303.
[0263] To a suspension ofthe lactam ester 16 (837 mg, 2.77 mmol) in THF (20 mL) at room température was add LiBHi (2 M in THF, 1.39 mL, 2.78 mmol). After the mixture was stirred at room température for 60 h, more L1BH4 (2 M în THF, 0.28 mL, 0.56 mmol) was added and it was stirred at room température for 24 additional h. A mixture of
EtOAc/EtOH ( 10 mL/10 mL) was added to the reaction and it was concentrated in vacuo. The residue was taken up in EtOAc/CH2CI2/MeOH and loose silica gel was added. After volatile solvents were evaporated, the solid was loaded onto a RediSep 24 g silica-gel • column. Chromatography (solvent A: EtOAc, solvent B: 10:1 v/v CH2CI2/MeOH;
gradient eluent: A to B) gave 540 mg (75% yield) ofthe alcohol 17 as white solid. MS:
[M+1] = 261.
[0264] To a solution ofthe alcohol 17 (105.4 mg, 0.40 mmol) and CBr4 (336 mg, LOI mmol) in DMF (3 mL) was slowly added a solution of PPhj (255 mg, 0.97 mmol) in
DMF (I mL) over 20 min. After addition, TLC showed the reaction went completion.
Water was added to quench the reaction and the mixture was extracted with EtOAc thrice.
The combined extracts were washed sequentially with H2O, brine and dried over Na2SO4.
177
Filtration and concentration gave the crude product. Chromatography (RediSep 12 g silica-gel column, CH2C12 to 30% EtOAc in CH2CI2) gave 439.2 mg of a mixture of the bromide 18 ([M+1 ] = 324) and Ph3PO. The above mixture (439 mg) în EtOAc/EtOH (8 mL/8 mL) with Pd/C (10 wt%, 200 mg, 0.19 mmol) was stirred under H2 (balloon) for 2 h, then was filtered through Celite. The filtrate was concentrated and residue was purified by chromatography (RediSep 12 g silica-gel column, solvent A: 1:1 v/v CH2Cl2/hexanes, solvent B: EtOAc; gradient eluent: A to B) to give 99 mg (-80% yield for 2 steps) of product 19 as a white solid. MS: [M+1] = 245.
[0265] In a separate flask, 1,2,3-triazole (55.3 mg, 0.80 mmol) ïn CH3CN (1 mL) at 0°C 10 was treated with f-Pr2NEt (146 pL, 0.84 mmol), followed by POCI3 (23 pL, 0.25 mmol).
The solution was stirred at 0°C for 2 h. The lactam 19 was added in one lot and the resulting suspension was heated în an 80°C oil bath for 20 h. Water was added to quench the réaction. It was extracted wîth EtOAc thrice. The combined extracts were washed wîth brine and dried over Na2SO4. Filtration and concentration gave 48.8 mg of the crude product 20, which was used directly in the next step. A solution of KO- /-Bu (37.2 mg,
0.33 mmol) in DMF (0.5 mL) was cooled to -50°C. Ethyl isocyanoacetate (40 pL, 0.36 mmol) was added drop wise. The mixture was stirred at -50°C for 1 h. The above crude product 20 in DMF (1 mL) was added drop wise. The mixture was allowed to warm to 10°C and stirred at 10°C for 1 h. Saturated NH4CI aqueous solution was added and it was 20 extracted with EtOAc thrice. The combined extracts were washed sequentîally with water, brine and dried over Na2SO4. Filtration and concentration gave the crude product.
[0266] Chromatography (RediSep 12 g silica-gel column, solvent A: 1:1 v/v CH2C12/hexanes, solvent B: EtOAc; gradient eluent: 20% to 80% B in A) to give 15 mg (21% yield for 2 steps) of Compound 1 (Example 1) as an off-whîte solid. MS: [M+1] = 25 340. ’H-NMR(500MHz, CDC13) δ: 7.74 (s,IH), 7.63 (d, IH, J=3Hz), 7.51 (d, IH,
J=8.5Hz), 7.14 (dd, IH, J=3.Q, 8.5Hz), 4.44 (q, 2H, J=7.0Hz). 3.95 (s, 3H), 2.44 (s, 3H), 1.45 (t, 3H, J=7.0Hz).
178
Example 2: Synthesis of Compound 2:
[0267| Compound of Example 2 was synthesized în an analogous synthetic route as that described for Example I, using 5-fluoro-2-nitro-anilîne as the starting material to give Compound 2 as a light brown solid: MS: [M+1] = 328. *H-NMR (500MHz, CDCI3) δ: 7.90 (brdd, 1H, J=2.5,8.5Hz), 7.77 (s, 1H), 7.62 (br dd, 1H, J=5.0,9.0Hz), 7.35 (m, I H), 4.45 (q, 2H, J=7.0Hz), 2.45 (s, 3H), 1.45 (t, 3H, J=7.0Hz).
Example 3: Synthesis of Compound 3:
[0268] Compound of Example 3 was synthesized in an analogous synthetic route as that described for Example 1, using 2-nitro-aniline as the starting material to give Compound 3 as a light yellow solid: MS: [M+1] = 310; *H-NMR (500MHz, CDCI3) δ: 8.161 (br d, 1H, J=8.5Hz), 7.81 (s, 1H), 7.66 (m, 3H), 4.45 (q, 2H, J=7.0Hz), 2.45 (s, 3H), 1.46 (t, 3H, J=7.0Hz).
Example 4: Synthesis of Compound 110
[0269] Acetamïde oxîme was azeotroped three times in toluene before use. To a suspension of acetamïde oxime (30 mg, 0.4 mmol) in THF (1 mL) was added NaH 60% in oil dispersion (16 mg, 0.4 mmol). The suspension was stirred at room température for 15 min. The ester compound 2 (65 mg, 0.2 mmol) was added. The vial containing the ester was rinsed with THF ( 1 mL) which was added to the reaction mixture. The resulting brown suspension was stirred at room température for 30 mins. then heated at 70 °C for 2h 30 min. The suspension was quenched with MeOH. The solvent was evaporated and the crude oil was purified by chromatography (RediSep 4 g silica-gel column, eluted with 70% EtOAc în Hexanes) to give 28 mg (41% yield) of product. MS: [M+1] = 338.
H’NMR (CDCIj) δ 7.92 (IH, dd, J= 2.5,8.5 Hz), 7.90 (IH, s), 7.67 (IH, dd, J= 4.5,9.5 Hz), 7.38 (IH, m), 2.51 (3H, s), 2.46 (3H, s).
Example 5: Synthesis of Compound 167
[0270] The compound was prepared analogously from Compound 1 to give
Compound 167: MS: [M+1] = 350. H'NMR (CDCIj) δ 7.87 (IH, s), 7.65 (IH, d, J= 3
Hz), 7.55 (IH, d, J= 9 Hz), 7.17 (IH, dd, J= 2.5,9 Hz), 3.96 (3H, s), 2.5 (3H, s), 2.45 20 (3H, s).
Scheme 12.
180
Ν’·ΟΜ·17 Κ'·ΟΜ·Ι1
0 y CN^COjEI - KQ-t-Bu DMF -eOXtolO’C ^R,
R’>OM>14 /
R1 · ΟΜ·, R* · Η tonpoun* 4 R’ · H. R1- H ««npound I R’ · f, R1 · H nmpeund 4
R' · F. R’ - 4-CHj <<xnpouM 44 R1 F. R’ - 4-Ct eempeund 44
R1 · F. R* · 4-F lompound 44 R1 · H. R’· «-Ct «ompand 47
R' - F, R* · H «ampaind 14*
Example 6: Synthesis of Compound 4:
(02711 To a solution of compound 17 prepared as in Exemple 1 (260 mg) in DMSO (4 mL) and CH2C12 (6 mL) was added Et3N (0.7 mL, 5 mmol), followed by Py· SO3 (398 mg, 2.5 mmol). it was stirred at room température for i h. The reaction mixture was poured into water and extracted with EtOAc thrice. The combined extracts were washed sequentially with H2O, brine and dried over Na2SO4. Filtration and concentration gave
198.5 mg of the crude aldéhyde 21, which was used without further purification. To a suspension of aldéhyde 21 (198.5 mg, 0.77 mmol) in THF (10 mL) at 0°C was added drop wise PhMgBr (I M in THF, 1.54 mL, 1.54 mmol). It was stirred at 0°C for 30 min. Saturated NH4CI aqueous solution was added and it was extracted with EtOAc thrice.
[0272] The combined extracts were washed with brine and dried over Na2SO4. Filtration and concentration gave 252.9 mg of the benzyi alcohol 22 as a brown foamy solid. This was used in the next step without further purification. To a solution of the above crude alcohol 22 in CH2CI2 (8 mL) with Et3SÎH (0.60 mL, 3.76 mmol) was added TFA (0.64 mL, 8.27 mmol). The reaction solution was stirred at room température for 4 h. After
181 concentration, the residue was purified by chromatography (RediSep 12 g silica-gel column, 20% to 80% EtOAc in hexanes) to give 34.1 mg (yield 12% for four steps) ofthe reduced product 23 as white foamy solid. MS: [M+1] = 321.
[0273] ln a separate flask, a solution of 1,2,4-triazole (27 mg, 0.39 mmol) in CH3CN (0.5 mL) at 0°C was treated with /-Pr2NEt (72 pL, 0.41 mmol), followed by POClj (11 pL, 0.12 mmol). The mixture was stirred at 0°C for 2 h. The lactam material 23 (32.2 mg, 0.1 mmol, solid) was added in one lot to the reaction mixture and ït was heated in an 80°C oil bath for 20 h. The mixture was cooled to room temperature and creamy solid precipitate was observed. Water (0.5 mL) was added and it was stirred at room temperature for 5 min. The solid precipitate was collected by filtration, and washed with 0.5 mL of water, followed by drying under high vacuum to give 15.8 mg (yield 42%) of the adduct 24 as a off-white flufïy solid. MS: [M+1] = 372. A solution of KO-Z-Bu (9.5 mg, 85 pmol) in DMF (0.5 mL) was cooled to -50°C. Ethyl îsocyanoacetate (10.4 pL, 95 pmol) was added drop wise. The resulting mixture was stirred at -50°C for I h. The trîazole amidine 24 (15.8 mg, 42 pmol, solid) was added în one lot. The stirred mixture was allowed to warm up to 10°C in 1 h and kept at 10°C for 1 h. Saturated NH4CI aqueous solution was added and it was extracted with EtOAc thrice. The combined extracts were washed sequentially with H2O, brine and dried over Na2SÜ4. Filtration and concentration gave the crude product. Chromatography (RediSep 4 g silica-gel column.
Solvent A: 1:1 v/v CH2Cl2/hexanes, solvent B: EtOAc; gradient eluent: A to 50% B în A) gave 16.8 mg (yield 95%) of the compound of Example 6 as a white solid. MS: [M+1] = 416. 'H-NMR(500MHz, CDC13) δ: 7.74 (s,lH), 7.63 (d, 1H, J=3.0Hz), 7.50 (d, 1H, J= 9.0Hz), 7.30 (br d, 2H, J=7.0Hz), 7.29 (br d, 2H, 7.5Hz), 7.20 (m, 1H), 7.13 (dd, 1H, J=2.5, 9.0Hz), 4.41 (q, 2H, J=7.5Hz), 4.17 (s, 2H), 3.95 (s, 3H), 1.43 (t, 3H, 7.5Hz).
Example 7: Synthesis of Compound 5:
182
[0274] Compound of Example 7 was synthesized in an analogous synthetic route as that described for Example 6, using 2-nitro-anilîne as the starting material to give Compound 5 as a brown solid: MS: [M+l] = 386. lH-NMR (500MHz, CDCI3) δ: 8.16 (br d, 1 H, J=7.0Hz), 7.81 (s, IH), 7.60-7.68 (m, 3H), 7.34 (br d, 2H, J=8.0Hz), 7.29 (br d,
2H, J=7.0Hz), 7.20 (m, ! H), 4.42 (q, 2H, J=7.0Hz), 4.18 (s, 2H), 1.44 (t, 3H, J=7.0Hz).
Example 8: Synthesis of Compound 6:
[0275] Compound of Example 8 was synthesized in an analogous synthetic route as that described for Example 6, using 5-fluoro-2-nitro-ani!ine as the starting material to give compound 8 as a brown solid: MS: [M+1] = 404. lH-NMR (500MHz, CDCI3) δ: 7.90 (dd, IH, J=3.5, 8.5Hz), 7.77 (s, IH), 7.61 (dd, IH, J=5.0,10.5Hz), 7.28-7.37 (m,
5H), 7.21 (m, 1H), 4.43 (q, 2H, J=7.0Hz), 4.17 (s, 2H), 1.44 (t, 3H, J=7.0Hz).
Example 9: Synthesis of Compound 44:
[0276] Compound of Example 9 was synthesized in an analogous synthetic route as that described for Example 6, using 5-fluoro-2-nitro-aniline as the starting material to give the compound of Example 9 as a brownish solid: MS: [M+1] = 418. 'H-NMR (500MHz, CDC13) δ: 7.89 (br d, IH, J=9.5Hz), 7.76 (s, IH), 7.60 (dd, IH, J=5.5,
10.0Hz), 7.35 (br t, IH, J=6.0Hz), 7.22 (br d. 2H, J=8.5Hz), 7.09 (br d, 2H, J=7.5Hz),
183
4.43 (q, 2H, J=7.5Hz), 4.12 (s, 2H), 2.30 (s, 3H), 1.44 (t, 3H, J=7.5Hz).
Example 10: Synthesis of Compound 45:
[0277] Compound of Example 10 was synthesized in an analogous synthetic route as that described for Example 6, using 5-fluoro-2-nitro-aniline as the starting material to give the compound of Example 10 as a brownish solid: MS: [M+1] = 438. ’H-NMR (500MHz, CDCH) 6: 7.90 (dd, 1H, J=3.0, 8.0Hz), 7.77 (s, 1H), 7.61 (dd, 1H, J=5.0,
9.0Hz), 7.36 (m, I H), 7.25 (br s, 4H), 4.42 (q, 2H, J=7.0Hz), 4.14 (s, 2H), 1.44 (t, 3H,
J=7.0Hz).
Example 11: Synthesis of Compound 46:
[0278] Compound of Example 11 was synthesized in an analogous synthetic route as that described for Example 6, using 5-fluoro-2-nitro-aniline as the starting material to give the compound of Example 11 as a yellowish solid: MS: [M+1] = 422. ’H-NMR (500MHz, CDCH) δ: 7.90 (dd, IH, J=3.0, 8.5Hz), 7.77 (s, 1H), 7.61 (dd, 1H, J=5.0,
9.0Hz), 7.36 (m, 1 H), 7.28 (m, 2H), 6.96 (m, 2H), 4.42 (q, 2H, J=7.5Hz), 4.14 (s, 2H),
1.44 (t, 3H, J=7.0Hz).
Example 12: Synthesis of Compound 47:
184
[0279] Compound of Example 12 was synthesized în an analogous synthetic route as that described for Example 6, using 2-nitro-aniIine as the starting material to give the compound of Example 12 as a yellowish solid: MS: [M+1 ] = 420. ’H-NMR (500 MHz, CDCB) δ: 8.16 (br d, I H, J=7.0 Hz), 7.80 (s, 1 H), 7.64 (m, 3H), 7.25 (m, 4H), 4.41 (q, 2H, J=7.0 Hz), 4.14 (s, 2H), 1.44 (t, 3H, J=8.0 Hz).
Example 13: Synthesis of Compound 109:
109 [0280] Acetamide oxime (50 mg, 0.67 mmol) was azeotroped with toluene 3 times. THF (5mL) was added, then NaH 60% în oil dispersion (25 mg, 0.62 mmol). The suspension was stirred at room température for 30 min. 2 mL of this suspension was added to ester compound 6 (40 mg, 0.099 mmol) and the resulting solution was heated at 70 °C for 3h. The solution was quenched with water. The solution was extracted with EtOAc (3x). The combined organic phases were washed with brine, dried over MgSO-i. Filtration and concentration gave the crude product. Chromatography (RediSep 12 g silica-gel column. Eluted with 50% EtOAc in Hexanes) gave 6 mg (yield 20%) ofthe product Compound 109 as yellow solid. MS: [M+1] =414). H’NMRÎCDCLj) δ 7.93 (1H, dd, J= 3,8.5 Hz), 7.89 (ΙΗ, s), 7.65 (lH, dd, J= 5.5,9 Hz), 7.38 ( 1 H, m), 7.23 (5Η, m), 4.2 (2H, s), 2.50 (3 H, s).
185
Exampie 14: Synthesis of Compound 7:
[0281] To a stirred mixture of 5-methoxy-2-nitroaniline (5g, 29.7 mmol) in HCl (conc.
12.9 mL) at 0°C was added drop wise a solution of NaNO2 (2.05 g, 29.7 mmol) in H2O (8 mL). The internai température was kept below 5°C. After addition, the mixture was allowed to warm up to room température in 1 h. The mixture was cooled to 0°C and a solution of SnCl2*2H2O (20.13 g, 89.2 mmol) in HCl (conc. 13 mL) was added slowly dropwise. After addition, it was stirred at room température for 2 h. The resulting yellow solid was collected by filtration and washed with cold (0°C) 6 N HCl. After drying in vacuum oven, it gave 3245 g (yield 50%) of brown solid as aryl hydrazine 25. MS: [M+H2O+Na] = 224. In a separate flask, a mixture of diethyl 1,3-acetonedîacrboxylate (2.426 g, 12 mmol) and diethoxymethyl acetate (1.946 g, 12 mmol) was heated under microwave radiation at 100°C for 1 h. The reaction mixture was concentrated in vacuo, and residual volatile component was co-distilled off with toluene (5ml) in vacuo to give condensation product 26, which was used directly in the next step.
186
Scheme 13.
NaN02
HCl (cône )
H20
EtQ O
SnCI?
HCl. H20
(-50% for 2 steps) 25
EtOjC^^A^COjEt
100‘C, 1h neat, mlcrowave no2 nhnh2«hck
MS (4À) _ EtOH, rt
Hj, Pd/C ËtOH '
(24% for 3 steps)
DibakH CH,Cl, 74%
H, Pd/C , EtOAc/EtOH (-78% for 2 steps)
2) CN^xCO2Et KO-t-Bu, DMF
1) 1,2,4-triazole POCh, FPr2NEt CH3CN
[0282] Product 26 from above was dissolved in EtOH (30 mL). Molecular sieves (4 A, 5 2 g) and hydrazine hydrochloride 25 (2.19 g, 10 mmol) were added. The suspension was stirred at room température for 24 h. It was filtered through Celite and the solid was washed with EtOAc (10 mLX 3). The filtrate was concentrated. The residue was purified by chromatography (RediSep 40 g silica-gel column, 10% to 40% EtOAc in hexanes) to give 2.091 g of pyrrole 27 which was used without further purification in the 10 next step. MS: [M+1] = 378.
[0283] The above nitro group on 27 (2.09 g, 5.5 mmoi) was reduced in EtOH (40 mL) with Pd/C (10 wt%, 295 mg, 0.28 mmol) under H2 (balloon) for 18 h. The mixture was filtered through Celite. The filtrate was concentrated and the residue was purified by
187 chromatography (RediSep 24 g silica-gel column. hexanes to 50% EtOAc in hexanes) to give 1.127g ofthe un-cyclized product 28 as a yellow stîcky oil ([M+1] = 348), plus 154 mg of cyclized product 29 as a gray solid (MS: [M+1] = 302). The un-cyclized aniline 28 (1.127 g, 3.2 mmol) in p-xylene (20 mL) was treated with catalytic amount ofp-TsOH· H2O (15 mg) în a 140°C oil bath for 20 h. The réaction mixture was cooled, concentrated, and the residue was triturated with cold (0°C) EtOAc. Filtration gave 559 mg of the lactam product 29 as a yellow solid. The total weight of the lactam product 29 combined is 713 mg (24% for 3 steps). MS: [M+1] = 302.
[0284] To a suspension ofthe ester 29 (566 mg, 1.88 mmol) in CH2C12 (35 mL) at -78°C was added Dibal-H (1 M in hexane, 6.60 mL, 6.60 mmol). The suspension was stirred for 10 min at -78°C. The cold bath was removed and it was stirred for 20 min while the température rose to room température. At this point, TLC showed -80% reaction completion. It was cooled to -78°C and more Dibal-H (1 M in hexane, 1.0 mL, 1.0 mmol) was added. After stirring at -78°C for 30 min, LCMS showed the reaction proceeded to completion. The reaction was quenched by addition of Rochelle’s sait aqueous solution (20%) followed by EtOAc. It was vigorously stirred at room température until it became a clear two-layer mixture. The layers were separated and the aqueous layer was extracted with EtOAc thrîce. The combined organic phase was washed with brine and dried over Na2SO4. Filtration and concentration gave 480 mg of the crude alcohol 30 as a slightly yellow solid. MS: [M+1] = 260.
[0285] To a solution of alcohol 30 (200 mg, 0.77 mmol) and CBr4 (640 mg, 1.93 mmol) in DMF (8 mL) was added a solution of PPI13 (486 mg, 1.85 mmol) in DMF (2 mL) slowly in 30 min. After addition, it was stirred at room température for 30 min. Water was added to quench the reaction and the mixture was extracted with EtOAc thrice. The combined extracts were washed sequentially with H2O, brine and dried over Na2SO4. Filtration and concentration gave the crude product. Chromatography (RediSep 12 g silica-gel column, solvent A: 1:1 v/v CH2Cl2/hexanes, solvent B: EtOAc; gradient eluent: 10% to 40% B in A) gave 221 mg of a mixture ofthe bromide 31 and PI13PO.
[0286] The above mixture in EtOAc/EtOH (8 mL/8 mL) with Pd/C (10 wt%, 200 mg, 0.19 mmol) was stirred under H2 (balloon) for 1 h. It was filtered through Celite. The filtrate was concentrated and residue was purified by chromatography (RediSep 12 g t
188 silica-gel column, solvent A: 1:1 v/v CH2CI2/hexanes, solvent B: EtOAc; gradient eluent: 10% to 40% B in A) to give 146 mg of a mixture of the réduction product 32 ([M+1] = 244) and Ph3PO.
[0287] ln a separate flask, 1,2,4-triazole (81 mg, 1.17 mmol) in CH3CN (I mL) at 0°C was treated with /-Pr2NEt (214 pL, 1.23 mmol), followed by POCI3 (34 pL, 0.36 mmol). The solution was stirred at 0°C for 2 h. The lactam 32 (-60% purity by LCMS) was added in one lot and the resulting suspension was heated in an 80°C oil bath for 18 h. Water was added to quench the reaction. It was extracted with EtOAc thrice. The combined extracts were washed sequentially with H2O, brine and dried over Na2SO4. Filtration and concentration gave 126.6 mg of the crude product 33 as a yellow glue, which was used directly in the next reaction. MS: [M+l] = 295. A solution of KO- r-Bu (97 mg, 0.86 mmoi) in DMF (1 mL) was cooled to -50°C. Ethyl isocyanoacetate (104 pL, 0.95 mmol) was added drop wise. The mixture was stirred at -50°C for 1 h. The above crude product 33 in DMF (1.5 mL) was added drop wise. The mixture was allowed to warm to 10°C and stirred at 10°C for 1 h. Saturated NH4CI aqueous solution was added and it was extracted wîth EtOAc thrice. The combined extracts were washed sequentially with water, brine and dried over Na2SO4. Filtration and concentration gave the crude product. Chromatography (RediSep 12 g silica-gel column, solvent A: 1:1 v/v CH2CI2/hexanes, solvent B: EtOAc; gradient eluent: 10% to 40% B in A) to give 22 mg of a white solid, which was further purified by préparative TLC (deveioped with 1:1 AZB) to give 12.8 mg ofthe final product Compound 7 (Example 14) as a white solid. MS: [M+l[ = 339.’H-NMR (500MHz, CDCI3) δ: 7.70 (s, 1 H), 7.56(s, lH),7.50(d, IH, J=3.0Hz), 7.43 (d, IH, J=8.5Hz), 7.00 (dd, IH, J=2.5,9.5Hz), 5.29 (brs, IH), 4.44 (q, 2H, J=7.0Hz), 3.92 (s, 3H), 3.55 (br s, IH), 2.17 (s, 3H), 1.45 (t, 3H, J=7.0Hz).
189
Example 15: Synthesis of Compound 8:
Scheme 14.
[0288] To a solution of the alcohol 30 (261 mg, 1.0 mmol) which was prepared in Example 14 in DMSO (4 mL) and CH2CI2 (6 mL) was added Et2N (0.7 mL, 5 mmol), followed by Py· SO3 (398 mg, 2.5 mmol). It was stirred at room température for 1 h. The reaction mixture was poured into water and extracted with EtOAc thrice. The combined extracts were washed sequentially with H2O, brine and dried over Na2SO.|. Filtration and concentration gave 226 mg of the crude aldéhyde 34 as a yellow solid. It was used in lhe next step without purification. MS: [M+1 ] = 258.
[0289] To a suspension ofthe crude aldéhyde 34 (202 mg, 0.79 mmol) in THF (10 mL) at 0°C was added drop wise PhMgBr (I M in THF, 1.58 mL, 1.58 mmol). It was stirred at 0°C for 30 min. Saturated NH4CI aqueous solulion was added and it was extracted with EtOAc thrice. The combined extracts were washed with brine and dried over
Na2SO4. Filtration and concentration gave 275 mg ofthe crude product 35 as a yellow
foamy solid, which was used in the next step without purification.
[0290] To a solution of the above crude alcohol 35 in CH2CI2 (10 mL) with EtjSiH (0.66 mL, 4.10 mmol) was added TFA (0.70 mL, 9.02 mmol). The reaction solution was stirred at room température for 1 h. After concentration, the residue was purified by chromatography (RediSep 24 g silica-gel column, 10% to 50% EtOAc in hexanes) to give
187.8 mg (yield 59% for three steps) ofthe product 36 as a gray solid. MS: [M+1] = 320.
[0291] In a separate flask, a solution of 1,2,4-triazole (127 mg, 1.83 mmol) in CH3CN (1.6 mL) at 0°C was treated with /-Pr2NEt (336 pL, 1.93 mmol), followed by POCI3 (53 pL, 0.56 mmol). The mixture was stirred at 0°C for 2 h. Lactam 36 (150 mg, 0.47 mmol, solid) was added in one lot to the réaction mixture and it was heated in an 80°C oil bath for 18 h. The mixture was cooled to room température and solid precipitate was observed. Water (2.1 mL) was added and it was stirred at room température for 10 min. Filtration, washing the solid with 2 mL of water, followed by drying under high vacuum gave 118.8 mg (yield 69%) ofthe triazole amidine 37 as an off-white fluffy solid. MS:
[M+1] = 371. A solution of KO-f-Bu (72 mg, 0.64 mmol) in DMF (2 mL) was cooled to 50°C. Ethyl isocyanoacetate (77 pL, 0.71 mol) was added drop wise. The resulting mixture was stirred at -50°C for 1 h. The triazole amidine 37 (118.8 mg, 42 pmol, solid) was added in lot. The stirred mixture was allowed to warm up to 10°C in 1 h and kept at IO°C for 1 h. Saturated NH4CI aqueous solution was added and it was extracted with
EtOAc thrice. The combined extracts were washed sequentially with H2O, brine and dried over Na2SO4. Filtration, concentration, then chromatography (RediSep 12 g silicagel column. solvent A: 1:1 v/v CH2Cl2/hexanes, solvent B: EtOAc; gradient eluent: A to 40% B ïn A) gave 125.1 mg (yield 94%) of Compound 8 as a white solid. MS: [M+1] = 415. 'H-NMR (500 MHz; CDCI3) 5:7.72 (s, IH), 7.54 (s, IH), 7.51 (br s,lH), 7.44 (br d,
IH, J=9.5Hz), 7.29 (br d, 2H, J=7.5Hz), 7.20 (m, 3H), 7,01 (br d, IH, J=7.5Hz), 5.30 (br s, IH),4.38(q,2H,J=7.0Hz),3.92(brs,5H),3.54(brs, IH), 1.41 (t,3H,J=7.0Hz).
Exemple 16: Synthesis of Compound 9:
Scheme 15.
Ri 4-F, X · OCHy’ compound 10
Ri · 3-OCHj. X OCHj compound 11
R, - 2.4-dl-CH,, X OCHj.· compound 12
R, H. X F, compound 107
R, CH,. X F compound 111 [0292] LiOH (l .09 g, 45.5 mmol) was added to a stirring solution of ester 16 (prepared in Exampie 1) (2.75g, 9.10 mmol) in THF (24 mL) and water (20 mL) at room température. MeOH (4mL) was added, and stirring contînued for 2 h at room température at which point LCMS indicated complété consumption ofthe ester. Upon concentration in vacuo, the reaction mixture was acidified to pH 3-4 by adding 2N HCl (20 mL). After min stirring, the reaction mixture was cooled to 0°C, a solid precipitate was collected by filtration, washed with 3-4 ml water, and dried to give 1.59 g (64%) of the
192 corresponding acid 38 as a grayish solid. MS: [M+l] = 275. To acid 38 (1.59 g, 5.8 mmol) suspended and stirred în DCM (30ml) was added EDC (5.6g, 29.2 mmol), benzyl alcohol (2.5 g, 23.2 mmol) and DMAP (3.54 g, 29.2 mmol). After 3 days of stirring at room température, the reaction was concentrated in vacuo. Water (80 mL) was added to the slurry, followed by diethyl ether (40 mL), and the mixture was stirred vigorously for min, at which point the slurry tumed into a precipitate, and was collected by suction filtration. The solid was washed with water and small amount ofdiethyl ether, and dried to give 1.65 g (78%) benzyl ester 39 as a white solid. MS: [M+1] = 365.
[0293] Compound 1,2,4-trîazole (1.22 g, 17.7 mmol) in CH3CN (15 mL) at 0°C was treated with /-Pr2NEt (3.24 mL, 18.6 mmol), followed by POC13 (0.507 mL, 5.44 mmol). The solution was stirred at 0°C for 2 h. Benzyl ester 39 (1.65 g, 4.53 mmol) was added in lot and the resulting suspension was heated în an 80°C oil bath for 18 h. LCMS showed 5-10% starting lactam remained. In a separate flask, 1,2,4-triazole (307 mg, total 4.9 eq) în CHjCN (3.8 mL) was treated with /-Pr2NEt (0.82 mL, total 5.1 eq) and POClj (0.127 ml; total 1.5 eq) at 0°C for 2 h. The resulting clear solution was transferred into the above reaction mixture. After 2 h heating at 80°C, the reaction was cooled to room température, water was added slowly to quench the reaction ( 10 min). Upon cooling în an ice bath, the solids formed were collected by filtration, washed with water (5ml), and dried to give 1.61g (86%) product 40 as a lightly yellow solid. MS: [M+1] =416.
[0294] A solution of KO- /-Bu (0.739 g, 6.59 mmol) in DMF (11 mL) was cooled to 50°C. Ethyl isocyanoacetate (0.8 ! 0 mL, 7.00 mmol) was added drop wise. The mixture was stirred at -50°C for 1 h. The above triazole intermediate 40 (1.61 g, 3.87 mmol) was added. The mixture was stirred at -50°C for 30 min. and slowly warmed to room température over 4-5 h. Saturated NHjCl aqueous solution (10 mL) was added, followed by EtOAc ( 10 mL). The mixture was sonicated to breakup solid chunks, then stirred thoroughly for 30 min. The precipitate was collected by filtration, washed with water, Et2O, and dried to give crude product as a white solid. Filtrate was partitioned between water and EtOAc; aqueous layer was separated and extracted with EtOAc twice; the combined EtOAc layer was washed with brine and dried over MgSOj. Filtration and solvent removal gave a solid residue which was combined with the solid obtained above for chromatographie purification, using RedîSep 24 g silica-gel column and gradient elution with 0.5 to 5% MeOH in DCM, to give 1.78 g (100%) imidazole 41 as a white
193 solid. MS: [M+1] = 460. The benzyl ester 41 (J .78 g, 3.87 mmol) was subjected to hydrogenolyis (hydrogen balloon) in the presence of catalytic amount of 10% Pd on charcoal in a solvent mixture of THF (40 mL), MeOH (20 mL) and EtOAc (20 mL) for 20 h. LCMS showed complété disappearance ofthe starting material. The solid catalyst was removed by filtration over Celite, and rinsed repeatedly with ample amount of 30% MeOH in DCM until almost ail products were recovered (TLC monitor). Filtrate containing the product was concentrated in vacuo to give 1.22 g (85%) ofacid product 42 was obtained as a yellowish solid. MS: [M+1] = 370.
[0295] To the acid 42 (1.22 g, 3.30 mmol) suspended and stirred in THF (25mL) at 0°C was added borane dimethylsulfide complex (2M THF; 19 mL, 38 mmol) dropwise. Ice bath was removed and the réaction mixture was stirred at room température for 16 h. Upon cooling in an ice bath, the réaction was carcfully quenched with MeOH (20 mL), and then stirred at room température ovemight. Solvents were removed in vacuo. MeOH was added and removed in vacuo two more times. ISCO purification (RediSep 24g column) using a gradient of 1 to 8% MeOH in DCM gave 0.625 g (53%) of alcohol product 43 as a white solid. MS: [M+1] = 356.
[0296] Diisopropyl azodicarboxylate (48.3 mg, 0233 mmol) was added drop-wîse into a stirring solution of alcohol 43 (37.5 mg, 0.106 mmol). phénol (14.9 mg, 0.158 mmol), and Ph3P (55.6 mg, 0212 mmol) in anhydrous THF (0.8 mL) at 0°C. Ice bath was removed and stirring continued at room température for 16 h. LCMS showed complété disappearance ofthe starting alcohol. The reaction mixture was partitioned between sat. NaHCO3 and EtOAc. The organic layer was separated and washed with water, brine, and dried over MgSOj. The desired product was isolated from the reaction mixture by two consecutive préparative TLC (4% MeOH in DCM, and hexanes/EtOAc/MeOH ~ 47.5 /
47.5 / 5, v/v/v) to give 5.3mg (12%) of product which is Compound 9 as a white solid.
MS: [M+1] = 432. ’H-NMR (500 MHz, CDC13) δ: 7.77 (s, IH), 7.63 (d, IH, J=3.5 Hz), 7.53 (d, IH, J=9,0 Hz), 7.31 (m, 2H), 7.17 (dd, IH, J=3.0, 8.5 Hz), 7.08 (d, 2H, J=7.0 Hz), 6.99 (t, IH, J=6.5 Hz), 5.30 (s, 2H), 4.40 (q. 2H, J=7.0 Hz), 3.96 (s, 3H), 1.38 (t, 3H, J=7.0 Hz).
194
Exampie 17: Synthesis of Compound 10:
F [0297] Compound of Example 17 was synthesized in an analogous synthetic route as that described for Example 16, using 4-fluoro-phenol in the ultimate step to give Compound 10 (4.9 mg) as a white solid: MS: [M+1] = 450. 'H-NMR (500 MHz, CDC13) δ: 7.76 (s, 1H), 7.64 (d, 1H, J=3.5 Hz), 7.53 (d, 1H, J=8.0 Hz). 7.17 (dd, 1H, J=2.5, 8.0Hz), 7.01 (m, 4H), 5.26 (s, 2H), 4.40 (q, 2H, J=7.0Hz), 3.96 (s, 3H), 1.40 (t, 3H, J=7.0Hz).
Example 18: Synthesis of Compound 11:
och3 [0298] Compound of Example 18 was synthesized in an analogous synthetic route as 15 that described for Example 16, using 3-methoxy-phenol in the ultimate step to give
Compound 11 (6.1 mg) as a white solid: MS: [M+1] = 462. lH-NMR (500 MHz, CDC13) δ: 7.76 (s, 1H), 7.63 (d, 1H, J=2.5 Hz), 7.53 (d, 1H, J=9.0Hz), 7.15-7.22 (m, 2H), 6.67 (m, 2H), 6.55 (br dd, 1H, J=2.5, 8.0 Hz), 5.28 (s, 2H), 4.39 (q, 2H, J=7.0 Hz),3.96 (s, 3 H), 3.81 (s, 3H), 1.39 (t, 3H, J=7.0 Hz).
195
Example 19: Synthesis of Compound 12:
H3C [0299] Compound of Example 19 was synthesized in an analogous synthetic route as that described for Example 16, using 2,4-dimethylphenol in the ultimate step to give Compound 12 (3.1 mg) as a white solid: MS: [M+1] = 460. lH-NMR (500 MHz, CDCI3) δ: 7.76 (s, IH), 7.65 (d, IH, J=3.0Hz), 7.53 (d, IH, J=9.0 Hz), 7.17 (dd. IH, J=2.5, 8.5 Hz), 6.98 (m, 3H), 5.26 (s, 2H), 4.37 (q, 2H, J=7.0 Hz), 3.96 (s, 3H), 2.26 (s, 10 3H), 2.20 (s, 3H), 1.36 (t, 3H, J=7.0Hz).
Example 20: Synthesis of Compound 107:
107 [0300] To a solution of alcohol 43 where X = F (prepared in an identical manner to example where X — OCH3) (60 mg, 0.17 mmol) in THF (0.8 mL) was added phénol (30 mg, 0.32 mmol), triphenylphosphine (84 mg, 0.32 mmol). The reaction mixture was stirred at room température for 15 min. It was then cooied with an ice bath and DIAD (64 pL, 0.32 mmoi) in THF (0.2 mL) was added slowiy. The ice bath was removed and the reaction mixture was stirred at room température for 18h. LCMS indicated still the presence of some starting materiai. Phénol (10 mg), triphenylphosphine (28 mg) and DIAD (21 pL) were added to the reaction mixture and stirred for another hour. The
196
solvent was evaporated and the crude material was purified by Chromatography (RediSep 12 g silica-gel column. Eluting solvent: EtOAc) and prep TLC (eluting solvent: 5% MeOH/47.5%EtOAc/47.5% Hexanes) to give 11.4 mg (yield 16%) of the product Compound 107. [M+1] = 421). H'NMR (CDCI3) δ 7.92 (IH, dd, J= 3.5, 8.5 Hz), 7.80 (IH, s), 7.63 (1 H, dd, J= 5, 10 Hz), 7.38 (1H, m), 7.31 (2H, t, J= 8.5 Hz), 7.07 (2H, d, J=
8.5 Hz), 7.00 (1H, t, J= 8.5 Hz), 5.3 (2H, s), 4.39 (2H, q, J= 7 Hz), 1.38 (3H, t, J= 7 Hz).
Example 21; Synthesis of Compound 111:
[0301] To a suspension of alcohol 43 (X = Me) (160 mg, 0.47 mmol) in acetonitrile (9 mL) was added POBr3 (405 mg, 1.41 mmol). The reaction mixture was heated at 80 C for
h. The reaction mixture was cooled down with an ice bath and sat. aq. NaHCOj solution was added. The resulting solution was extracted with DCM (3X). The combined organic phases were washed with brine and dried over MgSCh. The solvent was concentrated to afford the desired product, 166 mg, 88% yield, [M+1] = 403).
[0302] To a suspension of the above alkyl bromide dérivative (30 mg; 0.075 mmol) in deoxygenated DME (2.7 mL) was added 3’pyridine boronic acid (14 mg, 0.11 mmol) and a 2M Na2CO3 solution (0.22 mL, 0.44 mmol). The suspension was stirred at room température for 5 min, then PdCl2(PPh3)2 (10 mg, 0.015 mmol) was added. The suspension was heated in a MW at 85 C for I hour. The réaction mixture was cooled and diluted with water and extracted with EtOAc (twice). The combined extracts were washed with brine and dried over MgSOi. Filtration and concentration gave the crude product which was purified by 2 prep TLC (eluting system: 3% MeOH in DCM) to give 5.3 mg (yield 18%) ofthe product Compound 111. MS: [M+l|=401. H*NMR(CDCI3) δ 8.66 (1H, bs), 8.48 (1H, bs), 7.96 (IH, s), 7.79 (1H, s), 7.66 (1H, d, J= 8 Hz), 7.50 (1H, d, J= 8
Hz), 7.43 (1H, d, J= 7 Hz), 7.23 (IH, m), 4.42 (2H, q, J= 7 Hz), 4.18 (2H, s), 2.54 (3H, s),
1.44 (3H,t,J= 7Hz).
197
Example 22: Synthesis of Compound 48:
Scheme 16.
t+RRT, NaBHtOAc),
[0303] To alcohol 43 (186 mg, 0.523 mmol) stirring in DMSO (1 mL) and dichloromethane (2.5 mL) at room température was added triethylamine (0.394 mL, 2.82 10 mmol) and pyridine sulfur trioxide complex (225 mg, 1.4! mmol). After 3 h stirring, the réaction was quenched with water (5 mL), and extracted with ethyl acetate three times. The combined organic solution was washed with water, brine, and dried over MgSOj.
The aldéhyde product 57 was isolated by ISCO flash column chromatography (RediSep 4g column) using a gradient elution of 0.5 to 8% MeOH in DCM. 84.4 mg (46%) was obtained as a yellowish foamy solid. MS: [M+1] =354.
[0304] To a stirring solution of aldéhyde 57 (15.5 mg, 0.0439 mmol) in 1,2dichloroethane (0.3 mL) at room température was added pyrrolidine (5.5 uL, 0.0658 mmol). After 2 min stirring, the solution tumed clear, and NaBH(OAc)j (14.4 mg) was added. The reaction mixture was stirred for 4 h, and was quenched with saturated
NaHCOj, and extracted with ethyl acetate three times. The combined organic layer was
198 washed with water, brine, and dried over Na^SO». Prep TLC with 10% MeOH in DCM gave 13.1 mg (73%) of the desired Compound 48 as a clear filmy solid. MS: [M+1] = 409. 'H-NMR (500MHz, CDCI3) Ô: 7.74 (s, 1H), 7.62 (d, 1H, J=3.0Hz), 7.51 (d, 1H, J=9.0Hz), 7.14 (dd, 1H, J=3.5, 9.0Hz), 4.42 (q, 2H, J=6.5Hz), 3.94 (s, 3H), 3.87 (br s, 2H), 2.65 (br s, 4H), 1.79 (br s, 4H), 1.44 (t, 3H, J=7.0Hz).
Example 23: Synthesis of Compound 49:
[0305] Compound of Example 23 was synthesized in an analogous synthetic route as that described for Example 22, using morpholine in the ultimate step to give the compound of Example 23 as a clear filmy solid: MS: [M+1] - 425. ’H-NMR (500MHz, CDC13) δ: 7.75 (s, 1H), 7.63 (d, 1H, J=3.0 Hz), 7.52 (d, 1H, J=9.5 Hz), 7.15 (dd, 1H, J=3.0,9.0 Hz), 4.42 (q. 2H, J=7.5 Hz), 3.95 (s, 3H), 3.76 (br s, 2H), 3.71 (br s, 4H). 2.57 (br s, 4H), 1.44 (t, 3H, J=8.0 Hz).
Example 24: Synthesis of Compound 50:
[0306] Compound of Example 24 was synthesized in an analogous synthetic route as that described for Example 22, using diethylamine in the ultimate step to give the compound of Example 24 as a clear filmy solid: MS: [M+1] = 411. 'H-NMRfSOO MHz, CDCI3) δ: 7.74 (s, 1H), 7.64 (br d, 1H, J=3.0 Hz), 7.51 (d, 1H, J=9.0Hz), 7.15 (dd, 1H, J=2.5,9.0 Hz), 4.43 (q, 2H, J=6.5 Hz), 3.96 (s, 3H), 3.86 (br s, 2H), 2.64 (br s, 4H), 1.44 (t, 3 H, J=8.5 Hz), 1.15 (br s, 6H).
199
Example 25: Synthesis of Compound 51:
[0307] Compound of Example 25 was synthesized în an analogous synthetic route as that described for Example 22, using methyl benzyl amine in the ultimate step to give the compound of Example 25 as a clear filmy solid: MS: [M+1] =459. lH-NMR (500 MHz, CDC!3)ô:7.75 (s, IH), 7.63 (d, 1H,J=3.O Hz), 7.51 (d, IH, J=8.5 Hz), 7.36(brd, 2H, J=8.0 Hz), 7.30 (m, 2H), 7.23 (m, IH), 7.15 (dd, IH, J=3.0,9.0 Hz), 4.38 (q, 2H, J=7.5 Hz), 3.95 (s, 3H), 3.85 (br s, 2H), 3.63 (br s, 2H), 2.25 (s, 3H), 1.41 (t, 3H, J=7.0 Hz).
Example 26: Synthesis of Compound 170:
170 [0308] Isobutyramidoxime (41.8 mg, 0.41 mmol) and ester 48 (27.9 mg, 0.0683 mmol) in a round bottom flask was azeotroped in toluene on a Rotavap several times, suspended in anhydrous THF (0.6 mL), and then cooled to 0°C. NaH (60% oil suspension; 10.9 mg, 0.273 mmol) was added. Ice bath was removed and the reaction mixture was stirred at RT for 20 min before being heated at 70°C for 6hrs, and cooled. Water (4 mL) was added, and the mixture was extracted wîth EtOAc three times. The combined organic solution was washed with brine and dried over MgSO4. Prep. TLC with 10% MeOH in
200
EtOAc gave 10.4 mg (34%) ofthe desired product Compound 170 as a clear filmy solid. MS: [M+1] = 447.
Scheme 17.
Rl » OMe* 43
R, Me; R2 3 H: compound 102 (HO)2B
Pd(PPh3)4. Na2CO3 V orPdCl^ppf
R
Rt 3 OMe; R2 3 m-CI-Pti: compound 62
Rt « OMe; R2 3 m-CN-Ph: compound 53
Rt « Me; R2 3 oCI-Ph: compound 54
Rt 3 Me; R2 Ph: compound 101
Rt 3 OMe; o-CI-Ph: compound 108 [0309] The starting alcohol 43 (160 mg, 0.45 mmol) was treated with phosphorous oxide tribromide (400 mg, 1.4 mmol) in acetonitrile (10ml) at 80°C for 5 h. The reaction was then cooled down to 0°C, quenched with sat. NaHCO3, and extracted with dichloromethane twice. Combined dichloromethane solution was washed with brine and dried over MgSQ». Filtration and solvent removal in vacuo gave 173.3 mg (92%) ofthe bromide as a yellowish foamy solid. MS: [M+1] =418.
201 [0310] To a suspension of bromide (55 mg, 0.131 mmol) in dimethoxyethane (2 ml; degassed) was added 2M Na2CCh (0.39 ml, 0.78 mmol) and 3-chlorophenyl boronic acid (42.2 mg, 0.27 mmol). The reaction mixture was stirred at room température for 2 min, then Pd(PPh3)4 (75 mg, 0.065 mmol) was added, and the suspension was heated În a 85°C 5 oil bath for 90 min. Upon cooling, the réaction mixture was diluted with EtOAc and washed with brine. The aqueous layer was separated and extracted with EtOAc three times. Ail organic layers were pooled and dried over Na2SOj, then filtered and solvent was removed in vacuo. The product was isolated by successive prep TLC purifications, using 20% hexanes in EtOAc followed by 5% MeOH in DCM. 9.6 mg product (Compound 52) was obtained as a brownish solid. MS: [M+1 ] = 450. ’H-NMR (500 MHz, CDCB) δ: 7.75 (s, 1H), 7.64 (d, 1H, J=3.0 Hz), 7.51 (d, 1H, J=9.5 Hz), 7.31 (brs, 1H), 7.23 (brs, IH), 7.17 (m, 3H), 4.43 (q, 2H, J=7.0Hz), 4.15 (s, 2H), 3.96 (s, 3H), 1.44 (t, 3 H, J=8.0Hz).
Example 28: Synthesis of Compound 53:
[0311] Compound of Example 28 was synthesized in an analogous synthetic route as that described for Example 27, using 3-cyanophenyl boronic acid in the ultimate step to 20 give the compound of Example 28 as a brownish solid: MS: [M+1] = 441. ’H-NMR (500 MHz, CDCB) δ: 7.75 (s, 1 H), 7.66 (br s, 1H), 7.64 (d, 1H, J=3.0 Hz), 7.61 (br d, 1H,
J=7.5 Hz), 7.39 (t, 1H, J=7.5 Hz), 7.16 (dd, 1H, J=3.5, 9.5 Hz), 4.45 (q, 2H, J=7.0H), 4.20 (s, 2H), 3.96 (s, 3H), 1.45 (t, 3H, J=7.0 Hz).
202
Example 29: Synthesis of Compound 54:
[0312] Compound of Example 29 was synthesized in an analogous synthetic route as that described for Example 27, starting with the alcohol where Ri = methyl, and using 2chlorophenyl boronic acid in the ultimate step to give the compound of Example 29 as a brownish solid: MS: [M+1 ] = 434.
Example 30: Synthesis of Compound 101:
101 [0313] Compound of Example 30 was synthesized in an analogous synthetic route as that described for Example 27, starting with the alcohol where Ri = methyl, and using phenyl boronic acid in the ultimate step to give the compound of Example 30 as a brownish solid product which was purified by chromatography (RediSep 4 g silica-gel column. Eluting solvent: EtOAc) then a prep TLC (eluting system: 40% DCM/40% Hexanes/17% EtOAc/ 3% MeOH) to give 5.9 mg (yield 31%) ofthe product Compound 101. MS: [M+1 ]= 402. HlNMR (CDCIj) δ 7.96 (1H, s), 7.77 (1H, s), 7.55 (IH, m), 7.47 (IH, m), 7.32 (5H, m), 4.41 (2H, q, J= 7 Hz), 4.17 (2H, s), 2.53 (3H. s), 1.43 (3H, t, J= 7 Hz).
203
Example 31: Synthesis of Compound 102:
[0314] To a suspension of the bromîde in EtOAc (2mL) and MeOH (2mL) was added activated 10% Pd/C (5 mg). The suspension was stirred under a hydrogen atmosphère for 48 h. The solution was filtered over celite. The filtrate was concentrated and purified by chromatography (RediSep 4 g silica-gel column. Eluting solvent: EtOAc) to give 15.9 mg (33%) ofthe desired product Compound 102. MS: [M+1] = 324. H!NMR (CDClj) δ 7.96 (1H, s), 7.78 (1H, s), 7.49 (1H, d, J= 9 Hz). 7.42 (1H, d, J= 8 Hz), 4.43 (2H, q, J= 7.5 Hz), 2.53 (3H. s), 2.44 (3H, s), 1.45 (3H, t, J= 7.5 Hz).
Example 32: Synthesis of Compound 108:
108 [0315] To a suspension ofthe bromide dérivative where Ri = OMe, (18 mg; 0.043 mmol) in deoxygenated DME (2 mL) was added 2-chlorophenyl boronîc acid (10 mg, 0.065 mmol) and a 2M Na2CO3 solution (0.13 mL, 0.26 mmol). The suspension was stirred at room température for 15 min, then PdCI2dppf (7 mg, 0.009 mmol) was added. The suspension was heated în an oil bath at 85 C for 1 hour. The reaction mixture was diluted with water and extracted with EtOAc (twice). The combined extracts were washed with brine and dried over Na2SO4. Filtration and concentration gave the crude product whîch was purified by PrepTLC (eluting system: 5% MeOH/ 47.5% Hex/47.5% EtOAc) to give 3.5 mg (yield 18%) ofthe product Compound 108. MS: [M+l]=451. H*NMR
204 (CDCIj) δ 7.77 ( I H, s), 7.63 (1 H, d, J= 3 Hz), 7.52 (1 H, d, J= 11.5 Hz), 7.36 (1 H, m),
7.31 (IH, m), 7.18 (2H, m), 7.14 (IH, dd, J= 3,9 Hz), 4.38 (2H, q, J= 7 Hz), 4.27 (2H, s), 3.94 (3 H, s), 1.41 (3 H, t, J= 7 Hz).
Scheme 18a.
CHpprt ·μμ·«μ in
1)U0H nn
R^CHgOCH, 11·
R^CONHNHy
DlKA.PhCI r à-r1 -, N'N · θΐ61 R1 * CHjOPh M
R' CHjO-4-F-Pti cempMntf M R1 CHjOCHj CMipouftd tU
CHjOPtrç R « CH3C(CH1)> wmpmintf lit
CD1.NHPH . ___ __ iPOCO R^CMjOR»; R · W*T IM
i
R1 · CHjOCH, 1»
205
Scheme 18b.
Exemple 33: Synthesis of Compound 55:
[0316] To a solution of compound 58 (6.6 g, 33.5 mmol) in dichloromethane (100 mL) were added DIPEA (8.65 g, 67 mmol), HOBt (5.4 g, 36.85 mmol) and EDC1 (9.6 g, 50.3 mmol). After about 15 min stirring, to the homogeneous reaction mixture was added a solution of 2,4-dimethoxybenzyl amine (5.6 g, 33.5 mmol) in dichloromethane (50 mL) dropwise under nitrogen atmosphère. The resulting mixture was stirred under nitrogen atmosphère at room température for 16h. The reaction mixture was washed successively with INNaOH (100 mL), water (100 mL) and brine (100 mL). The organic phase was then dried over Na2SO4 and evaporated to give a crude solid product 59 that crystal I ized from ethyl ether. Filtration and open air suction drying afforded an off-white solid pure product 9.8g (96%), (MS: [M+1 ] = 347).
206 [0317] Toasolutionofcompound59(9.8g, 28.3 mmol) ιn MeOH/EtOAc (1:1, 100 mL) was added 10% wet Pd-C (1.8 g, 10% mmol). After three consecutive vacuuming and flushing with nitrogen, the heterogeneous reaction mixture was subjected to a balloon hydrogénation at atmosphère pressure up untîl the absorption of hydrogen ceases, about 4h. The réaction mixture was filtered through a celite pad and evaporated to afford the pure desired product 60 as a brown oil 8.63g (96%), (MS: [M+1 =317]). This product was used directly in the next step.
[0318] To a solution of compound 60 (8.63g, 27.3 mmol) in dichloromethane (100 mL) was added triethylamine (5.5g, 54.6 mmol). The mixture was cooled with îce bath and treated with bromo acetyl chloride (5.2g, 32.76 mmol) under nitrogen atmosphère. The ice bath was removed and the mixture left stirring for 18h. The reaction mixture was washed successively with saturated NaHCO3 (100 mL), water (100 mL) and brine (100 mL). The organic phase was then dried over Na2SÜ4 and evaporated to give a crude solid product 61. The crude product was crystallized from methanol, filtered and dried to afford a brown solid pure product 10.3 g (87%), [MS: 439].
[0319] To a solution of compound 61 (10 g, 22.9 mmol) în DMF (1000 mL) was added K2CO3 (4.8 g, 45.8 mmol). The mixture was heated at 50 °C for 24h. LCMS showed a complété conversion to the desired product. The mixture was cooled to room température and the inorganic solid was filtered. The solvent was removed under high vacuum. The resulting crude product 62 was crystallized from methanol, filtered and dried to give a pure brown solid product 6.4g (78%), (MS: [M+1] = 357).
[0320] To compound 62 (4.46 g, 12.52 mmol) dissolved in 2.5:1 THF/DMF (50 mL) at 20°C was added /-BuOK (97%, 1.88 g, 16.28 mmol). The mixture was warmed to 25 °C, and after stirring for 30 min was cooled again to -20 °C. Following dropwise addition of diethyl chlorophosphate (2.35 mL, 16.28 mmol), the mixture was stirred for 3 h while warming from -20 to 25 °C. The reaction mixture was re-cooled to 0 °C and to it was added ethyl isocyanoacetate (1.92 mL, 17.53 mmol). Subséquent cooling to -78 °C was followed by addition of /-BuOK (97%, 1.88 g, 16.28 mmol) and stirring at RT for 5 h. Progress was monitored by LC/MS. The reaction was quenched by addition of 1:1 saturated NaHCO3 / H2O (140 mL), the precipitate was filtered, washed with H2O and air
207 dried ovemight to afford 4.81 g (85%) of tmtdazole denvative 63 as a yellow solid (MS: [M+1] = 452).
[0321] To compound 63 (4.81 g, 10.65 mmo!) in dichloromethane (35 mL) at 0 °C was added trifluoroacetic acid (35 mL) followed by dropwise trifluoromethanesulfonic acid (1.9 mL, 21.31 mmol). The mixture was warmed to RT, stirred for 2 h, then concentrated to afford a residue which was dissolved in dichloromethane (120 mL). The crude solution was partitioned between chilled saturated NaHCOjand dichloromethane. The organic extractions were combined, dried (MgSOi), filtered and concentrated to afford 3.2 g (99%) of deprotected product 64 (brown solid) of sufficient purity to take on the next step (MS: [M+1] = 302).
[0322] To lactam 64 (51.8 mg, 0.172 mmo!) and N,N-dimethyl-p-toluidine (93.0 mg, 0.688 mmol) stirring in chlorobenzene (1 ml) under nitrogen was added POC13 (52.7 mg, 0.344 mmol). The reaction was then heated at 135°C for 2 h. Upon cooling to room température, phenoxy acetic acid hydrazïde (228.4 mg, 1.36 mmo!) was added in situ to the imino-chloride 65, followed by D1PEA (90 ul). The reaction was stirred at room température for 30 min, then heated at 100°C for 90 min. The reaction mixture was cooled, saturated NaHCOj (aq.) was added, and extracted with ethyl acetate three times; combined organic layer was washed with brine, and dried over MgSO+. After filtration and concentration, the product as Compound 55 was isolated by ISCO flash column chromatography (RediSep 4 g column, 1 to 10 % MeOH in DCM as eluting gradient) as a white solid, Wt: 8.6 mg. MS: [M+1] =432. *H-NMR(500 MHz, CDCB) δ: 7.81 (s, 1H), 7.71 (d, 1H, J=3.5 Hz), 7.52 (d, 1H, J=9.0 Hz), 7.32 (m, 2H), 7.2! (dd, IH, J=2.5, 8.5 Hz), 7.11 (d, 2H, J=8.5 Hz), 7.02 (m, 1H), 5.44 (s, 2H), 4.38 (q, 2H, J=7.5 Hz), 3.94 (s, 3H), '1.39 (t, 3H, J=7.0 Hz).
Example 34: Synthesis of Compound 56:
208 [0323] Compound of Example 34 was synthesîzed in an analogous synthetic route as that described for Example 33, using 4-fluoro-phenoxy acetic acid hydrazide in the ultimate step to give the compound of Example 34 as a yellowish solid: MS: [M+1] = 450. ‘H-NMR (500 MHz, CDCI3) δ: 7.82 (s, 1H), 7.73 (d, 1H, J=3.5 Hz), 7.53 (d, 1H, J= 10.0 Hz), 7.22 (dd, 1H, J=3.5, 9.0 Hz), 7.08-6.99 (m, 4H), 5.41 (s, 2H), 4.41 (q, 2H, J=7.0 Hz), 3.95 (s, 3H), 1.42 (t, 3H, J=6.5 Hz).
Example 35: Synthesis of Compound 103:
103 [0324] Compound of Example 35 was synthesîzed in an analogous synthetic route as that described for Example 33, using 2-methoxy acetic acid hydrazide in the ultimate step to give the compound of Example 35 as a yellowish solid: MS: [M+1] = 370.
Example 36: Synthesis of Compound 118:
118 [0325] Acetamide oxime (8.4 mg, 0.108 mmol) was azeotroped in toluene three times on a Rotavap, then suspended in THF (1.0 mL). NaH (60% minerai suspension; 3.3 mg, 0.081 mmol) was added, and the mixture was stirred at RT for 10 min. Ester 55 (23.2 mg, 0.054 mmol) was added next. After 40min stirring at RT, the reaction mixture was heated at 70°C for 4 h. Upon cooling, cold water (5 mL) was added to the reaction mixture, and ppts were collected by filtration, washed with water, and dried to give 9.7 mg (41%) of the desired product as a yellowish solid. MS: [M+1 ] = 442.
209
Example 37: Synthesis of Compound 128:
128 [0326] Compound of Example 37 was synthesized in an analogous synthetic route as that described for Example 36 above, using ester Compound 103 În the ultîmate step to 5 give the compound of Example 37 as a brownish solid: MS: [M+i] = 380.
Example 38: Synthesis of Compound 130:
130 [0327] Compound of Example 38 was synthesized în an analogous synthetic route as that described for Example 36, starting with ester Compound 103 and condensing with 10 isobutyramidoximeto give the compound of Example 38 asayeliowishsolid: MS:
[M+1] =408.
Example 39: Synthesis of Compound 119:
119 [0328] To the carboxylic acid (13.9 mg, 0.0345 mmol; obtained through LÎOH
210 hydroxysis of the precursor ester 55) stirring in DCM (0.2 mL) was added Neopentyl alcohol (30.4 mg, 0.345 mmol), DMAP (4.2 mg, 0.0345 mmol), and EDC (20 mg, 0.104 mmol). After five hour stirring, the réaction mixture was diluted with EtOAc, washed with sat. NH4CI, sat. NaHCOj, brine, and dried over MgSO4. Silica gel chromatographie 5 purification using a gradient of 0 to 8% MeOH in EtOAc gave 11.7mg (72%) of the desired product Compound 119 as a yellowish solid. MS: [M+1] = 474.
Example 40: Synthesis of Compound 120:
120 [0329] Compound of Exemple 40 was synthesized in an analogous synthetic route as 10 that described for Example 39 above, using 2-propyl alcohol in the ultimate step to give the compound of Example 40 as a yellowish solid: MS: [M+l] = 446.
Example 41: Synthesis of Compound 129:
129 [0330] Compound 103 (Scheme 18a) (66.1 mg, 0.179 mmol) was hydrolyzed in a solvent System of THF/water/MeOH (1.8 ml total, 6/5/1 ratio) by treating with LiOH (21.4 mg, 0.895 mmol) at RT for 2 h. Dil. HCl was added to acidify (pH ~3) the reaction mixture. The precipitate was collected by filtration, washed with water, and dried to give 49.0mg (80%) of the acid as a brownish solid.
[0331] The acid thus obtained was stirred in DMF (0.7 mL) at 0°C. NaHCOj (48.1 mg,
211
0.572 mmol) was added, followed by N-bromosuccinamide (96.7mg, 0.543 mmol). After ovemight stirring, the reaction was diluted with EtOAc, and washed with sat. NaHCOj.
Aq. Layer was separated and extracted with EtOAc. Combined organic layer was washed with brine, dried over MgSO4, filtered, and concentrated. The product bromide was obtained by silica gel column chromatography with a gradient elution of 0 to 13% MeOH in EtOAc as a white solid (Compound 129). Wt: 28.6 mg (53%). MS: [M+1] = 377.
Example 42: Synthesis of Compound 131:
[0332] Compound 129 (22.6mg, 0.060 mmol) was hydrogenated over 10% Pd-C in
EtOAc (1 mL) and MeOH (1 mL) for 16 h. Filtration over Celite, and solvent removal gave 14.9 mg (84%) of the des-bromo product Compound 131 as a lightly yellowish solid. MS: [M+1] = 298.
Example 43: Synthesis of Compound 122:
122 [0333] The phenoxy analog (Scheme 18a, Ri = OPh) of add 66 (20.4 mg, 0.0506 mmol) was suspended and stirred in DCM (0.5 mL) at RT. Carbonyl diîmidazole (16.4 mg, 0.101 mmol) was added. After 2 h stirring, the resulting suspension was cooled to 0°C, and ammonia (30 uL) was added dropwise. After 20min stirring, ice bath was removed and the reaction was allowed to proceed at RT for Ihr. The reaction was concentrated by removing DCM in vacuo. Water (3 mL) was added, and precipitate was collected by filtration, washed with water, and dried to give 16.2 mg of the crude primary
212
amide which was used without further purification.
[0334] The primary amide (16.2mg, 0.0402 mmol) was treated with POC13 (46.2 mg, 0.302 mmol) in 1,4-dioxane (0.5 mL) at 95°C ovemight. The reaction mixture was then quenched with sat. NaHCOj (5 mL), cooled to 0°C, and precipitate collected by suction filtration, washed with water, and dried to give 13.6 mg (88%) of the nitrile as a brownish solid, Compound 122. MS: [M+1] = 385.
Example 44: Synthesis of Compound 123:
123 [0335] To Add 66 (15.8mg, 0.0392 mmol) stirring in THF (0.15 mL) and DCM (0.15ml) was added Ν,Ο-dimethylhydroxylamine HCl (4.6 mg, 0.047 mmol) and Nhydroxylbenzotriazole hydrate (6.0 mg). EDC (11.3 mg, 0.0588 mmol) and triethylamine (11.9 mg, 0.118 mmol) were then added, and the reaction was stirred at RT for 12hrs, diluted with EtOAc, washed with sat. NH4CI, brine, and dried over MgSQ». Filtration and solvent removal în vacuo gave 14.4 mg (82%) ofthe Weînreb amide which was used without further purification.
[0336] To the Weînreb amide (14.4 mg, 0.0323 mmol) stirring in THF (0.3 mL) at 0°C was added ethyl magnésium bromide etherate (3M; 0.323 mL). The reaction was allowed to warm to RT and stirred for 14 hrs., quenched with sat. NH4CI, extracted with EtOAc three times; combined organic layer washed with brine and dried over MgSO4. Filtration and solvent removal gave the crude ketone product which was purified by prep. TLC using 8% MeOH in EtOAc. Wt: 4.6 mg (34%) of Compound 123. MS: [M+1] = 416.
Example 45: Synthesis of Compound 124:
213
124 [0337[ Weinreb amide (18.0 mg, 0.0403 mmol) described above was treated with
DIBAL (IM THF; 0.363 mL) at -78°C for Ihr, then still at -78°C quenched with Rochelle sait solution (20%) ovemight. The aq. solution was extracted with EtOAc three times;
combined organic layer was washed with brine, and dried over MgSOj. Filtration and solvent removal in vacuo gave 13.7mg ofthe crude aldéhyde which was used without further purification.
[0338] The crude aldéhyde (13.7 mg) in DCM (0.7 mL) at RT was treated with DeoxoFluor (54.8 mg, 0.248 mmol) for 16hrs. The reaction was quenched with sat. NaHCOj (5 10 mL) for 20 min, extracted with EtOAc three times; combined organic layer washed with brine, and dried over MgSOj. Filtration and solvent removal followed by prep. TLC purification using 10% MeOH in EtOAc gave 7.5 mg (52%) ofthe desired difluoride Compound 124 as a yellowish solid. MS: [M+1] = 410.
Example 46: Synthesis of Compound 142:
142 [0339] Weinreb amide (8.8 mg, 0.0197 mmol) from above in THF (0.15 mL) at 0°C was treated with phenylmagnesium bromide (IM THF; 0.54 mL) for 2.5hrs, quenched with sat. NH4CI, extracted with EtOAc twice; combined organic layer washed with brine
214
and dried over MgSO.4. Filtration and solvent removal gave the crude ketone which was used without further purification. The ketone in THF (0.5 mL) was treated with NaBIfi (6 mg) at RT for 2 hrs., then quenched with sat. NH4CI, extracted with EtOAc three times; combined organic layer washed with brine, and dried over MgSO4. Filtration and solvent 5 removal gave the crude alcohol which was used without further purification. The thus obtained alcohol in DCM (1.4 mL) was treated with triethylsîlane (86.4 mg, 0.75 mmol) and trifluoroacetic acid (171.0 mg, 1.5 mmol) at 40°C ovemight, then concentrated in vacuo, diluted wîth EtOAc, washed wîth sat. NaHCOj, brine, and dried over MgSO.4.
Filtration and solvent removal gave the crude benzyl product which was purified by silica 10 gel column chromatography using 0 to 12 % MeOH in EtOAc as eluent; 3.6 mg of
Compound 142 was obtained as a yellowish solid. MS: [M+1] = 450.
Scheme 19:
Example 47: Synthesis of Compound 106:
215
[0340] To lactam 64 ( 185.7 mg, 0.616 mmol) in chlorobenzene (5 mL) was added N,N-dimethyl-p-toluidine (333.3 mg, 2.465 mmol) and phosphorous oxychlorïde (188.9 mg, 1.232 mmol). The reaction mixture was heated at 135°C for 2hrs, cooled to RT, and formylhydrazide (296.0 mg, 4.93 mmol) was added, followed by dïisopropyl ethyl amine (238.8 mg, 1.85mmol). Following 30 min stirring at RT, the reaction was heated at 100°C for 1 hr., cooled, and sat. NaHCOj (15 mL) added, extracted with EtOAc twice; combined organic layer washed with brine, and dried over MgSO4. Filtration and solvent removal gave the crude triazole product which was purified by silica gel column chromatography using 0 to 15% MeOH in EtOAc elution, 35.9mg (18%) was obtained as a brownish solid. MS: [M+1] = 326.
[0341] The triazole from above in DCM ( 1 mL) was treated with N-bromosuccïnamide (37.6 mg, 0.21 mmol) at 0°C. The reaction was allowed to warm to RT slowly, and proceeded at RT ovemight, diluted with EtOAc, washed with sat. NaHCOj, brine, and dried over MgSO4. Filtration and solvent removal gave the crude bromide which was purified by silica gel column chromatography using 0 to 10% MeOH în EtOAc gradient; 22.9 mg (51%) of Compound 106 was obtained as an ofT-whîte solid. [MS]: 406.
Example 48: Synthesis of Compound 104:
[0342] A microwave reaction vessel was charged with phénol (20.3 mg, 0.216 mmol), the bromide substrate from Example 47 (29.1 mg, 0.0719 mmol), Cs2COj (117.0 mg,
216
0.360 mmol), diethyl 1,3-acetonedicarboxylate (14.5 mg, 0.0719 mmol), and DMF (0.5 ml). The vessel was flushed with nitrogen gas. Cul (6.8 mg, 0.036 mmol) was added, and the mixture was stirred at RT for 5min before heated @14O°C under MW radiation conditions for 60 min. The reaction mixture was diluted with EtOAc, washed with water, aq. Layer separated and extracted with EtOAc twice; combined organic solution was washed with brine and dried over MgSOj. Filtration and solvent removal gave the crude ether product which was purified by prep. TLC using 5% MeOH in DCM; 6.6 mg of Compound 104 was obtained as a yellowish solid. MS: [M+1] = 418.
Example 49: Synthesis of Compound 105:
[0343] Compound of Example 49 was synthesized in an analogous synthetic route as that described for Example 48 above, using 3-methoxy phénol in the place of phénol, to give the compound of Example 49 as a yellowish foamy solid: MS: [M+l ] = 448.
Scheme 20:
R · kPr compound 112
R « CHjC(CHj)j‘ compound 111
R>CHjCFi' compound 114
Compound IM R . Pn Compound 1J»
R J-PyrWyf Compound 140
R 1-Mo-4-Pyr»zolyt Compound 182
R 2-M»~+-Pyridyt Compound 1S4
217
Example 50: Synthesis of Compound 112:
112 [0344] To a solution of Compound 2 (160 mg, 0.49 mmol) in THF (6 mL), water (5 mL) and MeOH (1 mL) was added LiOH (59 mg, 2.45 mmol). The solution was stirred at room température for 3 h. The solution was concentrated and the crude material was acidified with IN HCl until pH 3-4. No solid was observed. EtOAc was added and the organic phase was extracted (3x). The combined extracts were washed with brine and dried over MgSO4. Filtration and concentration gave 112 mg (77% yield) ofthe desired carboxylic acid product as an orange solid MS: [M+1] = 300.
[0345] To a suspension of acid (30 mg, 0.1 mmol) in dichloroethane (0.2 mL) was added thionyl chloride (0.4 mL; 5 mmol) and DMF (20 pL). The resulting solution was heated at 70 C for 1 hour. Another 0.2 mL of thionyl chloride was added and the solution was heated for another 30 min. The solvent was removed. The crude material was dried under vacuo.
[0346[ The crude acid chloride (0.1 mmol) was suspended in isopropanol and stirred at room température for 18 h. The solvent was evaporated and the crude material was purified by chromatography. (RediSep 4 g silica-gel column, eluted with 10% MeOH in DCM) to give 8.6 mg (25% yield) of product Compound 112 [M+1] =342).
H’NMR (CDCb) δ 7.90 (1H, d, J= 9 Hz), 7.79 (1H, bs), 7.63 (1H, bs), 7.36 (1H, bs), 3.48 (1H, m), 2.45 (3H, s), 1.43 (6H. d, J= 6.5 Hz).
Example 51: Synthesis of Compound 113:
218
113 [0347] The crude acid chloride prepared above (0.066 mmol) was suspended in dichloroethane (lmL) and 2,2-dimethyl-l-propanol (300 mg, 3.4 mmol) was added. The solution was stirred at room température for 18 h. No product was formed. To the solution above, was added DMAP (5 mg, 0.004 mmol) and DCC (15 mg, 0.073 mmol). The solution was stirred at room température for 2 h. The reaction mixture was directly applied on a prep TLC (eluting System: 75 EtOAc în Hexanes) to give 7.2 mg (30% yield) of product Compound 113. MS: [M+1 ]=370. H’NMR (CDCh) δ 7.91 (1H, dd, J= 3,9 Hz), 7.79 (1H, s), 7.61 (1H, dd, J= 4.5,9 Hz), 7.35 (1H, m), 4.11 (2H, s), 2.44 (3H, s), 1.07 (9H,s).
Example 52: Synthesis of Compound 114:
114 [0348] The crude acid chloride prepared above (0.066 mmol) was suspended in dichloroethane (lmL) and 2,2,2-trifluoroethanol (0.1 mL, 1.4 mmol) followed by triethylamine (0.6 mL, 4.3 mmol) was added. The solution was stirred at room température for 2 h 30 min. The solvent was evaporated and the crude material was purified by chromatography. (RediSep 4 g silica-gel column, eluted with EtOAc) then purified with a prep TLC (eluting system: 70 % EtOAc in Hexanes) to give 8.1 mg (32% yield) of product Compound 114 [M+1] = 382).
H’NMR (CDCh) δ 7.91 (1H, dd, J= 3.5,9.5 Hz), 7.83 (1H, s), 7.63 (1H, dd, J= 4.5,9.5 Hz), 7.35 (1H, m), 4.77 (2H, m), 2.43 (3H, s).
Example 53: Synthesis of Compound 136:
219
136 [0349] To a solution of acid prepared in Example 50 (100 mg, 0.33 mmol) în DMF (1.5 mL) cooled with an ice bath was added NallCOj (111 mg, 1.32 mmol) followed by NBS (117 mg, 0.66 mmol). The solution was stirred at room température for 14 h. The reaction mixture was diluted with water and extracted with EtOAc (5X). The combined extracts were washed with brine (2x) and dried over MgSO.». Filtration and concentration gave a crude product. Chromatography (RediSep 4 g silica-gel column, eluted with EtOAc) to give 93 mg (85% yield) of product Compound 136 [M+1] =334).H’NMR (CDCIj) δ 7.87 (1H, dd, J= 2.5, 8.5 Hz), 7.72 (1H, s), 7.56 (1H, dd, J= 6,10 Hz), 7.33 (1H, m), 2.44 (3 H, s).
Example 54 Synthesis of Compound 139:
139 [0350] General coupling procedure: To a solution of Compound 136 (20 mg, 0.061 mmol) în degassed DME (0.9 mL) and water (0.1 mL) was added phenyl boronic acid (11 mg, 0.092 mmol), césium carbonate (80 mg, 0.24 mmol) and Pd Cl2dppf (5 mg, 0.066 mmol). The suspension was heated at 80 °C for one hour. The reaction mixture was diluted with water, extracted with EtOAc (3X). The combined extracts were washed with brine (2x) and dried over MgSO4. Filtration and concentration gave a crude product which was purified by prep TLC (eluting system: 3% MeOH in EtOAc).
[0351] Compound 139 was prepared using phenyl boronic acid. 10.8 mg (54% yield) of product was obtained. MS: [M+1] = 332. H'NMR (CDCIj) δ 7.87 (1H, dd, J= 3.5,9.5 Hz), 7.85 (1H, s), 7.63 (3H, m), 7.50 (2H, t, J= 6.5 Hz), 7.35 (2H, m), 2.41 (3H, s).
220
Example 55: Synthesis of Compound 140:
140 [0352] Compound 140 was prepared similarly using 3-pyridine boronic acid. 8.9 mg (27% yield) ofproduct was obtained. MS: [M+1] = 333. H’NMR (CDCIj) δ 8.86 (IH, s), 8.63 (IH, d, J= 5 Hz), 8.01 (IH, m), 7.90 (2H, m), 7.64 (IH, dd, J= 5.5,9 Hz), 7.44 (IH, m), 7.36 (IH, m), 2.39 (3 H, s).
Example 56: Synthesis of Compound 152:
152 [0353] Compound 152 was prepared using l-methylpyrazoie-4-boronic acid, HCl.
12.5 mg (63% yield) of product was obtained. MS: [M+1] = 336. H'NMR (CDCb+ MeOD<) δ 9.04 (IH, bs), 7.99 (IH, bs), 7.75 (2H, m), 7.41 (2H, m), 3.95 (3H, s), 2.32 (3H, s).
Example 57: Synthesis of Compound 154:
154 [0354] Compound 154 was prepared using 2-methylpyridine-4- boronic acid pinacol ester. 7.1 mg (34% yield) of product was obtained. MS: [M+1] = 347. H’NMR (CDCIj)
221
δ 8.6 (1Η, d, J= 6 Hz), 7.89 (1H, dd, J= 3.5, 8.5 Hz), 7.87 (1H, s), 7.64 (1H, dd, J= 5.5, 9 Hz), 7.48 (1H, s), 7.36 (2H, m), 2.64 (3H, s), 2.41 (3H, s).
Scheme 21:
117
222 [0355] In a 100 mL round-bottom flask, the lactam ester 16’ (2 g, 7.35 mmol; which was prepared in analogous fashion as 16 described in Scheme 11) was dissolved in 60 mL of anhydrous THF. The solution was stirred at room température under a nitrogen atmosphère. LÎBH* (2 M în THF, 4 mL, 8 mmol) was added slowly. The reaction mixture was stirred under a nitrogen atmosphère for 18 h. More LiBH* (2 M in THF, 2 mL, 4 mmol) was added slowly. The réaction mixture was stirred for another 24 h. A mixture of EtOAc/EtOH (20 mL/20 mL) was added to the reaction mixture and it was concentrated. The residue was taken up in MeOH and silica gel was added. After volatile solvents were evaporated, the solid was loaded onto a RediSep 40 g silica-gel column. The desired product was eluted with 5:1 v/v CHîCh/MeOH. The alcohol was obtained as a white solid (1.14 g, 67% yield). MS: [M+1] =231.
[0356] The alcohol ( L14 g, 4.96 mmol) was suspended in 16 mL of HBr 33% in AcOH and heated at 80°C for 18 h. The solution was cooled down with an ice bath and diluted with EtOAc. A white solid could be observed. Slowly, a sat. aq. NaHCO3 solution was added. Large amount of EtOAc and MeOH were used to solubilize the solid. The organic phase was extracted (3x) and the combined organic phases were washed with brine, dried over MgSO*. Filtration and concentration gave a crude product which was used in the next step without further purification. MS: [M+1] = 293.
]0357] To a solution of alkyl bromide dérivative (4.96 mmol) in EtOAc (50 mL), MeOH (200 mL) and THF (50 mL) was added wet 10% Pd/C (250 mg) and the resulting suspension was stirred under a hydrogen atmosphère for 7 days. The suspension was filtered through Celite and the resulting solution was concentrated and co-evaporated with toluene. The crude product was used in the next step without further purification.
[0358] To a solution of 1,2,4-triazole (2.7 g, 39.7 mmol) in anhydrous CH3CN (20 mL) at 0°C was added f-Pi^NEt (7.6 mL, 43.6 mmol). Once ail the triazole was dissolved, POC13 (1.11 mL, 11.9 mmol) was added. The mixture was stirred at 0°C for 2 h. The solution was transferred into the flask containing the lactam (4.96 mmol). The resulting solution was heated in an oil bath at 80°C for 16 h. The viscous mixture was cooled with an ice bath and the solvent evaporated. Diluted with EtOAc and water was added. It was extracted with EtOAc five times. The combined extracts were washed with brine and dried over MgSO.*. Filtration and concentration gave a crude product, which was used directly in the next reaction. MS: [M+l]=266.
[0359] A solution of KO/Bu (1.11 g, 9.92 mmol) in DMF (10 mL) was cooled to -50°C
under a nitrogen atmosphère. Ethyl isocyanoacetate (1.2 mL, 10.9 mmol) was added slowly. The mixture was stirred between -60°C to -40°C for 1 h. The above crude 1,2,4triazolo intermediate from step 4 (4.96 mmol) in DMF (5 mL) was added slowly. The mixture was allowed to warm to room température over 16 h. Saturated NH4CI aqueous solution was added and it was extracted with EtOAc three times. The combined extracts were washed with brine (3x) and dried over MgSO4· Filtration and concentration gave a crude product. Chromatography (RediSep 24 g silica-gel column, eluted with 70% EtOAc in Hexanes) to give 296 mg (20% yield for 4 steps) of product. MS: [M+1] =310. [0360] To a solution of ester dérivative (260 mg, 0.84 mmol) in THF (6 mL), water (5 mL) and MeOH (1 mL) was added LiOH (117 mg, 4.85 mmol). The solution was stirred at room température for 3 h. The solution was concentrated and the crude material was acidified with IN HCl until pH 3-4. The solid was collected by multiple filtrations to give 178 mg (75% yield) of the desired product. MS: [M+1] = 282.
[0361] To a suspension of acid (80 mg, 0.28 mmol) in THF (2 mL) was added CD1 (50 mg, 0.31 mmol). The suspension was heated at 65 C for 3 h. LCMS indicated that the reaction was incomplète. More CD1 (10 mg) was added and the solution heated for another hour. The solution was cooled down to room température and a NH4OH solution (1 mL) was added. The solution was stirred for one hour. The solid was collected by filtration to give 33 mg (42%) ofthe Compound 117 as the desired product as a white solid. MS: [M+1] = 281. H'NMR (MeOD4) δ 8.1 (IH, s), 7.9 (IH, s), 7.73 (3H, m), 7.07 (2 H, s), 2.40 (3H, s).
Example 59: Synthesis of Compound 115:
115 [0362] To a suspension of Compound 117 (8 mg, 0.029 mmol) and triethylamine (8 pL; 0.058 mmol) in THF (1 mL) was added trifiuoroacetic anhydride (8 pL; 0.058 mmol). The reaction mixture was stirred at room température for 16 h. LCMS indicated only 30% conversion. More trifiuoroacetic anhydride (30 pL) and triethylamine (30 pL)
224 were added. The solution became clear and stirred for another hour. The reaction was quenched with MeOH. The solvent was evaporated and the crude material was purified by prep TLC (eluting system: 70% EtOAc in Hexanes) to give 6.6 mg (83%) of the Compound 115. MS: [M+1] = 263. H!NMR(CDCh)δ 8.17(IH, d,J= 7 Hz), 7.88 (IH, s), 7.67 (3H, m), 2.46 (3 H, s).
Example 60: Synthesis of Compound 127:
127 [0363] To a suspension of Compound 115 (16 mg, 0.06 mmol) în EtOH (0.8 mL) and water (0.2 mL) was added hydroxylamine hydrochloride (6 mg, 0.09 mmol) and potassium carbonate (12 mg, 0.09 mmol). The suspension was heated at 80 °C for 16 h. The solution was diluted with EtOAc and washed with water. Aq. Layer was separated and extracted with EtOAc (3x). The combined organic phases were washed with brine, dried over MgSO4. Filtration and concentration gave 12.2 mg (67% yield) of the desired product. MS: [M+1] = 296.
[0364] A suspension of oxime (10 mg, 0.034 mmol) in acetic anhydride (0.5 mL) was heated at 110 C for 1 hour. Then, the solution was heated at 130 C for 1 hour. Finally, the température was increased to 140 °C and heated for another 2 h. The réaction mixture was cooled down and EtOH (imL) was added to the reaction mixture which was heated for 16 h at 80 °C. The solvent was evaporated and the crude material was purified by prep TLC (eluting system: EtOAc) to give 6.1 mg (56% yield) ofthe desired product Compound 127. MS: [M+1] =320). H’NMR(CDCh) δ 8.16 (IH, m), 7.92 (IH, s), 7.65 (3H, m), 2.68 (3 H, s), 2.46 (3 H, s).
Example 61: Synthesis of Compound 133:
225
133 [0365] To a solution of isobutyric acid ( 19 pL, 0.2 mmol) in THF (0.5 mL) was added CDI (10 mg, 0.062 mmol). The solution was stirred at room température for 2 h. The solution was then transferred into a vial contaîning the oxime dérivative described above (12 mg, 0.041 mmol) and heated at 70 °C for 2 h. LCMS indicated that the réaction was incomplète. Another batch of reagent (isobutyric acid and CDI) was prepared and added to the reaction mixture which was heated at 70 °C for another hour. LCMS indicated that ail starting material was consumed. The solvent was evaporated and the crude material was suspended in isobutyric acid (1 mL) and heated at 130 °C for one hour. The solvent was evaporated and the crude material was purified by Prep TLC (eluting system: 70% EtOAc in Hexanes) to give 6.7 mg (71%) ofthe desired product Compound 133. MS: [M+1] = 348.
H'NMR (CDCIj) δ 8.16 (1H, m), 7.92 (1H, s), 7.65 (3H, m), 3.32 (1H, m), 2.46 (3H, s), 1.5 (6H, d, J= 7 Hz).
Example 62: Synthesis of Compound 126:
126 [0366] Acetamide oxime was azeotroped three times in toluene before use. To a suspension of acetamide oxime (24 mg, 0.32 mmol) in THF (1 mL) was added NaH 60% in oil dispersion (13 mg, 0.32 mmol). The suspension was stirred at room température for 15 min. Compound 3 (50 mg, 0.16 mmol) was added. The vial contaîning the ester was rinsed with DMF (1 mL) which was added to the reaction mixture. The resulting brown suspension was stirred at room température for 30 min then heated at 70 °C for 2 h. The suspension was quenched with water and the solution was kept in the fridge ovemight.
226
The solid was collected by multiple filtrations to give 16 mg (31% yield) of product
Compound 126. MS: [M+1] = 320. H’NMR (CDCb) δ 8.18 (1H, m), 7.94 (1H, s), 7.67 (3H, m), 2.51 (3H, s), 2.46 (3H, s).
Example 63; Synthesis of Compound 125:
125 [0367] To a suspension of the carboxylic acid derived from Compound 3 (30 mg, 0.11 mmol), Ν,Ο-dimethylhydroxylamine hydrochloride (13 mg, 0.13 mmol), 1hydroxybenzotriazole hydrate (17 mg, 0.11 mmol) and triethylamine ( 46 pL, 0.33 mmol) in THF (0.3 mL) and DCM (0.3 mL) was added l-(3-dimethylaminopropyl)-3ethylcarbodiimide hydrochloride (32 mg, 0.17 mmol). The solution was stirred at room temperature for 16 h. The reaction mixture was quenched with a saturated ammonium chloride solution and extracted with EtOAc (3x). The combined extracts were washed with brine and dried over MgSO<. Filtration and concentration gave 31.2 mg (88% yield) of an orange solid which was used in the next step without further purification. MS: [M+l] = 325.
[0368] To a solution of above Weinreb amide dérivative (31.2 mg, 0.093 mmol) in THF (0.5 mL) cooled at -78 °C was added a solution of 3 M ethyl magnésium bromide (0.31 mL, 0.93 mmol). The reaction mixture was stirred below -10 °C over a period of 60 min. Then, it was quenched with a saturated ammonium chloride solution and extracted with EtOAc (2X). The combined extracts were washed with brine and dried over MgSO4. Filtration and concentration gave a crude product. Chromatography (RediSep 4 g silicagel column, eluted with 80% EtOAc in Hexanes) to give 11,1 mg (41% yield) of product Compound 125. MS: [M+1] =294. H*NMR (CDCh) δ 8.15 (1H, m), 7.76 (1H, s), 7.65 (3H, m), 3.08 (2H, q, J= 7 Hz), 2.44 (3H, s), 1.22 (3H, t, J= 7 Hz).
Example 64: Synthesis of Compound 132:
132 [0369] To a solution of isobutyronitrile (2.6 mL; 29 mmol) in EtOH (30 mL) and water (10 mL) was added hydroxylamine hydrochloride (2.01 g, 29 mmol) and potassium carbonate (4 g, 29 mmol). The resulting suspension was heated at 80 °C for 16 h. The solvent was removed under vacuo. The residue was co-evaporated with toluene. The crude material was washed with EtOH and filtered to remove the sodium chloride. The filtrate was evaporated, co-evaporated with toluene several times and dried under vacuo to give 2 g (69%) of N-hydroxybutyramidine.
[0370] To a suspension of N-hydroxybutyramidine (47 mg, 0.46 mmol) in THF (1 mL) was added NaH 60% in oil dispersion (18 mg, 0.46 mmol). The suspension was stirred at room température for 30 min. Compound 3 (47 mg, 0.15 mmol) in THF (lmL) was added. The resulting suspension was stirred at room température for 30 min then heated at 70 °C for 2 h. After one hour, only 50% conversion was observed. No change was observed after another hour. More reagent (N-hydroxybutyramidine and NaH) as described above was prepared and added to the reaction mixture which was heated for another 40 min. At this point, LCMS showed that the réaction was complété. The suspension was quenched with water. Some MeOH was added to help a complété dissolution, and the solution was extracted with EtOAc (3x). The combined extracts were washed with brine (3x) and dried over MgSO4. Filtration and concentration gave a crude product. Chromatography (RediSep 4 g silica-gel column, eluted with EtOAc) to give 20 mg (38% yield) of product Compound 132. MS: [M+1 ] =348. H'NMR (CDCIj) δ 8.18 (1H, d, J= 8 Hz), 7.93 (1H, s), 7.69 (3H, m), 3.22 (1H, m), 2.46 (3H, s), 1.43 (6H, d, J= 9.5 Hz)
Example 65: Synthesis of Compound 161:
228
161 [0371] Το a solution of acid derived from Compound 3 (90 mg, 0.32 mmol) in DMF (2 mL) cooled with an ice bath was added NaHCOj (108 mg, 1.28 mmol) followed by NBS (114 mg, 0.64 mmol). The solution was stirred at room température for 18 h. The reaction mixture was diluted with water and extracted with EtOAc (3X). The combined extracts were washed with brine (2x) and dried over MgSO4. Filtration and concentration gave a crude product. Chromatography (RediSep 4 g silica-gel column, eluted with EtOAc) to give 54 mg (53% yield) of product. MS: [M+1 ]=316.
[0372] To a solution of bromide dérivative (30 mg, 0.1 mmol) in dioxane (1 mL) and triethylamine (I mL) was added TMS-acetylene (71 gL, 0.5 mmol), Cul (2 mg, 0.01 mmol) and PdCI2(PPh3)2 (7 mg, 0.01 mmol). The solution was heated at 110 °C for 6 h. More Pd catalyst (7mg) and TMS-acetylene (0.2 mL) were added and the reaction mixture heated for an additional 12 h. At this time, LCMS showed about 80% conversion. More Pd catalyst (7mg) and TMS-acetylene (0.2 mL) were added and the reaction mixture heated for an additional 12 h. LCMS showed complété conversion. The reaction mixture was then diluted with water and extracted with EtOAc (3x). The combined extracts were washed with brine (2x) and dried over MgSOj. Filtration and concentration gave a crude product. Chromatography (RediSep 4 g silica-gel column, eluted with 70% EtOAc in Hexanes) to give 23 mg (69% yield) of product. MS: [M+I] =334.
[0373] To a solution of alkyne dérivative (23 mg, 0.069 mmol) in MeOH (0.6 mL) and
H2O (0.2 mL) was added KOH (4 mg, 0.076 mmol) at 0 C. The solution was let warm to room température over 16 h. The reaction mixture was diluted with a saturated aqueous ammonium chloride solution and extracted with EtOAc (2X). The combined extracts were washed with brine (2x) and dried over MgSO4. Filtration and concentration gave a crude product which was purified by prep TLC (eluting system: 80% EtOAc in Hexanes) togive 8.1 mg (45% yield) of product Compound 161. MS: [M+l]=262. H’NMR (CDCh) 6 8.13 (1H, m), 7.76 (IH, s), 7.62 (3H, m), 4.09 (2H, bs), 3.28 (1H, s), 2.44 (3H,
s)·
Example 66: Synthesis of Compound 146:
229
146 [0374] To a solution of 3-amino-2-methylacrolein (65 mg, 0.76 mmol) in anhydrous THF (2 mL) was added NaH 60% in oil dispersion (30 mg, 0.76 mmol). The suspension was stirred at room température for 15 min. Compound 115 (50 mg, 0.19 mmol) was added and the reaction mixture was heated at 65°C for 3 h. The reaction mixture was cooled down with an ice bath and water was added. The reaction mixture was stored in the fridge ovemight. The solid was collected by filtration to give 27.5 mg (44% yield) of a white solid Compound 146. MS: [M+1] = 330. h'NMR (CDCb) δ 8.66 (2H, s), 8.15 (1H, m), 7.89 (1H, s), 7.65 (3H, m), 2.44 (3H, s), 2.36 (3H, s).
Example 67: Synthesis of Compound 153:
153 [0375] To a suspension of acid derived from Compound 3 (30 mg, 0.11 mmol) in dichloroethane (0.2 mL) was added thionyl chloride (1 mL; 13.8 mmol) and DMF (20 pL). The resulting solution was heated at 70 °C for 1 hour. The solvent was removed. The crude material was dried under vacuo. The crude material was suspended în isopropanol (2 mL) and stirred at room température for 16 h. The solvent was evaporated, coevaporated with methanol and the crude material was purified by prep TLC (eluting System: EtOAc) to give 7.2 mg (21% yield) of the product Compound 153. MS: [M+1] =324. H* NMR (CDCb) ô 8.15 (1H, d, J= 8 Hz), 7.81 (1H, s), 7.64 (3H, m), 5.32 (1H, q, J= 7 Hz), 2.45 (3H, s), 1.43 (6H, d, J= 7Hz).
230
Scheme 22
Cempwid 144
CCH
TriflourwcB'icarhydnd·
RiCFÿCtfTvawtf IM
fornita tod COI propionte »dd
CCH phnHcadd CU
R»HC«TvaiMri 111
R.BCOnvow* 1M
R · t-BuCompourid 1M
Example 68: Synthesis of Compound 116:
[0376] An altemate route to the nîtrile-substitutcd imidazole dérivatives was also implemented. As an example, Compound 116 was prepared from imino-derivative as shown in Scheme 22. A solution of isocyanoacetonitrile (206 mg, 3.12 mmol) in DMF (7 mL) was cooled to -50°C under a nitrogen atmosphère. KO/Bu (320 mg, 2.85 mmol) was added. The mixture was stirred at -50°C for 1 h. The tmîno dérivative (prepared in
231 identical fashion to the imino dérivative shown above in Scheme 21) (350 mg, 1.24 ' mmol) was added slowly at -50 °C. The mixture was allowed to warm to room température over 16 h. Saturated NH4CI aqueous solution was added and it was extracted with EtOAc three times. The combined extracts were washed with brine (3x) and dried over MgSO4. Filtration and concentration gave a crude product. Chromatography (RediSep 12 g silica-gel column, eluted with 70% EtOAc in Hexanes) to give 230 mg (70% yield) ofthe product Compound 116. MS: [M+1] =281. H'NMR (CDCh) δ 7.92 (1H, dd, J= 3, 8.5 Hz), 7.81 (1H, s), 7.61 (1H, dd, J= 4.5, 9 Hz), 7.38 (1 H, m), 2.47 (3H, s).
Example 69: Synthesis of Compound 145:
145 [0377] To a suspension of cyanide dérivative Compound 116 (50 mg, 0.18 mmol) in EtOH (1.6 mL) and water (0.4 mL) was added hydroxylamine hydrochloride (17 mg, 0.24 mmol) and potassium carbonate (28 mg, 0.2 mmol). The suspension was heated at 80 °C 15 for 30 min then cooled down to room température. A solid precîpitate was collected by filtration to give 37.8 mg (68% yield) of the desired amino oxime product, [M+1] = 314.
[0378] A suspension of amide oxime (10 mg, 0.032 mmol) in acetic anhydride (0.5 mL) was heated at 140 C for 4 h. The réaction mixture was cooled down and EtOH (1 mL) was added to the reaction mixture which was heated for 16 h at 80 °C. The solvent was evaporated and the crude material was purified by prep TLC (eluting system: EtOAc) to give 6.6 mg (61% yield) ofthe desired product Compound 145. MS: [M+1] = 338.
H‘NMR (CDCh) δ 7.91 (1H, dd, J= 3.5, 8.5 Hz), 7.89 (1H, s), 7.65 (1H, dd, J= 5.5,10 Hz), 7.35 (1H, m), 2.69 (3H, s), 2.45 (3H, s).
232
Example 70: Synthesis of Compound 149:
149 [0379] To a solution of isobutyric acid (30 pL, 0.32 mmol) in THF (0.5 mL) was added CDI ( ! 6 mg, 0.096 mmol). The solution was stirred at room température for 2 h. The above amide oxime dérivative (10 mg, 0.032 mmol) was added and the reaction mixture was heated at 70 C for 45 min. The solvent was evaporated and the crude material was suspended în isobutyric acid (I mL) and heated at 130 °C for 3 h. The solvent was evaporated and the crude material was purified by Prep TLC (eluting system: 80% EtOAc in Hexanes) to give 10.6 mg (91%) ofthe desired product Compound 149. MS: [M+1] =
366. H'NMR (CDCh)S 7.90 (IH, dd, J= 3.5, 9 Hz), 7.89 (IH, s), 7.66 (IH, dd, J= 4.5,
8.5 Hz), 7.36 (IH, m), 3.32 (IH, q, J= 6.5 Hz), 2.46 (3H, s), 1.49 (6H, d, J= 8 Hz).
Example 71: Synthesis of Compound 150:
150 [0380] A suspension of the above amide oxime ( 10 mg, 0.032 mmol) in trifluoroacetic 15 anhydride (0.5 mL) was heated under reflux for 10 min. The solvent was evaporated and the crude material was purified by Prep TLC (eluting system: 80% EtOAc in Hexanes) to give 11.8 mg (94%) ofthe desired product Compound 150. MS: [M+1] = 392. H*NMR (CDCIj) δ 7.92 (2H, m), 7.69 (IH, dd, J= 5.5, 9.5 Hz), 7.39 (IH, m), 2.45 (3H, s).
Example 72: Synthesis of Compound 151:
151 [0381] To a solution of formic acid (12 pL, 0.32 mmol) in THF (0.5 mL) was added CDI (16 mg, 0.096 mmol). The solution was stirred at room température for 2 h. The above amide oxime dérivative ( 10 mg, 0.032 mmol) was added and the reaction mixture was heated at 70 °C for 45 min. The solvent was evaporated and the crude material was suspended în formic acid (I mL) and heated at 60 °C for 3 h. The solvent was evaporated and the crude material was purified by Prep TLC (eluting system: 80% EtOAc in Hexanes) to give 2.1 mg (20%) ofthe desired product Compound 151. MS: [M+1] =
324. H'NMR (CDCb) δ 8.83 (IH, s), 7.92 (IH, dd, J= 3.5, 8 Hz), 7.91 (IH, s), 7.65 (IH, dd, J= 4.5,9Hz), 7.37 (1 H, m), 2.45 (3H, s).
Example 73: Synthesis of Compound 155:
155 [0382] To a solution of propionîc acid (22 pL, 0.29 mmol) in THF (0.5 mL) was added 15 CDI (14 mg, 0.087 mmol). The solution was stirred at room température for 1 hour. The above amide oxime dérivative (10 mg, 0.032 mmol) în THF (0.5 mL) was added and the reaction mixture was heated at 70 °C for 90 min. The solvent was evaporated and the crude material was suspended in propionîc acid (1 mL) and heated at 130 °C for 1 h. The solvent was evaporated and the crude material was purified by Prep TLC (eluting system: 20 80% EtOAc in Hexanes) to give 9.4 mg (94%) of the desired product Compound 155.
MS: [M+1] = 352. H'NMR (CDCh) δ 7.91 (IH, dd, J= 2,8.5 Hz), 7.88 (IH, s), 7.65 (IH.
234 dd, J= 6,9.5 Hz), 7.36 (1H, m), 3.01 (2H, q, J= 8.5 Hz), 2.46 (3H, s), 1.48 (3H, t, J= 8.5 Hz).
Exampie 74: Synthesis of Compound 160:
160 [0383] To a solution of pivalic acid (30 mg, 0.29 mmol) in THF (0.5 mL) was added CDI (14 mg, 0.087 mmol). The solution was stirred at room température for 1 hour. The above amide oxime dérivative (10 mg, 0.032 mmol) in THF (0.5 mL) was added and the reaction mixture was heated at 70 °C for 90 min. The solvent was evaporated and the crude material was suspended in acetic acid (1 mL) and heated under reflux for 3 h. The solvent was evaporated and the crude material was purified by Prep TLC (eluting system: 80% EtOAc in Hexanes) to give 7.4 mg (67%) ofthe desired product Compound 160. MS: [M+1] = 380. H*NMR (CDCIj) δ 7.90 (1H, dd, J= 2.7, 9 Hz), 7.88 (1H, s), 7.65 (IH, dd, J= 4.5,9 Hz), 7.35 (1H, m), 2.47 (3H, s), 1.53 (9H, s).
Example 75: Synthesis of Compound 143:
143 [0384] A solution of KOfBu (40 mg, 0.36 mmol) in DMF (3 mL) was cooled to -50 °C under a nitrogen atmosphère. ρ-Tolueneslfonylmethyl isocyanide (76 mg, 0.39 mmol) was added. The mixture was stirred at -50 °C for 1 h. The imino-derivative from Scheme 22 (50 mg, 0.18 mmoi) was added and the mixture was allowed to warm to room température over 16 h. Saturated NH4CI aqueous solution was added and it was extracted
with EtOAc five times. The combined extracts were washed with brine (3X) and dried over MgSO4. Filtration and concentration gave the crude product. Chromatography (RediSep 4 g silica-gel column, eluted with 70% EtOAc in Hexanes) followed by a prep TLC (eluting system: 30% EtOAc in DCM) to give 22.2 mg (30% yield) of a white solid 5 Compound 143. MS: [M+1] =410.
H'NMR (CDCh) δ 7.91 (2H, d, J= 8 Hz), 7.87 (IH, dd, J= 2.5, 8.5 Hz), 7.74 (IH, s), 7.65 (IH, dd, J= 5.5,9 Hz), 7.34 (3 H, m), 2.50 (3H, s), 2.42 (3H, s).
Example 76: Synthesis of Compound 144:
144 [0385] To 3-ethoxymethacrolein (100 mg, 0.88 mmol) was added 7 N ammonia in methanol (1.3 mL, 8.8 mmol). The solution was stirred at room température for 16 h. The solvent was evaporated and the crude yellow solid corresponding to 3-amino-2methylacrolein was used in the next step without further purification.
[0386] To a solution of 3-amino-2-methylacroIein (7 mg, 0.087 mmol) in anhydrous
THF (1 mL) was added NaH 60% in oil dispersion (6 mg, 0.16 mmol). The suspension was stirred at room température for 15 min. The cyanide dérivative (22 mg, 0.079 mmol) in THF (ImL) was added and the reaction mixture was heated at 65 °C for 1 hour. As described above, a new batch of reagents was prepared with 3-amino-2-methylacrolein (20 mg) and NaH (20 mg) in THF (1 mL), and added to the reaction mixture which was heated at 65 °C for another hour. LCMS indicated completion of the reaction. The reaction mixture was quenched with methanol. The solvent was evaporated. The crude material was suspended in water and a solid was collected by filtration to give 5.2 mg (19% yield) ofa light red solid Compound 144. MS: [M+1] = 348. H*NMR (CDCIj) δ 8.67 (2H, s), 7.90 (IH, d, J=9.5 Hz), 7.85 (IH, s), 7.65 (IH, dd, J= 4.5,9 Hz), 7.34(IH, 25 m), 2.44 (3H, s), 2.36 (3H, s).
236
Scheme 23
Example 77: Synthesis of Compound 121:
121 [0387] To a solution of 1,2,4-trîazole, (2.03 g, 29.4 mmol) in anhydrous CHjCN (20 mL) at 0°C was added f-Pr2NEt (5.6 mL, 32.4 mmol). Once ail the triazole was dissolved, POClj (0.82 mL, 8.8 mmol) and compound 16* (I g, 3.68 mmol) were added. The mixture was stirred at 0 °C for 2 h. The resulting solution was heated in an oil bath at 80°C for 16 h. The mixture was cooled with an ice bath, diluted with EtOAc, and water was added. It was extracted with EtOAc three times. The combined extracts were washed with brine and dried over MgSOj. Filtration and concentration gave 1.05 g (88% yield) of an orange solid which was used directly in the next step. MS: [M+1] =324, [0388] A solution of KO/Bu (696 mg, 6.2 mmol) in DMF (15 mL) was cooled to -50°C
237 under a nitrogen atmosphère. Ethyl isocyanoacetate (0.75 mL, 6.8 mmol) was added slowly. The mixture was stirred at -50 °C for 1 h. The above crude product from step 1 (1 g, 3.1 mmol) was added and the mixture was allowed to warm to room température over 18 h. Saturated NH4CI aqueous solution was added and it was extracted with EtOAc eight times. The combined extracts were washed with brine (3X) and dried over MgSOj. Filtration and concentration gave the crude product. Chromatography (RediSep 24 g silica-gel column, eluted with 70% EtOAc in Hexanes) to give 950 mg (83% yield) of product. MS: [M+1] = 368.
[0389] To a solution of diester (200 mg, 0.54 mmol) in anhydrous THF (4 mL) stirred at room température under a nitrogen atmosphère was added LÎBH4 (2 M în THF, 0.66 mL, 1.3 mmol). The reaction mixture was stirred under a nitrogen atmosphère for 24 h. A mixture of EtOAc/EtOH (3 mL/3 mL) was added to the reaction mixture and it was concentrated. The residue was taken up in MeOH and silica gel was added. After volatile solvents were evaporated, the solid was loaded onto a RediSep 4 g silica-gel column. The desired product was eluted with 10:1 v/v CH2C12/MeOH. The diol was obtained as a solid (60 mg, 39% yield). MS: [M+1] = 284.
[0390] The diol (60 mg, 0.21 mmol) was suspended in 5 mL of HBr 33% in AcOH and heated at 80°C for 18 h. The solution was cooled down with an îce bath and diluted with EtOAc. Slowly, a saturated aqueous NaHCO3 solution was added. The solution was extracted with EtOAc (3x), and the combined organic phases were washed with brine, dried over MgSO4. Filtration and concentration gave a crude product which was used in the next step without further purification. MS: [M+1] = 408.
[0391] To a solution of dialkyl bromide dérivative (0.21 mmol) in EtOAc (10 mL) and MeOH (10 mL) was added wet 10% Pd/C (catalytic amount) and the resulting suspension was stirred under a hydrogen atmosphère for 60 h. The suspension was filtered through Celite and the resulting solution was concentrated. The crude product was purified by multiple prep TLC (eluting system: 3% MeOH in EtOAc) to give 6.2 mg (12% yield over 2 steps) ofthe desired product Compound 121. MS: [M+1] = 252. h'NMR (CDCIj) δ 8.09 (IH, m), 7.74 (IH, s), 7.56 (3H, m), 7.90 (2H, m), 2.42 (3H, s), 2.29 (3H, s).
Example 78: Synthesis of Compound 135:
135 [0392] Compound 135 was synthesized în an analogous manner to Compound 121 as follows: To a solution of 1,2,4-triazole (952 mg, 13.8 mmol) in anhydrous CHaCN (20 mL) at 0°C was added r-P^NEt (2.6 mL, 15.2 mmol). Once ali the triazole was dissolved,
POC13 (0.45 mL, 4.8 mmol) and the lactam ester (1 g, 3.45 mmol) was added. The mixture was stirred at 0°C for 2 h. The resulting solution was heated in an oil bath at 80°C for 16 h. The mixture was cooled with an ice bath, diluted with EtOAc, and water was added. It was extracted with EtOAc three times. The combined extracts were washed with brine and dried over MgSO4. Filtration and concentration gave 1.03 g (87% yield) of an orange solid which was used directly in the next step. MS: [M+l]=342.
A solution of KO/Bu (658 mg, 5.9 mmol) in DMF (15 mL) was cooled to -50°C under a nitrogen atmosphère. Ethyl isocyanoacetate (0.71 mL, 6.5 mmol) was added slowly. The mixture was stirred at -50°C for 1 h. The above crude product from step 1 (1 g, 2.9 mmol) was added and the mixture was allowed to warm to room température over 18 h.
Saturated NHtCI aqueous solution was added and it was extracted with EtOAc eight times. The combined extracts were washed with brine (3X) and dried over MgSO4. Filtration and concentration gave the crude product. Chromatography (RediSep 24 g silica-gel column, eluted with 70% EtOAc in Hexanes) to give 1.02 g (90% yield) of product. MS: [M+1] - 386.
[0393] To a solution of dîester (600 mg, 1.56 mmol) in anhydrous THF (8 mL) stirred at room température under a nitrogen atmosphère was added LiBU» (2 M in THF, 3.1 mL, 6.24 mmol). The reaction mixture was stirred under a nitrogen atmosphère for 24 h. A mixture of EtOAc/EtOH (10 mL/10 mL) was added to the reaction mixture and it was concentrated. The residue was taken up in MeOH and silica gel was added. After volatile solvents were evaporated, the solid was loaded onto a RediSep 12 g silica-gel column. The desired product was eluted with 10:1 v/v CHiCh/MeOH. The diol was obtained as a solid (187 mg, 40% yield). MS: [M+1] = 302.
[0394] The diol (80 mg, 0.27 mmol) was suspended in 7 mL of HBr 33% in AcOH and • 239 heated at 80°C for 48 h. The solution was cooled down with an ice bath and diluted with EtOAc. Slowly, a saturated aqueous NaHCOj solution was added. The solution was extracted (3x) and the combined organic phases were washed with brine, dried over MgSO-i. Filtration, concentration and co-evaporation with toluene gave 100 mg (88% yield) of a beige solid which was used in the next step without further purification. MS:
[M+1] = 426.
[0395] To a solution of dialkyl bromide dérivative (70 mg, 0.16 mmol) in EtOAc ( 10 mL) and MeOH (10 mL) was added 10% Pd/C (catalytic amount) and the resulting suspension was stirred under a hydrogen atmosphère for 48 h. The suspension was filtered through Celite and the resulting solution was concentrated. The crude product was purified by multiple prep TLC (eluting system 1:75% EtOAc in Hexanes; eluting system 2:5% MeOH in EtOAc; eluting system 3: EtOAc) to give 4.1 mg (10% yield) of the desired product Compound 135. MS: [M+1] =270. H'NMR (CDClj) δ 7.84 (IH, dd, J= 2.5, 9 Hz), 7.70 (1Η, s), 7.54 (IH, dd, J= 5, 8 Hz), 7.30 (1Η, m), 2.42 (3H, s), 2.28 (3H,
s).
Example 79: Synthesis of Compound 134:
134 [0396] To a suspension of dialkyl bromide dérivative described in Scheme 23, R = H, (30 mg, 0.074 mmol) in EtOH (ImL), and heated at 80 °C was added a freshly prepared
NaOEt 2Μ solution (75 pL, 0.15 mmol). The solution was heated for 10 min. The solvent was evaporated. The crude material was suspended in EtOAc and filtered. The filtrate was concentrated and purified by prep TLC (eluting system: EtOAc) to give 3.1 mg (12% yield) ofthe desired product Compound 134. MS: [M+1] = 340.
240
Scheme 24
Ο,Η
EDC,H081, DCM (EtOjjPtOJCI, THF
CNCHjCOjEt. THF
Cl
1) UOH
OM·
R’CONHNHj E!jN PhCI
O
DIPEA. DCM
CDI/R-COjH
CMjOPtt oompowrt 157
R CH». R1 a CHjOPtl MfnpûuM IM R kPr, R1 · CHjOPtt ·Φ«ηρ«υη6 14 f
< ,c£1)UOH aCDt/NHfOH SJTFFA
ΝΗΛΗ / KsCO,
Exemple 80: Synthesis of Compound 137:
[0397] To a solution of 5-fluoro-2-nitrobenzoic acid (6.6g, 35.66 mmol) in dichloromethane (100 mL) were added DIPEA (9.22 g, 71.3 mmol), HOBt (6.0 g, 39.2 mmol) and EDCI (10.2 g, 53.5 mmol). After about 15 min stirring, to the reaction mixture was added a solution of 2,4-dimethoxybenzyl amine (5.96 g, 35.66 mmol) in dichloromethane (50 mL) dropwise under nitrogen atmosphère. The resulting mixture
241 was stirred under nitrogen atmosphère at room température for 16 h. The reaction mixture was washed successively with IN HCl (100 mL), sat. NaHCOj (100 mL) and brine (100 mL). The organic phase was then dried over MgSQi. Filtration and solvent removal în vacuo afïorded a yellowish solid, wt: 9.3g (78%). MS: [M+1] = 335.
[0398] To the nitro benzene analog (9.3 g, 27.8 mmol) suspended and stirred in a solvent mixture of HOAc/THF/MeOH/H2O (25/100/50/25 mL) at RT was added Zn powder. The mixture was heated to 70°C for 20 hr., cooled, and filtered. Solid was rinsed with THF, and the combined filtrate was concentrated in vacuo. To the resulting slurry was added sat. NaHCOj slowly and carefully to avoid excessive forming formation 10 until pH reach 7 to 8. The mixture was extracted with EtOAc (3x); combined organic layer washed with brine, and dried over MgSO4. Filtration and solvent removal gave the crude amine product as a dark brown gummy paste, wt: 8.7 g.
[0399] To a solution of the aniline from above (8.7 g) in dichloromethane (150 mL) was added triethylamine (3.37 g, 33.4 mmol). The mixture was cooled with ice bath and treated with bromo acetyl chloride (4.81g, 30.6 mmol) under nitrogen atmosphère. The ice bath was removed and the mixture left stirring for 72 hr. The reaction mixture was concentrated în vacuo, the resulting slurry treated with Et2O (100 mL) and water (100 mL). Product precipitate was collected by filtration, and dried to give 5.6g product as a brown solid. Et2O layer was separated from aq. Layer and diluted with DCM (50 mL), 20 washed with brine, and dried over MgSQ|. Filtration and solvent removal gave 5.3 g additional product as a foamy brown solid. Total wt: 11 g (100%).
[0400] To a solution ofthe bromide (11 g) in DMF (550 mL) was added K2COj (7.1 g, 51.7 mmol). The mixture was heated at 50 °C for 48hrs. The mixture was cooled to room température and the inorganic solid was filtered off. Filtrate was concentrated in vacuo, 25 treated with water/MeOH (60/10 mL), extracted with DCM (3x); combined organic layer was washed with brine and dried over MgSOi. Filtration and solvent removal followed by silica gel column chromatography using 5 to 50% EtOAc in DCM gave 3.2 g (36%) of the 7-member lactam as a brownish solid. MS: [M+1] = 345.
[0401] To the lactam (1.32 g, 3.83 mmol) dissolved and stirred în THF (20 mL) and
DMF (3 mL) at -20°C was added r-BuOK (0.645 g, 5.75 mmol). After 30mîn stirring at 20 °C, diethyl chlorophosphate (1.19 mL, 6.89 mmol) was added dropwise, and the
242 mixture was stirred for 3 h while wamring from -20 to 20 °C. The reaction mixture was cooled to -78 °C and to it was added ethyl isocyanoacetate (0.791 mL, 6.89 mmol), followed by addition of Z-BuOK (0.645 g, 5.75 mmol) and stirring continued ovemight while température reached to RT. The reaction was quenched with saturated NH4CI, extracted with EtOAc (2x); combined organic solution was washed with brine and dried over MgSOi. Filtration and solvent removal gave a crude product which was purified by silica gel column chromatography using 15 to 100% EtOAc in DCM, wt: 0.861 g (47%), as a brown solid. MS: [M+1] = 440.
[0402] To the imidazole ester from above (861 mg) in dichloromethane (5 mL) at 0 °C was added trifluoroacetîc acid (5 mL) followed by trifluoromethanesulfonic acid (0.345 • mL). The mixture was warmed to RT, stirred for 3 h, then concentrated to afford a residue which was dissolved in dichloromethane (50 mL). To which was added sat. NaHCOj (50 mL), followed by 20min stirring. pH of the top aq. Layer was tested basic, and was separated, extracted with DCM (3x); combined DCM solution washed with brine and dried over MgSOi. Filtration and solvent removal gave 0.58g (100%) ofthe lactam as a yellowish solid. MS: [M+1] = 290.
[0403] To lactam (209.1 mg, 0.723 mmol) and N,N-dimethyl-/>toluidine (234.7 mg, 1.74 mmol) stirring in chlorobenzenc (2.5 mL) under nitrogen was added POClj (133.0 mg, 0.867 mmol). The reaction was then heated at 135°C for 2 h. Upon cooling to room température, phenoxy acetic acid hydrazide ( 189.0 mg, 1.08 mmol) was added, followed by D1PEA (0.455 mL). The reaction was stirred at room température for 30 min, then heated at 100°C for 60 min. The reaction mixture was cooled, saturated NH4C! (aq.) was added, and extracted with ethyl acetate three fîmes; combined organic layer was washed with brine, and dried over MgSOi. After filtration and concentration, the product was isolated by ISCO flash column chromatography using 0 to 10% MeOH in EtOAc, wt:
116.7mg (36%) of Compound 137 as a yellowish filmy solid. MS: [M+1] = 420.
243
Exemple 81: Synthesis of Compound 156:
156 [0404] Ethyl ester Compound 137 (244.2 mg, 0.582mmol) in a solvent system of THF/water/MeOH (6.0 mL total, 6/5/1 ratio) was treated with LiOH (69.7 mg, 2.91 mmol) at RT for 4hrs, concentrated in vacuo, acidified to pH~3, and precipitate collected by filtration. After water washing and drying, 179.3 mg (79%) ofthe acid was obtained as a yellowish solid. MS: [M+1] ~ 392.
[0405] To the acid (10.8 mg, 0.0276 mmol) stirring in DCM (0.1ml) at RT was added EDCI (21.3 mg, 0.11 mmol), DMAP (6.7mg, 0.0552mmol) and isopropyl alcohol (13.2 mg, 0.221 mmol). After 12hrs, the reaction was diluted with EtOAc, washed with sat.
NaHCO3; aq. Layer separated and extracted with EtOAc, combined organic layer washed with brine, and dried over MgSÛ4. Filtration and prep. TLC purification ofthe concentrate using 10% MeOH in EtOAc gave 8.7 mg (73%) of the isopropyl ester Compound 156 as a yellowish foamy solid. MS: [M+1 ] = 434.
Example 82: Synthesis of Compound 138:
138 [0406] Acetamide oxime (10.7 mg, 0.144 mmol) was azeotroped four times in toluene, and added to the ethyl ester Compound 137 (9.5 mg, 0.0226 mmol). THF (0.3 mL) was added, followed by NaH 60% oil suspension (4.5 mg, 0.112 mmol). The réaction mixture was stirred at RT for 30 min, then heated at 70°C for 2 h, cooled to RT, and solvent
244 removed in vacuo, water (1.5 mL) added to quench the reaction, stirred for 20 min, and cooled to 4°C. Precipitate was collected by filtration, washed with water, and dried to *give 5.2 mg (59%) ofthe oxadiazole product Compound 138 as a light yellow solid. MS: [M+1] =430.
Example 83: Synthesis of Compound 141:
141 [0407] Compound of Example 83 was synthesized in an analogous synthetic route as that described for Example 82, using isobutyramidoxime in place of acetamide oxime to give the compound of Example 83 as a yellowish solid: MS: [M+1] = 458.
Example 84: Synthesis of Compound 157:
157 [0408] To the acid prepared above in Example 81 (60.2 mg, 0.154 mmol) stirring in DCM (0.7 mL) at RT was added carbonyl diimidazole (49.9 mg, 0.308 mmol). The mixture was stirred for 40 min, then cooled to 0°C, and ammonia (0.112 ml) added, warmed to RT while stirring continued ovemight. The reaction was concentrated, water (8 mL) added, and stirred well for 30 min. Resulting precipitate was collected by filtration, washed with water, and dried to give 51.1mg (85%) ofthe primary amïde as a brownish soiid. MS: [M+1] = 391.
[0409] The amide (51.1 mg) from above was treated with POCI3 (200.8 mg, 1.31 mmol) în 1,4-dioxane (0.9 mL) at 90°C for 14hrs. Upon cooling to RT, the réaction was carefully
245 quenched with sat. NaHCCh (5 mL), stirred for 20 min. Precîpitate was collected by filtration, washed with water, and dried to give 40.9 mg (85%) of nitrile product Compound 157 as a brownish solid. MS: [M+1] = 373.
Example 85: Synthesis of Compound 147:
147 [0410] To the nitrile (45.8 mg, 0.123 mmol) in a round bottom flask was added hydroxylamine hydrochloride (14.5 mg, 0.209 mmol), K2CO3 (22.3 mg, 0.161 mmol), éthanol (0.6 mL), and water (0.15 mL). The reaction mixture was heated at 80°C for 30min, cooled down, and concentrated in vacuo. The resulting slurry was treated with water (1.5 mL), sonicated to help mixing, and stirred at RT for I h before being cooled to 4°C. The resulting precîpitate was collected by filtration, washed with cold water (1 mL), and dried to give 40.8 mg (82%) ofthe adduct as an off-white solid. MS: [M+1] = 406.
[0411] Isobutyric acid (31.4 mg, 0.582 mmol) was treated with carbonyl diimidazole (28.4 mg, 0.175 mmol) in THF (0.5 mL) for 2hrs. The N-hydroxycarboxamide adduct (l 1.8 mg, 0.0291 mmol) was added, and the reaction was stirred at RT for 30 min. More isobutyric acid (0.5 mL) was added and the reaction mixture was heated at 110°C for 16 h, cooled, sat. NaHCCh (8 mL) added, and extracted with EtOAc (3x); combined organic layer washed with brine, and dried over MgSO4. Prep. TLC (5% MeOH in EtOAc) ofthe concentrated filtrate gave 11.2 mg (84%) of the oxadiazole Compound 147 as a white solid. MS: [M+1] = 458.
246
Example 86: Synthesis of Compound 148:
[0412] Compound of Example 86 was synthesîzed in an analogous synthetic route as that described for Example 85, using acetic acid in place of isobutyric acid to give the compound of Example 86 as a white solid: MS: [M+1] = 430.
Example 87: Synthesis of Compound 158:
158 [0413] Compound of Example 87 was synthesîzed in an analogous synthetic route as that described for Example 85, using propionîc acid in place of isobutyric acid to give 10 the compound of Example 87 as a white solid: MS: [M+1] = 444.
Example 88: Synthesis of Compound 159:
[0414] Trifluoroacetic anhydride (196.9 mg. 0.938 mmol) was added to the Nhydroxycarboxamide adduct (19.0 mg, 0.0469 mmol) suspended and stirred in THF (0.2
247 mL) at RT. After 30min stirring, the reaction was heated to 70°C for 1 h, cooled to RT, and diluted with EtOAc (10 mL), to which was added sat. NaHCOj and stirred for 30min. Aq. Layer was separated and extracted with EtOAc (lx); combined organic layer was washed with brine, and dried over MgSO4. Filtration and solvent removal gave a paste to 5 which was added nBuOH (5 ml) and HOAc (0.5 mL). This was heated at 115°C for 16 h, cooled and concentrated in vacuo, diluted with EtOAc, washed with sat. NaHCOj, brine, and dried over MgSO4. Prep. TLC (5% MeOH in EtOAc) ofthe concentrated filtrate gave 11.5 mg (51%) ofthe desired trifluoromethyl oxadiazole analog Compound 159 as a yellowish solid. MS: [M+1] = 484.
Scheme 25
R · CHaO4-F-Ptv compound 183
R CHjOCH,· compound 144
R CHjOCHjPH compound 148
Example 89: Synthesis of Compound 162:
162 [0415] To lactam 62 (503.4 mg, 1.42 mmol) stirring în THF (2.9 ml) and DMF (0.8 mL) at -20°C was added tBuOK (240.2 mg). After 30 min stirring, diethyl chlorophosphate (377.7 mg, 2.12 mmol) was added dropwise, and the reaction mixture was slowly warmed to 8°C in 3 h before being cooled down to -20°C. 2.26 mL (226
248 mmol) of oxadiazole isocyanate (ref. JA/C, 1996,39, 170; prepared as IM THF solution) was added. The reaction mixture was further cooled to -78°C, tBuOK (238.4 mg) was added, and the reaction was slowly warmed to RT ovemight. Sat. NH4CI (5 mL) was added and the mixture was extracted with EtOAc (2x), washed with brine, and dried over MgSO4. Upon filtration and concentration, the product was isolated by silica gel column chromatography using a gradient elution of 0 to 10% MeOH în EtOAc to give 246.0 mg imidazole product as ayellowish solid. MS: [M+1] = 462.
[0416] The imidazole (246.0 mg, 0.533 mmol) obtained above was stirred in DCM (3 ml). Trifluoroacetic acid (3 mL) was added, followed by trifluoromethyl sulfonic acid (160.0 mg, 1.07 mmol). After 3 h stirring, the réaction was diluted wîth DCM (20 mL), washed wîth sat. NaHCOj; aq. Layer was separated and extracted with DCM (2x); combined DCM solution was washed with brine, and dried over MgSO4. Filtration and solvent removal în vacuo gave 208.7 mg of the crude lactam product as a yellowish flaky solid. [M+1] = 312.
[0417] Phosphorous oxychloride (29.9 mg, 0.195 mmol) was added to a solution of the above obtained lactam (22.5 mg, 0.0723 mmol) and N,N-dimethyl-p-toluidine (51.8 mg, 0.383 mmol) stirring în chlorobenzene (0.45 mL) under nitrogen atmosphère. The reaction mixture was heated at I35°C for 3 h, then cooled to RT. Diisopropylethylamine (75.7 mg, 0.586 mmol) and phenoxyacetic hydrazide (50.1 mg, 0.302 mmol) was added, and the reaction mixture was heated at 100°C for 14 h, cooled to RT, and partitioned between sat. NH4CI and EtOAc. Aq. Layer was separated and extracted with EtOAc; combined EtOAc solution was washed with brine, and dried over MgSO4. Upon filtration and concentration, the product Compound 162 was isolated by silica gel column chromatography using a gradient elution ofO to 10% MeOH in EtOAc as ayellowish solid. Wt: 11.8 mg (37%). MS: [M+1] =442.
249
Example 90: Synthesis of Compound 163:
163 [0418] Compound of Example 90 was synthesized in an analogous synthetic route as that described for Example 89, using 4-fluorophenoxyacetic hydrazide in place of phenoxyacetic hydrazide to give the compound of Example 90 as a yellowish solid: MS: [M+1] = 460.
Example 91: Synthesis of Compound 164:
164 [0419] Compound of Example 91 was synthesized in an analogous synthetic route as that described for Example 89, using methoxyacetic hydrazide in place of phenoxyacetic hydrazide to give the compound of Example 91 as a yellowish solid: MS: [M+1] = 380.
Example 92: Synthesis of Compound 165: .
165
250 [0420] Préparation ο f benzyloxy acetic hydrazide: carbony! dîimidazole (1.52 g, 9.39 mmol) was added to benzyloxy acetic acid (1.2 g, 7.22 mmol) stirring in THF (60 mL) at 0°C. Ice bath was removed and the stirring continued for I hr. The resulting cloudy solution was added to hydrazîne (0.927 g, 28.9 mmol) stirring in THF (40 mL) at
RT. After 16hrs, the reaction mixture was concentrated to a slurry, to which was added water (120 mL), extracted with DCM (3x); combined DCM solution washed with brine, and dried over MgSOj. Filtration and solvent removal gave 0.908 g (70%) of the hydrazide as a clear viscous oil. This was azeotroped in toluene a few times before use. Compound of Example 92 was synthesized in an analogous synthetic route as that ! 0 described for Example 89, using benzyloxy acetic hydrazide in place of phenoxyacetic hydrazide to give the compound of Example 92 as a yellowish solid: MS: [M+1] = 456.
Example 93: Synthesis of Compound 166:
[0421] Compound 165 from above (58.5 mg, 0.128 mmol) was treated with 10% Pd-C 15 (catalytic) in EtOAc (4 mL) and MeOH (4 mL) under hydrogen atmosphère for 2 h.
Catalyst was removed by filtration over Celîte. To the filtrate was added conc. HCl (0.89 mL), and the mixture was stirred at RT for 16 h. Excess Na2COj (aq.) was added, and the solution was extracted with EtOAc (2x); combined organic solution was washed with brine, and dried over MgSOj. Prep. TLC ofthe concentrated filtrate using 15% MeOH in 20 EtOAc gave 14.9 mg ofthe primary amide ([M+I] =417) as a yellowish solid. This primary amide was treated with phosphorous oxychloride (54.9 mg, 0.358 mmol) în 1,4dioxane (1 mL) at 90°C for 14 h. Upon cooling, the reaction mixture was diluted with EtOAc, washed with sat. NaHCOj; aq. layer separated and extracted with EtOAc (Ix), combined organic solution was washed with brine, and dried over MgSQi. Prep. TLC of 25 the concentrated filtrate using 5% MeOH în EtOAc gave 5.2 mg of the desired nîtrile product Compound 166 as white needles. [M+1] = 399.
251
Scheme 26
R1 CHjOCHj. compound 16»
R’ » CHjOPh compound 171
R1 · CHjO-6-F-Ptt compound 171
R’ - CHjOEL compound 173
R’ CHjO-2-F-Ph: compound 174
R1 CHjO-2-Q-PTt compound 173
R’ CHjO-3-Pyr compound 17(
R1 * CHjO-l-Napmyl compound 177 R’ * CHjO-3-F-Ph. compound 17»
compound ln |21M)
Example 94: Synthesis of Compound 169:
169 [04221 To lactam 62 (2.23 g, 6.24 mmol) stirring in THF ( 10 mL) and DMF (3 mL) at 5 20°C was added tBuOK (L05 g, 9.36 mmol). After 30 min stirring, diethyl chlorophosphate (1.66 g, 9.36 mmol) was added dropwise, and the reaction mixture was slowly warmed to 8-10°C in 3 h before beîng cooled down to -20°C. 10.0 ml (10.0 mmol) of oxadiazole isocyanate (ref. JMC, 1996,39, 170; prepared as IM THF solution) was added. The reaction mixture was further cooled to -78°C, tBuOK (l.05g, 9.36 mmol) was
252 added, and the réaction was slowly warmed to RT ovemight. Sat. NH4CI (20 mL) was added and the mixture was extracted with EtOAc (3x), washed with brine, and dried over MgSOi. Upon filtration and concentration, the product was isoiated by silica gel column chromatography usîng a gradient elution of 10 to 100% EtOAc in DCM to give 1.07 g (35%) imidazole product as a yellowish foamy solid. MS: [M+1] = 490.
[0423] The imidazole (1.07 g, 2.18 mmol) obtained above was stirred in DCM (11 mL). Trifluoroacetic acid (11 mL) was added, followed by trifluoromethyl sulfonic acid (0.656 g, 4.37 mmol). After 4 h stirring, the reaction was concentrated in vacuo, diluted with DCM (50 mL), washed with sat. NaHCOj; aq. Layer was separated and extracted with DCM (2x); combined DCM solution was washed with brine, and dried over MgSOj. Filtration and solvent removal in vacuo gave 0.872 g of the crude lactam product as a brownish solid. [M+1 ] = 340.
[0424] Phosphorous oxychloride (51.0 mg, 0.333 mmol) was added to a solution of the above obtained lactam (45.0 mg, 0.133 mmol) and N,N-dimethyl-p-toluidine (89.6 mg, 0.663 mmol) stirring in chlorobenzene (0.60 mL) under nitrogen atmosphère. The reaction mixture was heated at 135°C for 3 h, then cooled to RT. Diisopropylethylamine (137.5 mg, 1.06 mmol) and methoxyacetic hydrazide (83.1 mg, 0.798 mmol) was added, and the reaction mixture was heated at 100°C for 4 h, cooled to RT, diluted with EtOAc, washed with sat.NaHCO3, brine, and dried over MgSOd. Upon filtration and concentration, the product Compound 169 was isoiated by silica gel column chromatography using a gradient elution of 0 to 13% MeOH in EtOAc as a brownish solid. Wt: 14.3 mg (26%). MS: [M+1] =408.
Example 95: Synthesis of Compound 171:.
MeO' ”'N
171
253 [0425] Compound of Example 95 was synthesîzed in an analogous synthetic route as that described for Example 94, using phenoxyacetic hydrazide in place of methoxyacetic hydrazide to give the compound of Example 95 as a yellowish solid: MS: [M+1] = 470.
Example 96: Synthesis of Compound 172:
[0426] Compound of Example 96 was synthesîzed in an analogous synthetic route as that described for Example 94, using 4-fluoro-phenoxyacetîc hydrazide în place of methoxyacetic hydrazide to give the compound of Example 96 as a yellowish solid; MS: [M+1] = 488.
Example 97: Synthesis of Compound 173:
MeO
173 [0427] Compound of Example 97 was synthesîzed in an analogous synthetic route as that described for Example 94, using ethoxyacetic hydrazide in place of methoxyacetic hydrazide to give the compound of Example 97 as a yellowish solid: MS: [M+1] = 422.
Example 98: Synthesis of Compound 174:
174 φ 2S4 [0428] Compound of Example 98 was synthesized in an analogous synthetic route as that described for Example 94, using 2-fluoro-phenoxyacetic hydrazide in place of methoxyacetic hydrazide to give the compound of Example 98 as a yellowish solid: MS: [M+1] = 488.
Example 99: Synthesis of Compound 175:
175 [0429] Compound of Example 99 was synthesized in an analogous synthetic route as that described for Example 94, using 2-chloro-phenoxyacetic hydrazide in place of methoxyacetic hydrazide to give the compound of Example 99 as a yellowish solid:
MS: [M+l] = 504.
Example 100: Synthesis of Compound 176:
176 [0430] Préparation of 3-pyrldyloxy acetic hydrazide: a solution of ethyl 3pyridyloxy acetate (0.50 g, 2.76 mmol) and hydrazine (0.31 g, 9.66 mmol) în isopropyl alcohol (35 mL) was heated at 85°C for 30 hr., cooled, and concentrated in vacuo. The resulting white solid was dissolved in small amount of sat. NaCl solution, and extracted with EtOAc repeatedly. The combined organic solution was dried over MgSO4. Filtration and solvent removal gave 177 mg of the desired acetic hydrazide as a white solid. Residual water moisture was removed by azeotroping in toluene.
Compound of Example 100 was synthesized in an analogous synthetic route as that described for Example 94, using 3-pyridyloxy acetic hydrazide in place of
255 tnethoxyacetic hydrazide to give the compound of Example 100 as a yellowish solid: MS: [M+I] = 471.
Example 101: Synthesis of Compound 177*.
[0431] Compound of Example 101 was synthesized in an analogous synthetic route as that described for Example 94, using 1-naphthoxy acetic hydrazide in place of methoxyacetic hydrazide to give the compound of Example 101 as an off white solid: MS: [M+1] = 520.
Example 102: Synthesis of Compound 179:
[0432] Compound of Example 102 was synthesized in an analogous synthetic route as that described for Example 94, using 3-fluorophenoxy acetic hydrazide in place of methoxyacetic hydrazide to give the compound of Example 102 as a yellowish solid: MS: [M+1] = 488.
256
Example 103: Synthesis of Compound 178:
178 [0433] Phosphorous oxychloride (64.8 mg, 0.422 mmol) was added to a solution of the oxadiazolyl imidazole lactam (57.5 mg, 0.169 mmol) and N,N-dimethyl-p-toluidine (114.6 mg, 0.847 mmol) stirring in chlorobenzene (0.70 ml) under nitrogen atmosphère. The reaction mixture was heated at I35°C for 3 h, then cooled to RT.
Dïisopropylethylamine (174.7 mg, 1.35 mmol), r-BuOH (0.3ml), and 2-hydroxy acetic hydrazide (9l.3mg, 1.0Immol) was added. The reaction mixture was stirred at RT for 20min, then warmed at 50°C for one hour followed by 80°C heating for one hour before finally heated at 100°C ovemight. Upon cooling to RT, the reaction was diluted with EtOAc, washed with brine, and dried over MgSO4. Silica gel column chromatography of the concentrated filtrate using a gradient elution of 0 to 20% MeOH in EtOAc gave the desired hydroxymethyl triazole product as a yellowish solid. Wt: 18.1 mg (27%). MS: [M+1] = 394.
[0434] To a solution of hydroxymethyl triazole from above (18.1 mg, 0.046 mmol), cyclopentyl bromide (274.0 mg, 1.84 mmol), and HMPA (16.5 mg, 0.092 mmol) stirring in THF (0.5 ml) was added NaH (60% suspension; 18.4 mg, 0.46 mmol). After 10min, the reaction was heated at I00°C for 6hrs, cooled, quenched with sat. NaHCOj, and extracted with EtOAc (2x), washed with brine, and dried over MgSO4. Prep. TLC of the concentrated filtrate using 8% MeOH in EtOAc gave 5.5mg (26%) of the desired ether Compound 178 as a yellowish solid. [M+1] = 462.
257
Scheme 27
tanpond 1U
Exemple 104: Synthesis of Compound 168:
[0435] To a suspension of benzyl glycinate hydrochloride (5 g, 24.8 mmol) in DCM (100 mL) was added EDC.HC1 (6.2 g, 33.2 mmol) and triethylamine (5.2 mL, 37.2 mmol). The suspension was cooled down to -50 °C then formic acid (1.4 mL, 37.2 mmol) in DCM (5 mL) was added. The reaction mixture was stirred at -50 C for one hour then at 4°C for 3 h. The solution was diluted with IN HCl and extracted with DCM (2x). The combined organic phases were washed with brine and dried over MgSO4. Filtration and concentration gave 3.89 g (81% yield) of formylated glycine as an oil (M+l= 194) [0436] To a solution of formylated glycine dérivative (1 g, 5.2 mmol) in DCM (30 mL) was added triethylamine (3.2 mL, 23 mmol). The solution was cooled down to -50°C and
POClj ( 1.9 mL, 20.8 mmol) was added slowly. The solution was stirred at - 50 C for 10 min, then stirred at room temperature for 40 min. The solution tumed light red-brown. It was diluted with DCM and a 20% sodium carbonate solution (100 mL) was added. The
258 reaction mixture was stirred vigorously for 15 min. The organic phase was separated twice and dried over MgSO4. Filtration and concentration to give the desired benzyl isocyanoacetate in quantitative yield which was used in the next step without further purification.
[0437] To a solution of 1,2,4-triazole (914 mg, 13.2 mmol) in anhydrous CHjCN (20 mL) at 0°C was added /-PriNEt (2.5 mL, 14.6 mmol). Once ail the triazole was dissolved, POCI3 (0.43 mL, 4.6 mmol) was added. The mixture was stirred at 0°C for 2 h. The lactam ester 16’ (1 g, 3.31 mmol) was added. The resulting solution was heated in an oil bath at 80°C for 16 h. The mixture was cooled with an ice bath. Diluted with EtOAc then water was added. Aq. layer was separated and extracted with EtOAc four times. The combined organic extracts was washed with brine and dried over MgSQ». Filtration and concentration gave a light yellow solid which was used directly în the next step (M+l=354).
[0438] A solution of benzyl isocyanoacetate (892 mg, 5.1 mmol) in DMF (10 mL) was cooled to -50°C under a nitrogen atmosphère. KO/Bu (514 mg, 4.6 mmol) was added. The mixture was stirred at -50°C for 1 h. The triazole dérivative prepared above (900 mg. 2.55 mmol) în DMF (5 mL) was added slowly at -50 °C. The mixture was allowed to warm to room température over 16 h. Saturated aqueous NH4CI solution was added and it was extracted with EtOAc three times. The combined extracts were washed with brine (3x) and dried over MgSÛ4. Filtration and concentration gave a crude product.
Chromatography (RediSep 24 g silica-gel column, eluted with 70% EtOAc in Hexanes) to give 886 mg (76% yield) of product (M+1 =460).
[0439] To a solution of benzyl ester dérivative (770 mg, 1.68 mmol) in EtOAc (10 mL) and MeOH (30 mL) was added wet Pd/C (60 mg) and the resulting suspension was stirred under a hydrogen atmosphère for 48 h. The suspension was filtered through Celite and the resulting solution was concentrated. The crude debenzylated product (530 mg, 86%yield) was used in the next step without further purification (M+l= 370).
[0440] To a suspension of acid (530 mg, 1.44 mmol) in DCM (10 mL) was added CD!
(931 mg, 5.75 mmol). The solution was stirred at room température for 2 h. The solution was cooled down with an ice bath and a NH4OH solution (6 mL) was added. The solution was stirred for 30 min and it was concentrated. The solid was collected by filtration and washed with water to give 422 mg (80%) ofthe desired product as a brown solid. (M+l=
369).
259 [0441] To a suspension of primary amïde dérivative (422 mg, 1.15 mmol) in dioxane (10 mL) was added POClj (160 pL, 1.7 mmol). The suspension was heated at 90°C for 2 h. The resulting solution was cooled down with an ice bath and quenched with a saturated aqueous NaHCOj solution. The solid was collected by filtration to give 308 mg (77% yield) ofthe desired cyanîde dérivative. (M+l= 351).
[0442] To a suspension of cyanîde dérivative (150 mg, 0.44 mmol) in EtOH (4 mL) and water (1 mL) was added hydroxylamîne hydrochloride (40 mg, 0.57 mmol) and potassium carbonate (67 mg, 0.48 mmol). The suspension was stirred at room température for 16 h. LCMS indicated about 50% conversion. More hydroxylamîne hydrochloride (40 mg, 0.57 mmol) and potassium carbonate (67 mg, 0.48 mmol) were added, and stirred for another 24 h. The solution was diluted with EtOAc and washed with water. The combined organic phases were washed with brine, dried over MgSOi. Filtration and concentration gave 145 mg (86% yield) of the desired product. (M+l= 384).
[0443[ To a solution of acetic acid (0.22 mL, 3.8 mmol) in THF (5 mL) was added CDI (123 mg, 0.76 mmol). The solution was stirred at room température for 2 h. The solution was then poured into a flask containing the oxime dérivative (145 mg, 0.38 mmol) and heated at 70 C for I hour. The solvent was evaporated and the crude material was suspended în acetic acid (8 mL) and heated at 130°C for one hour. The solvent was evaporated and the crude material was triturated with water to give 134 mg (86%) ofthe desired product (M+1 - 408).
[0444] To a suspension of ester dérivative (50 mg, 0.12 mmol) în THF (I mL) was added lithium aluminum hydride (7 mg, 0.18 mmol). The suspension was stirred at room température for 2 h. LCMS indicated about 70% conversion along some other side products and some remaining starting material. More lithium aluminum hydride (4 mg) was added and the reaction mixture was stirred at room température for another 30 min. The reaction mixture was quenched with IN HCl. The solution was extracted with EtOAc (3x). The combined organic phases were washed with brine, dried over MgSOi. Filtration and concentration gave 20 mg (45% yield) of the desired alcohol product. (M+l= 366).
[0445] To a suspension of alcohol (20 mg, 0.055 mmol) in dioxane (I mL) was added
POBrj (31 mg, 0.11 mmol). The reaction mixture was heated at 110°C for 1 hour. The reaction mixture was cooled down with an ice bath and sat. aq. NaHCOj solution was added. The resulting solution was extracted with EtOAc (3X). The combined organic
260 phases were washed with brine and dried over MgSO4. The solvent was concentrated to give 22 mg (96% yield) of the desired product (M+l= 428).
[0446] To a vial containîng alkyl bromide dérivative (22 mg, 0.052 mmol) was added 3fluorophenol (58 mg, 0.52 mmol) in dioxane (I mL) and potassium carbonate (72 mg,
0.52 mmol). The reaction mixture was heated at 90°C for 1 hour. The réaction mixture was diluted with sat. aq. NaHCO3 solution. The resulting solution was extracted with EtOac (3X). The combined organic phases were washed with brine and dried over MgSO4. Filtration and concentration gave a crude product. Purification by prep TLC (eluting System: EtOAc) to give 5 mg (21% yield) of the desired product Compound 168 (M+l= 460). H’nMR (CDCb) δ 7.87 (IH, s), 7.65 (IH,d, J= 3.5 Hz), 7.57 (1H, d, J= 10
Hz), 7.24 (1H, m), 7.19 (IH, dd, J= 3.5, 9 Hz), 6.77 (1H, dd, J= 2.5, 9.5 Hz), 6.72 (2H, m), 5.26 (2H, s), 3.97 (3H, s), 2.48 (3H. s).
Synthesis of Compounds 215 - 313
Scheme 28
THF, 0-2S*C ° h T
N1OH
MeOH, H2O J h, RT
• 261
Synthesis of Intermediate A (ethyl 15-chloro-9-(methoxymethyl)-2,4,8,l0,l 1-perttaazatetracyclo[11.4.O.O3,é.O8, ’2]heptadeca-1(17), 3,5,9,11,13,15-heptaene-5-carboxylate). [0447] Ethyl bromoacetate (Scheme 28) (10.0 gm, 59.87 mmol) solution in 20.0 mL of anhydrous THF was added dropwise to a solution of (2,4-dimethoxybenzyl)amine (10.0 gm, 59.81 mmol) and triethyl amine (6.06 gm, 59.87 mmol) in anhydrous THF (20.0 mL) at 0 °C under nitrogen atmosphère. The reaction mixture was warmed to room température and stirred ovemight. Brine was added - 100 mL, and the reaction mixture was extracted with ethyl acetate (2 x - 100 mL). Combined extracts were dried over anhydrous MgStliand concentrated under reduced pressure. The purification was performed using combiFlash chromatography. Gradient: 20:80 to 50:50 v/v Ethylacetate:Hexane. 7.6 gm (yield 50.2 %) of the alkylation product was obtained as a colorless liquid. m/z calculated for C13H19NO4 [M+H]+: 254; Obtained: 254.1. The ester (7.5 gm, 29.6 mmol) was dissolved in 40.0 mL of methanol. The reaction mixture was cooled and 2N aq. NaOH (88.82 mmol, 44.0 mL) solution was added dropwise. The reaction mixture was warmed to room température and stirred for 2 h. The reaction mixture was diluted with -75.0 mL of water, cooled in ice bath and neutralized down to ~ 5.0 to 4.5 pH using 2N aq. HCl. The excess water was concentrated under reduced pressure and air streamed to obtain white solid powder. The solid was dissolved in 85:15 v/v, DCMiMeOH (100.0 mL) and filtered, the filtrate was evaporated to obtain
7.1 gm of carboxylic acid as a white powder (Hygroscopic). m/z calculated for
CnHlsNO4 [M+Na]+: 248; Obtained: 248.1.
[0448] The above compound (7.0 gm, 31.08 mmol) and 6.14 gm, 31.08 mmol of 5chloroîsatoic anhydride were mixed in 70.0 mL of p-Xylene and refluxed at 140 °C température for 3 h. The reaction mixture filtered and crude product recrystallized from methanol. 8.5 gm of 7-chloro-4-[(2,4-dimethoxyphenyl)methyl]-2,3,4,5-tetrahydro -1Hl,4-benzodiazepine-2,5-dione was obtained as a white powder (75.8 % yield). m/z calculated for C18H17CIN2O4 [M+H]+: 361; Obtained: 361.1.
[0449] Theabove benzodiazepine-2,5-dîone(4gm, 1 l.l mmol) wasdissolved in
THF/DMF (57.2/12.7 mL) and cooled at-20 °C température. Fînely divided potassium30 tert-butoxide powder (1.9 gm, 16.6 mmol) was added and reaction mixture stirred at -20 °C for 20.0 min. 3.1 gm, 17.7 mmol of diethylchlorophosphate was dropwise added to the reaction mixture at -20 °C and allowed to 0-5 °C for 3 h. The reaction mixture was stirred at ambient température for 10.0 min. 2.1 gm, 18.4 mmol of ethylisocyanoacetate was
W 262 added to the reaction mixture at -20 °C and the reaction mixture was further cooled down to -78 °C. 1.9 gm, 16.6 mmol of finely divided potassîum-tert-butoxide powder was added at -78 °C and the reaction mixture was stirred ovemight by slowly warmîng to ambient température. The reaction mixture was quenched with saturated aq. NH4C!
solution (10 mL), extracted with ethyl acetate (3 x 20 mL). Combined extracts were dried over anhydrous MgSCh and concentrated under reduced pressure. The crude product was recrystallized from ethylacetate to obtain 2.2 gm of ethyl 12-chloro-8-[(2,4dimethoxyphenyl)methyl]-9-oxo-2,4,8-triazatricyclo[8.4.0.0î,6]tetradecal(14),3,5,IO,12-pentaene-5-carboxylate as a white solid. A second crop was obtained from the mother liquor to afford another 3.5 g of product (64% yield).
[0450] The dimethoxybenzyl protecting group was removed by dîssolvîng the above compound (2.2 gm, 4.83 mmol) in DCM (25.0 mL), followed by addition of 25.0 mL of trifluoroacetic acid and 1.45 gm, 9.65 mmol of trifluoromethanesulfonic acid. The reaction mixture was stirred at room température for 90 min. The reaction mixture was neutralized with aq. NaHCCh and the ppts were filtered, washed with water and dried to afford 1.9 gm of ethyl 12-chloro-9-oxo-2,4,8-triazatricyclo[8.4.0.02,6]tetradecal(14),3,5,10,!2-pentaene-5-carboxylate as a solid product. m/z calculated for C14H12CIN3O3 [M+H]+: 306; Obtained: 306.1.
[0451] In the first step, the ethyl !2-chloro-9-oxo-2,4,820 triazatricyclo[8.4.0.03,6]tetradeca-l(!4),3,5,10,12-pentaene-5-carboxylate from above (1.9 gm, 6.21 mmol) was dissolved in 25.0 mL of chlorobenzene, followed by addition of 2.52 gm, 18.64 mmol of4,N,N-trimethylaniline, 1.42 gm, 9.32 mmol of POCI3 and the reaction mixture was refluxed at 135 °C for 2 h. LCMS shows ~ 50 % starting material remained unreacted. 1.68 gm, 12.42 mmol of additional 4,N,N-trimethylaniline and 0.95 gm, 6.21 mmol of POCI3 were further added to the réaction mixture at room température and refluxed at 135 °C for 1 h. LCMS shows ~ 10 % starting material remained unreacted. An additional 0.84 gm, 6.21 mmol of 4,N,N-trîmethylaniline (total 6.0 eq.) and 0.48 gm, 3.11 mmol of POCI3 (total 3 eq.) were further added to the reaction mixture at room température and refluxed at 135 °C for 1 h.
[0452] In the second step, 4.67 gm, 44.75 mmol of methoxyaceticacid hydrazide (total
7.2 eq.), followed by 7.71 gm, 59.66 mmol of Ν,Ν-diisopropylethylamine were added to the reaction mixture at room température and refluxed at 100 °C for 1 h. The reaction mixture was cooled to room température and neutralized with aq. NaHCOj solution (18506
263
25.0 mL). The organic was extracted with ethyl acetate (75 mL x 3), followed by DCM (50.0 mL x 3) and washed with brine. The EtOAc organic layer was separated by filtering the insoluble ppts (0.805 gm pure product) and combined organic layers were dried over anhydrous MgSOj, concentrated under reduced pressure. The crude product was purified by Combiflash chromatography (Mobile phase: 0-10 % MeOH:EtOAc) to yield an additional 0.8 gm ofyellow solid. Total yield for the last two steps of Intermediate A (ethyl 15-chIoro-9-(methoxymethyl)-2,4,8,lO,l 1-penta azatetracycIoJlM.O.O’/.O8,11]heptadeca-l(17)3.5,9,ll,13,15-heptaene-5-carboxy'Iate) was 72.58 %. m/z calculated for C|7Hi6C1N5O3 [M+H]+: 374; Obtained: 374.1.
Synthesis of Intermediate B (15-chloro-9-(methoxymethyl)-2,4,8,10,ll-pentaazatetracyclofl 1.4.0.0a, 6.0e,n]heptadeca~l(l7),3,5,9,11,13,15-heptaene-5-carboxylic acid).
[0453] Intermediate A (0.4 gm, 1.07 mmol) was dissolved in mixture of
THF/HjO/MeOH (3.2/4.8/8.0 v/v mL). 0.05 gm, 2.14 mmol of LiOH was added and the reaction mixture was stirred at room température for 3 h. The reaction mixture was acidified with aq. 2N HCi solution, ppts were collected and washed with DI water. After drying 0.36 gm of Intermediate B (15-chIoro-9-(methoxymethyI)-2,4,8,10,ll-pentaazatetracyclo[11.4.0.0I,e.0e,]heptadeca-l(17),3,5,9,ll,13,15-heptaene-5-carboxyllc acid) was obtained as a white solid. m/z calculated for CuHijCiNjOj [M+H]+: 346; Obtained: 345.9.
Scheme 29 illustrâtes some selected examples using Intermediate A to generate new analogs.
Scheme 29
264
ooffipoundzx
oofflpound233
Synthesis of Compound 233:
compound 233 [0454] Acetoxime (0.22 gm, 0.31 mmol) was dissolved in anhydrous THF (0.5 mL). ' 0.38 mL, 0.62 mmol of 1.6 M n-BuLi was added dropwise and reaction mixture stirred at
0-5 °C for 1 h in separate flask. A solution of Intermediate A (0.05 gm, 0,13 mmol) in 1.0 mL of THF was added by cannula at 0-5 °C and the rxn was stirred for 16 h by gradually warming at room température. LCMS indicated starting material and intermediate m/z:
374.1 (- 45/14 °/o, two peak merged), m/z 401(-10 %), m/z 402 ( 18 %).
[0455] The reaction mixture was quenched with 0.03 mL of Conc. H2SO4, followed by
0.03 mL of Dl water and refluxed for 2 h. LCMS indicated starting material, product and intermediate m/z: 374.1 (- 43 %), m/z 383 (~ 40 %), m/z 402 (17 %).
265 [0456] The reaction mixture was concentrated under reduced pressure and neutralized with aq. NaHCOj solution, the ppts collected and washed with DI water. After drying gave 22.0 mgof crude ppts. Compound was purified by prep-TLC plate using 1:99 MeOH : CHClj.
Synthesis of Compound 238:
compound 238 ]0457] Step: 1 Intermediate A (0.045 gm, 0.12 mmol) was dissolved in anhydrous toluene (3.0 mL). 0.05 mL, 0.25 mmol ofaminoethanol (35.0 eq) was added and réaction mixture was refluxed for 16 h. The toluene was evaporated and reaction mixture was dissolved în DCM (25.0 mL). The DCM layer was washed with brine followed by Dl water, separated and dried over anhydrous MgSÛ4. The évaporation of organic layer gave
38.3 mgofthe correspondingamide. LCMS indicated product formation m/z: 389 [0458] Step: 2 The above amide (0.038 gm, 0.09 mmol) was dissolved in dry DCM (2.0 mL). 0.026 mL, 0.2 mmol of DAST (2.0 eq) was added to the reaction mixture at 0 °C température and stirred for 1.5 h at 0 °C. 0.065 gm solid K2CO3 (4.8 eq) was added at 0 °C and reaction mixture was stirred for 30 min. The reaction mixture was diluted with aq. NaHCO3 solution and extracted with DCM (15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous MgSO». The évaporation of solvent gave 36 mg of white solid product. m/z calculated for CnHuClNôOifM+H]*: 371; Obtained: 371.
Synthesis of Compound 239:
compound 239
266 [0459] Step: 1 Intermediate A (0.05 gm, 0.13 mmol) was dissolved in anhydrous toluene (3.0 mL). 0.28 gm, 2.67 mmol of aminoethanol (20.0 eq) was added and reaction mixture was refluxed for 16 h. The toluene was evaporated and reaction mixture was dissolved in DCM (25.0 mL). The DCM layer was washed with brine followed by DI water, separated and dried over anhydrous MgSCV The évaporation of organic layer gave the amide. LCMS indicated product formation m/z: 431 [0460] Step: 2 The above amide (0.057 gm, 0.13 mmol) of was dissolved in dry DCM (2.0 mL). 0.035 mL, 0.3 mmol of DAST (2.0 eq) was added to the reaction mixture at 0 O 0
C température and stirred for 1.5 h at 0 C. LCMS indicated product formation m/z 413. O
0,088 gm solid K2CO3 (4.8 eq) was added at 0 C and reaction mixture was stirred for 30 min. The reaction mixture was diluted with aq. NaHCO^ solution and extracted with DCM (15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous MgSO^. Concentration of the organic layer afforded product which was triturated with 20/80 Hex/EtOAc to give a solid which was collected by filtration and dried: 49.4 mg (89%).
Synthesis of Compound 243:
compound 243 [0461] Step: 1 Intermediate A (0.05 gm, 0.13 mmol) was dissolved in anhydrous toluene (3.0 mL). 0.02 mL, 2.67 mmol of the amino alcohol (20.0 eq) was added and reaction mixture was refluxed for 16 h. LCMS indicated starting material left. Xylene was placed (3.0 mL) and 10.0 eq of 3-aminobutan-l-o! added and reaction mixture refluxed for 16 h. Finally total 40.0 eq of amino éthanol was required to convert all starting material into product în refluxing xylene. The rxn mixture cooled to 0 oC and ppts filtered. The filtrate was extracted with DCM (15.0 mL x 4). The DCM layer was washed with brine followed by DI water, separated and dried over anhydrous MgSCL. The
267 évaporation of organic layer gave the corresponding amide. LCMS indicated product formation m/z: 403.
[0462] Step: 2 The above amide (0.054 gm, 0.13 mmol) was dissolved in dry DCM (2.0 mL). 0.05 mL, 0.33 mmol of DAST was added to the réaction mixture at 0 °C température and stirred for 1.5 h at 0 °C. LCMS indicted product formation. 0.09 gm solid K2CO3 was added at 0 °C and reaction mixture was gradually warmed to room température. The reaction mixture was diluted with aq. NaHCO3 solution and extracted with DCM ( 15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous MgSQ». The évaporation of solvent gave crude product. Purification was performed by prep TLC, Mobile Phase: 95:05, DCM:MeOH. m/z calculated for CigHnCINôOi [M+H]+: 385; Obtained: 385.
Synthesis of Compound 244:
compound 244 [0463] Compound 243 from above (0.011 gm, 0.03 mmol) was dissolved in toluene (2.0 mL). 0.010 gm, 0.04 mmol of DIX? was added and reaction mixture was stirred at 50 °C for 1 h. LCMS indicated starting material m/z 385 and little amount of product m/z 383. The rxn mixture was stirred at 60 °C for 3 h. LCMS indicated starting material m/z 385, product m/z 383. The rxn mixture was stirred at 70 °C for 2 h. LCMS indicated starting material m/z 385, product m/z 383 and side product m/z 421. The reaction mixture was stirred at 40 °C for 16 h. LCMS indicated major amount of product m/z 383 and little amount of side product m/z 421 and starting material. The toluene was evaporated and crude product was purified by prep-TLC plate. Mobile phase DCM:MeOH, 95:05 v/v to obtain 4.4 mg of product. m/z calculated for CisHiîCINèOî [M+H]+: 383; Obtained: 383.
Synthesis of Compound 249:
268
compound 249 [0464] Compound 238 from above (0.016 gm, 0.05 mmol) was dissolved in toluene (2.0 mL). 0.015 gm, 0.07 mmol of DIX) was added and reaction mixture was stirred at 50 °C for t h. LCMS indicated starting material m/z 371. The rxn mixture was stirred at 60 °C for 5 h. LCMS indicated starting material m/z 371, product m/z 369 and undesïred m/z 407. The rxn mixture was stirred at 30 °C for 16 h. LCMS indicated starting material m/z 371, product m/z 369 and side product m/z 407. The reaction mixture was stirred at 65 °C for 4 h. LCMS indicated product m/z 369, side product m/z 407 and little amount of starting material. The toluene was evaporated and crude product was purified by prep10 TLC plate. Mobile phase DCMiMeOH, 95:05 v/v to obtain 2.3 mg of product. m/z calculated for CitHijCINîOî [M+H]+; 369; Obtained: 369.
Synthesis of Compound 256:
compound 258 [0465] Step 1: Intermediate A (0.1 gm, 0.27 mmol) was dissolved in anhydrous THF (3.0 mL). 0.67 mL, 0.67 mmol of 1.0 M solution of D1BAL in THF was added dropwise and réaction mixture stirred at 0-5 °C for 2 h. LCMS shows alcohol réduction product formation m/z 332. The reaction was quenched with MeOH (1.0 mL), followed by water (0.5 mL). The saturated solution of NaHCOj was added and ppts were filtered through celite bed. The product was extracted using DCM (25.0 mL x 3). The combined DCM layers was washed with brine, separated and dried over anhydrous Na2SO4. The évaporation of solvent gave 46.1 mg of [15-chloro-9-(methoxymethyl)-2,4,8,10,l 1pentaazatetracyclo[l 1.4.0.0I,6-0e,li]heptadeca-l(l7),3,5,9,l 1,13,15 -heptaen-5yljmethanol as a solid product, Yield 51.9%. m/z calculated for CisHhCINjOj [M+H]+: 332; Obtained: 332.
269 [0466] Step 2: The above alcohol (0.05 gm, 0.14 mmol) of was dissolved in anhydrous DCM (3.0 mL). 0.09 gm of Dess-Martin Periodinane was added and reaction mixture was stirred at room température for 2 h. LCMS shows product formation m/z 330. The reaction was quenched with IN NaOH solution (2-mL). The saturated solution of
NaHCCh was added and the product was extracted using DCM (20.0 mL x 3). The combined DCM layers was washed with brine, separated and dried over anhydrous Na2SO4. The évaporation of solvent gave desired aldéhyde (15-chloro-9(methoxymethy 1)-2,4,8,10,1 l-pentaazatetracyclo[l 1.4.0.02,6.08,l2]heptadeca1(17),3,5,9,1 l,13,15-heptaene-5-carba!dehyde) as a solid product, Yield Quantitative, m/z 10 calculated for C15H12CIN5O2 [M+H]*: 330; Obtained: 330.
[0467] Step 3: 1.6 M n-BuLi solution in hexane (0.68 mL, 1.08 mmol) was added dropwise into 1.4 mL, 0.86 mmol oftrimethylsîlyldiazomethane solution in hexane dissolved in 3.0 mL of THF at -78 °C température. The reaction mixture was stirred at -78 15 °C température for 30.0 min. The aldéhyde obtained in Step 2 (0.142 gm, 0.43 mmol) in solution in 3.0 mL of THF was added dropwise into the reaction mixture at -78 °C température and graduai ly warmed to room température. LCMS shows product formation m/z 326 and starting material m/z 330. The reaction mixture was quenched with saturated NH4CI solution. The product was extracted using DCM ( 15.0 mL x 3). The combined 20 DCM layers was washed with brine, separated and dried over anhydrous Na2SC>4. The purification of crude product was performed by 1SCO Combiflash purification system, Mobile Phase: Ethyl acetate/Hexane. 19.0 mg of Compound 256 was obtained and 71.6 mg of starting material was isoiated. m/z calculated for CiéH^CINjO [M+H]*: 326; Obtained: 326.
·
Compound 285
270 [0468] Compound 256 (0.025 gm, 0.08 mmol) was dissolved in degassed DMF (2.0 mL). 0.03 mL, 0.23 mmol of iodobenzene was added to the reaction mixture followed by 0.06 mL, 0.41 mmol of TEA. The reaction mixture was stirred at room température. 0.04 gm, 0.04 mmol of Pd(PPh3)4 and 0.003 gm, 0.015 mmol of Cul mixture was added to the reaction mixture and stirred for 16 h. LCMS shows product formation m/z 402. The reaction mixture was diluted with DI water. The product was extracted using DCM (10.0 mL x 3). The combined DCM layers was washed with brine, separated and dried over anhydrous Na2SO4. The crude reaction mixture was purified through prep-TLC plate. Mobile Phase: EtOAc/MeOH. m/z calculated for CuHièCINjO [M+H]+: 402; Obtained: 402.
Compound 254 ]0469] Isobutyronitrile (10.0 gm, 144.70 mmol) was dissolved în EtOH: Water (150:50 mL, v/v), followed by addition of 10.0 gm, 144.70 mmol of hydroxylamine hydrochloride and 20.0 gm, 144.70 mmol of K2CO3. The réaction mixture was refluxed at 80 °C for 6 h. The solvent was evaporated under reduced pressure and the resulting solid was treated with 150 mL ofethanoi, sonicated, filtered and washed with 100 mL of ethanoi. The combined filtrate was evaporated under reduced pressure and azeotrope with toluene (25.0 mL x 3) to afford 8.1 gm of N’-hydroxy-2-methylpropimidamide as a colorless liquid slurry (54.8 % yield). The above amide-oxime (1.37 gm, 13.38 mmol) was azeotroped with toluene (10 niL x5) before use and dissolved in 20.0 mL of anhydrous THF. 0.27 gm, 6.69 mmol of NaH was added in three portion to the reaction mixture at 0 °C and stirred at ambient température for 30.0 min. 0.5 gm, 1.34 mmol of Intermediate A was added and reaction mixture was stirred for 45.0 min at ambient température and refluxed at 67 °C for 90.0 min. The solvent was evaporated under reduced pressure and resulting yellow paste treated with 25.0 mL of aq. saturated NaHCO3 solution. The ppts were filtered through funnel and washed with water 10.0 mL and hexane 10.0 mL to afford 0.380 gm solid (69.1 % yield). m/z calculated for CiçHisCINjCh [M+H]+: 412.0; Obtained: 412.1.
271
Synthesis of Compound 215
Compound 215 [0470] The alcohol [ 15-chloro-9-(methoxymethyl)-2,4,8,10,11-pentaazatetracyclo [11.4.0.0^.08.11] heptadeca-ί (17),3,5,9,11,13,15-heptaen-5-yl]methanoi (prepared in Compound 256, Step l) (34 mg, 0.1025 mmol) was suspended in dry THF (2 mL). HMPA (36.7 mg, 0.205 mmol) was added followed by ethyl iodide (0.33 mL) and NaH (41 mg of 60% suspension în oil). The reaction was stirred at RT for 5 min, then heated to 70° C ovemight. The mixture was cooled and partitioned between EtOAc and brine. The organic phase was dried and concentrated to afford an oil whîch was purified by column chromatography (0% to 10% MeOH in DCM) to give 3.7 mg of compound 215 as an oil.
Compound 274 [0471] [15-chloro-9-(methoxymethyl)-2,4,8,10,l 1 pentaazatetracyclo[ 11.4.0.0I,6.08,“]heptadeca1(17),3,5,9,1 l,13,15-heptaen-5-yl]methanol (0.02 gm, 0.06 mmol) was dissolved în anhydrous THF (3.0 mL). 0.003 gm of NaH was added and reaction mixture was stirred at room température for 30.0 min. 0.012 mL, 0.12 mmol of 2-bromopyridine was added dropwise and reaction mixture stirred at room température for 16 h. The reaction mixture was refluxed for additional 2 h. LCMS shows m/z 409. The reaction was concentrated under reduced pressure and diluted with saturated solution of NaHCOj. The product was extracted using DCM (10.0 mL x 4). The combined DCM layers was washed with brine,
272
separated and dried over anhydrous Na2SO4. Purification was performed by prep TLC, Mobile Phase: 95:05, DCM:MeOH. ~ 1.0 mg of product obtained. m/z calculated for CjoHnClNfiOilM+H]*: 409; Obtained: 409.
'3 Scheme 30 illustrâtes some selected exampies using Intermediate B to generate new analogs.
Scheme 30
Synthesis of Compound 234:
compound 234
273 [0472] Intermediate B (0.043 gm, 0.12 mmol), 0.3 mmol of EDC.HC1 and 0.048 gm, 0.31 mmol of HOBt hydrate were dissolved in THF/DCM (1:1, v/v 1.5 mL), followed by addition of 0.09 mL, 0.62 mmol oftrimethylaniline and 0.016 mL, 0.25 mmol of propargylamine. The reaction mixture was stirred at room température for 16 h. The reaction mixture was diluted with aq. Ammonium chloride and extracted with ethylacetate. Combined layers were washed with brine, separated and dried over anhydrous MgSOi. Evaporation of organic layer gave crude product ~ 13.0 mg. The crude product was purified through préparative TLC plate, Mobile Phase: 5:95, MeOH, Ethylacetate. m/z calculated for CisHiîCINôOî [M+H]+: 383; Obtained: 383.1
Synthesis of Compound 240:
a· compound 240 [0473] Step 1 Intermediate B (0.05 gm, 0.15 mmol) was dissolved in dry DCM (2.0 mL). 0.05 mL, 0.36 mmol of trimethylamine (2.5 eq), followed by 0.024 mL, 0.29 mmol of oxalylchloride (2.0 eq) were added and reaction mixture stirred for 60 min at room température. 0.076 mL, 0.7 mmol of amino-alcohol (5.0 eq) was added to reaction mixture at 0 °C and stirred for 2.5 h. The reaction mixture was diluted with aq. solution of NaHCOj and extracted with DCM (15.0 mL x 3). The combined organic layers were washed with brine, separated and dried over anhydrous MgSO.». The évaporation of organic layer gave 54.1 mg the amide. LCMS indicated product formation m/z: 431 [0474] Step2 (2S)-2-amino-3-methylbutyl 15-chloro-9-(methoxymethyi)-2,4,8,10,llpentaazatetracyclo[l 1.4.0.02,6.08,l2]heptadeca-l(17),3,5,9,1 l,13,15-heptaene-5carboxylate (0.027 gm, 0.06 mmol) was dissolved in dry DCM (2.0 mL). 0.016 mL, 0.13 mmol of DAST was added to the reaction mixture at 0 °C température and stirred for 3 h at 0-5 °C. LCMS indicted product formation. 0.04 gm solid K2CO3 was added at 0 °C and reaction mixture was gradually warmed to room température. The reaction mixture was diluted with aq. NaHCOj solution and extracted with DCM (15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous MgSO4. The évaporation of solvent gave crude product Purification was performed by prep TLC,
274
Mobile Phase: 95:05, DCM:MeOH. 23.7 mg of solid product was obtained. Mass, m/z calculated for C20H2tCIN6O2[M+H]+: 413; Obtained: 413.
Synthesis of Compound 246:
compound 248 [0475] Compound 240 was converted to Compound 246 using DDQ, Toluene at 50 C în an analogous manner to Compound 245 to give 5.5 mg (37%) of Compound 246. LCMS indicated product formation m/z: 411.
Synthesis of Compound 242:
compound 242 [0476] Step 1: Intermediate B (0.025 gm, 0.07 mmoi) was dissolved in dry DCM (2.0 mL). 0.03 mL, 0.21 mmol oftrimethylamine(3.0eq), followed by 0.015 mL, 0.18 mmol of oxalylchloride (2.5 eq) were added and reaction mixture stirred for 60 min at room température. 0.05 gm, 0.36 mmol of (R,S)-2-amîno-2-phenylethan-l-ol (5.0 eq) was added to reaction mixture at 0 °C and stirred for 2.5 h at room température. The reaction mixture was diluted with aq. solution ofNaHCOj and extracted with DCM (15.0 mL x 3). The combined organic layers were washed with brine, separated and dried over anhydrous MgSCh. The évaporation of organic layer gave the desired amide. LCMS indicated product formation m/z: 465 (0477] Step 2: The above amide (0.034 gm, 0.07 mmol) was dissolved in dry DCM (2.0 mL). 0.03 mL, 0.22 mmol of DAST was added to the reaction mixture at 0 °C température and stirred at 0 °C for 1.5 h. LCMS indicated product formation. 0.05 gm
275 solid K2CO3 was added at 0 C and reaction mixture was gradually warmed to room température. The reaction mixture was diluted with aq. NaHCO3 solution and extracted with DCM (15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous MgSO4. The évaporation of solvent gave crude product. Purification was performed by prep TLC, Mobile Phase: 95:05, DCM:MeOH. m/z calculated for C23Hi9ClN6O2 [M+H]*: 447; Obtained: 447.
[047S| Compound 242 (0.015 gm, 0.03 mmol) was dissolved in toluene (1.5 mL). 0.009 gm, 0.04 mmol of DDQ was added and reaction mixture was stirred at 50 °C for 1.5 h. LCMS indicated starting material m/z 447 and product m/z 445 in 1:3 ratio. 0.005 gm, 0.022 mmol of DDQ was further added and rxn mixture was stirred at 50 °C for 1.5 h. starting material m/z 447 and product m/z 445 in 1:6 ratio. The reaction mixture was stirred at room température for 16 h. Purification was performed by prep TLC, Mobile Phase: 95:05, DCM:MeOH. The band with m/z: 445 was isolated and 9.3 mg of solid compound was obtained (Yield 62.4 %). m/z calculated for C23Hi7ClN6O2 [M+H]*: 445; Obtained: 445.
Synthesis of Compound 237: .
compound 237 [0479] Step 1: Intermediate B (0.025 gm, 0.07 mmol) was dissolved in dry DCM. 0.009 mL, 0.02 mL, 0.14 mmol of trimethylamine, followed by 0.11 mmol of oxalylchloride
276
were added and reaction mixture stirred for 30 min at room température. 0.028 mL, 0.36 mmol of 3-amino-l-propanol was added to reaction mixture at 0 °C and stirred for 2.5 h and then concentrated. LCMS indicated product formation m/z: 403, lîttle starting material left.
[0480] Step 2: The crude amide from Step I (0.018 gm, 0,045 mmol) was dissolved in dry DCM (2.0 mL). 0.012 mL, 0.09 mmol of DAST was added to the reaction mixture at - 78 °C température and gradually warmed to 0 °C. 0.03 gm solid K2COj was added at 78 °C and reaction mixture was gradually warmed to room température. The reaction mixture was diluted with aq. NaHCCh solution and extracted with DCM (15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous MgSQf.
The évaporation of solvent gave 14.7 mg of compound 237 as a white solid product. m/z calculated for CigHI7CIN6O2[M+H]+: 385; Obtained: 385.1.
Synthesis of Compound 263:
Compound 263 [0481] Step 1: Intermediate B (0.03 gm, 0.09 mmol), 0.034 gm, 0.17 mmol of EDC.HC1 and 0.027 gm, 0.17 mmol of HOBt.xH2O were dissolved in anhydrous DCM (2.5 mL). 0.024 gm, 0.17 mmol of R-(-)-2-Pheny!glycinol was added and reaction mixture was stirred for 6 h at room température. LCMS indicated product formation m/z 464.9. The rxn mixture was diluted with DI water and extracted with DCM (10.0 mL x 3). The combined DCM layers were washed with brine, separated and dried over anhydrous Na2SQt. The évaporation of organic layer gave crude product. A liquid syrup was obtained. m/z calculated for CzjH^CINôOj [M+H]+: 465; Obtained: 464.9.
[0482] Step 2: The above amide (0.04 gm, 0.086 mmol) of was dissolved in dry DCM (2.0 mL). 0.03 mL, 0.21 mmol of DAST was added and reaction mixture was stirred at 0 °C température for 2 h. LCMS indicated product formation m/z 446.9.0.06 gm solid
K2COî was added at 0 °C and reaction mixture was gradually warmed to room
277 température. The reaction mixture was diluted with aq. NaHCOj solution and extracted with DCM (15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous Na2SO4. The évaporation of solvent gave crude product. Purification was performed by prep TLC, Mobile Phase: 95:05, DCM:MeOH. 25.0 mg of solid product was obtained. m/z calculated for C2jHi9ClN6O2 [M+H]+: 447; Obtained: 446.9.
Synthesis of Compound 264:
Compound 264 [0483] Step 1: Intermediate B (0.03 gm, 0.09 mmol), 0.034 gm, 0.17 mmol of EDC.HCI and 0.027 gm, 0.17 mmol of HOBt.xH2O were dissolved in anhydrous DCM (2.5 mL). 0.024 gm, 0.17 mmol of S-(+)-2-Phenylglycinol was added and reaction mixture was stirred for 6 h at room température. LCMS indicated product formation m/z 464.9. The rxn mixture was diluted with DI water and extracted with DCM (10.0 mL x 3). The combined DCM layers were washed with brine, separated and dried over anhydrous Na2SO4. The évaporation of organic layer gave crude product. A liquid syrup was obtained. m/z calculated for CuIhiClNfjOj [M+H]+: 465; Obtained: 464.9.
[0484] Step: 2: The above amide (0.04 gm, 0.086 mmol) was dissolved in dry DCM (2.0 mL). 0.03 mL, 0.21 mmol of DAST was added and reaction mixture was stirred at 0 °C température for 2 h. LCMS indicated product formation m/z 446.9.0.06 gm solid K2COj was added at 0 °C and reaction mixture was gradually warmed to room température. The reaction mixture was diluted with aq. NaHCOj solution and extracted with DCM ( 15.0 mL x 3). The organic layer was washed with brine, separated and dried over anhydrous Na2SÛ4. The évaporation of solvent gave crude product. Purification was performed by prep TLC, Mobile Phase: 95:05, DCM:MeOH. 26.4 mg of solid product was obtained. m/z calculated for C2jH]9C1NôO2 [M+H]+: 447; Obtained: 446.9.
[0485] Compounds 180,181, and 182 were prepared using a synthetic procedure that is similar to the one used for the synthesis of Compound 168 as depicted in Scheme 27.
[0486] Compounds 183 - 193 were prepared using a synthetic procedure that is similar to the one used for the synthèses of Compounds 169 - 179 as depicted in Scheme 26.
[0487] Compounds 194 and 195 were prepared using a synthetic procedure that is similar to the one depicted în Schemes 21 and 22.
[0488] Compounds 196-198, and 206 were prepared using a synthetic procedure that is similar to the one depicted in Scheme 18a.
[0489] Compound 202 was prepared using a synthetic procedure that is similar to the 10 one used for the synthesis of Compound 129 as depicted in Scheme 18a.
[0490] Compounds 199, 200, 204, and 205 were prepared using a synthetic procedure that îs similar to the one depicted in Scheme 18b.
[0491] Compounds 201 and 203 were prepared using a synthetic procedure that is similar to the one depicted in Scheme 24.
[0492] Compounds 207 - 210 were prepared using a synthetic procedure that is similar to the one depicted in Scheme 17.
[0493] The nïtrïle substîtuents in Compounds 207 - 210 were generated analogously to those transformations shown in Scheme 22.
[0494] Compounds 211 - 214 were prepared using a synthetic procedure that is similar 20 to the one depicted in Scheme 20.
[0495] Compound 255 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 254.
[0496] Compound 259 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 243.
[0497] · Compound 260 was prepared from the appropriate starting materials using the synthetic routes described în Schemes 28 and 30; similar to compound 242.
[0498] Compound 261 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 30; similar to compound 256.
[0499] Compound 265 was prepared from the appropriate starting materials using the 30 synthetic routes described in Schemes 28 and 30; similar to compound 264.
[0500] Compound 266 was prepared from the appropriate starting materials using the synthetic routes described în Schemes 28 and 30; similar to compound 264.
279 [0501] Compound 267 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 30; similar to compound 264.
[05021 Compound 268 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 30; similar to compound 263.
[0503J Compound 270 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 30; similar to compound 264.
[0504] Compound 271 was prepared from the appropriate starting materials using the synthetic routes described În Schemes 28 and 30; similar to compound 264.
]0505) Compound 275 was prepared from the appropriate starting materials using the 10 synthetic routes described in Schemes 28 and 30; similar to compound 264.
|0506[ Compound 276 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 30; similar to compound 245.
[0507] Compound 278 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 233.
[0508] Compound 281 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 233.
[0509] Compounds 282, 283, 286, 287 were prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 243.
[0510] Compound 288 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 256.
1051II Compound 293 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 285.
[0512] Compounds 294, 295, and 296 were prepared from the appropriate starting 25 · materials using the synthetic routes described in Schemes 28 and 29; similar to compounds 243 and 244.
[0513] Compound 303 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 233.
[0514] Compound 304 was prepared from the appropriate starting materials using the 30 synthetic routes described in Schemes 28 and 29; similar to compound 264.
[0515] Compound 297 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 243.
280 [0516] Compound 307 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 285.
[0517] Compound 308 was prepared from the appropriate starting materials using the synthetic routes described in Scheme 28; similar to Intermediate A.
[0518] Compound 309 was prepared from the appropriate starting materials using the synthetic routes described în Schemes 28 and 29; similar to compound 238.
[0519] Compound 310 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 285.
[0520] Compound 311 was prepared from the appropriate starting materials using the 10 synthetic routes described în Schemes 28 and 29; similar to compound 285.
[0521] Compound 312 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 244.
[0522] Compound 313 was prepared from the appropriate starting materials using the synthetic routes described in Schemes 28 and 29; similar to compound 244.
Synthesis of Compounds 305 and 306
Scheme 31
Compound 288
Compound 308
Compound 308 [0523] Compound 288 (0.015 gm, 0.042 mmol) was dissolved in anhydrous THF (3.0 mL). 0.003 mL, 0.05 mmol of methyl iodide was added at -78 °C température, followed by 0.05 mL, 0.05 mmol of 1.0 M LDA solution. The reaction mixture was stirred at -78 °C and gradually warmed at room température. LCMS shows product formation m/z 368 major, unreacted starting material m/z 354 and dimethylated unknown product m/z 382.1.
The reaction mixture was quenched with saturated NH4CI solution and extracted with 25 EtOAC. Organic layer was dried and concentrated. The purification of crude reaction mixture was performed by prep-TLC plate. Mobile Phase: EtOAc:Hexane 75:25 v/v mL to isolate three bands. It was found through MS that l“ band confirmed m/z 354 of starting material, 2nd band confirmed m/z 368 of mono methyl substituted product Compound 305 and 31 band confirmed m/z 382.1 of dimethyl substituted product 30 Compound 306. ’H NMR (CDCI3) data confirmed the mono methyl substitution on
280
Imidazole ring. Note: ’H NMR data confirmed products formation and pure products isolation.
[0524] Compound 216 was prepared similarly as compound 129 in Scheme 18a. MS: [M+1] = 395.
[0525[ Compound 217 was prepared similarly as compound 129 in Scheme 18a. MS:
[M+1] = 381.
Scheme 32
NRff » NHM« compoird 247 NRff » N(CHj)4 compoird 248
Î1 UOH (CCO)i DMF. (min*
ccmparid 298
R Cl canpanl 219
Synthesis of Compound 218:
Compound 218 . [0526] To 5-(ethoxycarbonyl)-16-methoxy-2,3,4,10,12-pentaazatetracyclo [11.4.0.02-6.0sl2]heptadeca’l(17),3,5,8,10,13,15-heptaene-9-carboxylic acid from Scheme 27 (0.609g, 1.65mmol) stirring in DMF (10ml) at 0°C was added NaHCO3 (0.749g,
8.9mmol) and NBS (0.793g, 4.45mmol). The reaction was allowed to proceed to ambient temperature ovemight. The réaction was then diluted with EtOAc, cooled to 0°C, and sat. sodium thiosulfate was added carefully under stirring. After foaming stopped, organic layer was separated, washed with sat. NaHCO3, brine, and dried over MgSQj. Filtration and solvent removal gave the crude bromide which was purified by flash column chromatography using a gradient elution of 0 to 80% EtOAc in hexanes. 424.2 mg (64%) was obtained as a yellowish solid. MS: [M+1] =405.
[0527[ To the bromide (286.7mg, 0.709mmol) from above in a thick walled rbf was added Cul (121.5mg, 0.638mmol), trimethylsilyl acetylene (1.04g, 10.7mmol), triethyl amine (0.717g, 7.09mmol), dicyclohexyl(2’,6’-dimethoxybiphenyl-2-yl) phosphine (0.349g, 0.85Immol) and 1,4-dioxane (2.5ml; degassed). The reaction vessel was flushed with nitrogen gas, and bis(triphenylphosphine) palladium(II) dichloride (298.2mg, 0.425mmol) was added. The reaction mixture was stirred at rt for 30 min then heated at 100°C under sealed tube conditions for 16 hrs, diluted with EtOAc, and washed with sat. NaHCO3, brine, and dried (MgSO.»). Silica gel column chromatography ofthe filtered and concentrated reaction mixture using a gradient of 0 to 100% EtOAc in hexanes gave I57.9mg (53%) ofthe desired trimethylsilyl acetylene product as a brownish solid. MS: [M+1] =422.
[0528] The trimethylsilyl alkyne obtained above (128.7mg, 0.305mmol) was treated with lithium hydroxide (36.6mg, 1.53mmol) in a solvent mixture of THF (0.9ml), water (0.75ml) and MeOH (0.15ml) at rt for two hrs. The mixture was then acidified to pH 3-4 with dil. Hydrochloric acid, and extracted with EtOAc (3x). The remaining precipitate in the aq. Layer was found to be product and was collected by filtration, and was combined φ 283 with the product isolated from the organic layer to give 95.6mg ofthe acid as a yellowish solid.
[0529] To the acid (95.6mg, 0.298mmol) in THF (1.3ml) and dichloromethane (1.3ml) was added N,O-dimethylhydroxy lamine hydrochloride (232.4mg, 2.38mmol), EDC $ hydrochloride (456.7mg, 2.38mmol), HOBt hydrate (91.2mg), and triethyl amine (0.833ml, 5.93mmol). After 16 hrs stirring, the reaction was diluted with EtOAc, and washed with sat. NH4CI. Aq. Layer was separated and extracted with EtOAc (3x), combined organic layer was washed with sat. NaHCO3, brine, and dried (MgSOi). Filtration followed by solvent removal gave IO4.8mg of the amide as a yellowish solid.
[0530] To the Weînreb amide from above (20. Img, 0.0552mmol) stirring in anh. THF (0.8ml) cooled in an ice-salt bath was added 4-fluorophenyl magnésium bromide solution (IM THF; 0.828ml) slowly. The reaction mixture was stirred to ambient température over 4 hrs, then quenched with sat. NH4CI, extracted with EtOAc (3x), washed with sat. NaHCO3, brine, and dried (MgSO4). Prep. TLC of the filtered concentrated mixture using
5% MeOH in DCM gave 2.0mg of Compound 218 as an off-white solid. MS: [M+1 ] =400.
[0531] Compound 219 was prepared similarly as compound 218 as depicted in Scheme 32. MS: [M+1] = 416.
[0532] 5 -benzoy l-9-ethyny I-16-methoxy-2,3,4,10,12- pentaazatetracy cio [1 l,4,0.026.08,l2]heptadeca-l(17),3,5,8,10,13,15-heptaene (90.3mg, 0.237mmol; obtained similarly as 218, was stirred in THF (1.5ml) at rt. NaBHj (26.8mg, 0.71mmol) was added.
After lhr, the reaction was quenched with NH4CI for 5 min, and extracted with EtOAc.
Organic layer was separated and washed with brine and dried over MgSÜ4. Filtration and
284 solvent removal in vacuo gave a clear viscous oil, which was treated with triethylsilane (241.9mg, 2.08mmol) and trifluoroacetic acid (0.32ml) in DCM (1,5ml) for 3hrs. The reaction mixture was placed on Rotovap for solvent removal, diluted with EtOAc, and washed with sat. NaHCOj. Aq. Layer was separated and extracted with EtOAc, the combined organic layer was washed with brine, and dried over MgSO^. Prep. TLC of the filtered concentrate using 2% MeOH in DCM/EtOAc (1:1) gave 2.5mg of Compound 220 as a clear filmy solid. MS: [M+1] =370.
Compound 221 was prepared similarly as compound 220 as depicted in Scheme 32. MS: [M+1] = 384.
Scheme 33
R 4-F. compound 222 R 2-F: compound 223
R H compoml 224 R · 3-Br compound 225
NRR'· NfCHi).· compound 250
NRR'· N(CHjV compound 231
NRR'· NEIj:compound 252
NRR' 4-phenyl plppentn· compoind 253
BrPhjT
Compound 222
Synthesis of Compound 222:
Compound 222
285 [0533] The cyano ester (407. lmg, 1.16mmol) was treated with lithium hydroxide (83.5mg, 3.49mmol) în a solvent mixture ofTHF (6ml), water (5ml) and MeOH (lml) at rt for 16 hrs, then concentrated in vacuo, acidified to pH 3-4 with dil. HCl, and cooled at 0°C. Precipitate was collected by filtration, washed with small amount ofwater, and dried to give 271.9mg (73%) acid as a greyish solid. This acid (271.9mg) was suspended and stirred in THF (2ml) at 0°C, to which was added borane dimethylsulfîde solution (2M THF; 8.4ml) dropwise. The reaction was allowed to proceed to ambient température ovemight, cooled in an ice bath, quenched with MeOH (10ml) for two hrs, and concentrated in vacuo. The resulting solid residue was partitioned between DCM and sat.
NaHCO3 and stirred for 20 min. Aq. Layer was separated and extracted with DCM (3x).
Combined organic layer was washed with brine and dried over MgSO4. Filtration and solvent removal gave 137.8mg ofthe crude alcohol product as a yellowîsh waxy solid. The alcohol from above (137.8mg) was treated with phosphores oxybromide (256.3mg, 0.894mmol) in 1,4-dioxane (5ml) at 100°C for 3hrs. Upon cooling in an ice bath, the reaction mixture was treated with sat. NaHCO3 (15ml) and EtOAc (15ml) under stirring conditions for about 20 min. The basic aq. Layer was separated and extracted with EtOAc (2x). Combined organic layer was washed with brine and dried over MgSO4. Filtration and solvent removal in vacuo gave the crude primary bromide as a solid paste which was stored in cold and used without further purification when needed.
The crude bromide from above (27.0mg, 0.0727mmol) was treated with 4-fluorophenol (652mg, 0.585mmol) and césium carbonate (47.4mg, O.I45mmol) at rt for 16 hrs. The reaction mixture was diluted with EtOAc, washed with brine, and dried over MgSO4. Prep. TLC ofthe filtered concentrate using 5% MeOH in DCM/EtOAc ( 1:1 ) gave I2mg of Compound 222 as a yellowîsh solid. MS: [M+1] =403.
[0534] Compound 223 was prepared similarly as compound 222 as depicted in Scheme 33. MS: [M+1] = 403.
[0535] Compound 224 was prepared similarly as compound 222 as depicted in
Scheme 33. MS: [M+1] = 385.
[0536] Compound 225 was prepared similarly as compound 222 as depicted în Scheme 33. MS: [M+1] = 464.
286
Scheme 34
R Bn compound 299 R Me' compound 300
1.2.4-tiazole, POCIa
Prepared tlmliarfyas in Scheme 11
SnCI
N^N
R· H. compound 220 R 3-F: compound 227 R 4-F. compound 228
[0537] Ethyl l-(5-chloro-2-nitrophenyl)-5-(2-ethoxy-2-oxoethyl)-lH-l,2,3-triazole-45 carboxylate (21.2g; obtained similarly as 14 in Scheme 11) was treated with tin (II) chloride hydrate (60g) in a mixture solvent of EtOAc ! EtOH (1:2,300ml) at 70°C for 3hrs. HCl (40ml; 37%) was added and heating continued for 3 days. More tin (II) chloride hydrate (25g) and HCI (15ml) added and heating continued for 2 days. The reaction was cooled, concentrated under reduced pressure to a brownish oïl, diluted with EtOAc (250ml), and carefully basified to pH 8-9 with sodium carbonate solution. The aq. Layer was separated and extracted with EtOAc repeatedly. Combined organic layer was washed with brine and dried over MgSO4. Filtration and solvent removal followed by recrystallization in MeOH gave 3.3g (51%) of the cyclized mono-ester as a yellowtsh
287 solid. MS: [M+1] = 307.
Préparation of tert-butyl isocyanoacetate:
[0538| To a suspension of fcrf-butyl glycinate hydrochlorîde (10.0 g, 60 mmol) în DCM (200m!) was added EDC.HC1 (14.9 g, 78 mmol) and triethylamine (12.5 mL, 89.8 mmol). The reaction mixture was cooled down to -50°C, formic acid (3.4 mL, 89.8 mmol) in DCM (10 mL) was added slowly. The reaction mixture was stirred at -50°C for one hour then at 4°C for 3 h. Water (150m!) was added. After 30 min stirring, aq. Layer was separated and extracted with DCM (3x). Combined organic layer was washed with brine and dried over MgSQj. Filtration and solvent remova! under reduced pressure gave 10g (100%) of the formy! amide as a clear viscous oil. H*NMR (CDCIj) δ 8.23 (IH, s), 6.17 (1H, br s), 3.98 (2H, d, J=5.5Hz), and 1.48 (9H, s).
[0539| To a solution of formy! amide (10.5 g, 66 mmol) in DCM (180 mL) was added triethylamine (36.8 mL, 264 mmol). The solution was cooled în a salt-ice bath, and POCh (7.4 mL, 79.2 mmol) was added slowly. The reaction was stirred in the cold bath for one hr. Then sodium carbonate (7.7g, 72.6mmol) in water (90ml) was added to the cold reaction mixture. After 15 min, cold bath was removed and stirring continued atambient température for one hr. Aq. Layer was separated and extracted with DCM (3x). Combined organic layer was washed with brine and dried over MgSO4. Filtration and solvent removal under reduced pressure gave 7.9g (84%) rert-butyl isocyanoacetate as a dark brown liquid. H'NMR (CDCh) δ 4.12 (2H, s), and 1.51 (9H, s).
[0540[ A solution of terf-butyl isocyanoacetate (1.51g, 10.7mmol) in DMF (43ml) was cooled to -50°C under nitrogen atmosphère. Potassium z-butoxide (1.05g, 9.4mmol; finely pressed) was added. After one hr stirring at -50°C, the 1,2,4-triazole intermediate (2.32g, 6.48mmol; prepared similarly as compound 20 in Scheme 11) was added to the resulting reddîsh clear solution, and the reaction was stirred to ambient température ovemight. Sat. NaHCCh (15ml) was added, and the reaction mixture was extracted with diethyl ether (5x), washed with brine, and dried (MgSCh). Silica gel chromatography ofthe filtered concentrate using a gradient of 0 to 100% EtOAc in hexanes gave 2.5g (89%) of the
288 imidazole /-butyl ester product as a yellowish solid. MS: [M+1-/Bu] = 374.
[0541 ] The imidazole /-butyl ester from above ( 1.1 g, 2.56mmol) was treated with trifluoroacetic acid (13ml) in DCM (!3ml) for 3hr or until ail starting /-butyl ester was hydrolyzed. The reaction was then concentrated under reduced pressure. Residual TFA 5 was removed with repeated addition and évaporation of toluene. The acid product was obtained as a dark brown viscous oily material, and was used without further purification. MS: [M+1] = 374.
[0542] Ethyl 16-chloro-9-cyano-2,3,4,10,1210 pentaazatetracyclofl !.4.0.026.08'12]heptadeca-l(17),3,5,8,10,13,!5-heptaene-5carboxylate (477mg, l.34mmol); obtained similarly as ethyl 9-cyano-16-methoxy2,3,4,10,12-pentaazatetracyclo[ 11.4.0.02,6.08·12] heptadeca-1 ( 17),3,5,8, ! 0,13,15-heptaene5-carboxylate in Scheme 27) was treated with lithium hydroxide (80.5mg, 3.36mmol) in a solvent mixture of THF (6ml), water (5ml) and MeOH (1ml) at rt for 16 hrs. The reaction was concentrated under reduced pressure, acidified to pH 3-4 with dil. HCl, and cooled to 0°C. Precipitate was collected by filtration, washed with small amount of water, and further dried to give 396.2 mg crude triazolo carboxylic acid product, MS: [M+1] = 327.
[0543] To a suspension of the crude acid from above (396.2mg) in anhydrous THF (7ml) at 0°C was added borane dimethylsulfide complex (10.9ml; 2M THF) dropwise.
The reaction was allowed to proceed to ambient température ovemight, and was cooled to 0°C, then slowly quenched with MeOH. After 30 min stirring, the reaction mixture was concentrated in vacuo. The resulting slurry was treated with MeOH which was subsequently removed in vacuo. This process was repeated several times. The resulting residue was then treated with 5% MeOH in DCM, and washed with sat. NaHCOj. Aq.
Layer was extracted with DCM (3x), combined organic layer was washed with brine and drîed over MgSO.4. Filtration and solvent removal gave a mixture of the crude alcohol product ([M+1] = 313) and the corresponding primary amide due to hydrolysis of the cayno group ([M+1] = 331). 388.8mg ofthis crude mixture was obtained and was used
289 without further purification.
[0544] The alcohol mixture (388.8mg) from above was treated with phosphores oxybromide (2.02g) in 1,4-dioxane ( 10ml) at 100°C for 8hrs. The reaction was cooled to 0°C, and carefully quenched with sat. NaHCOj (15ml). After 20 min stirring, the reaction 5 mixture was extracted with EtOAc (3x), washed with brine, and dried over MgSO4.
Filtration and solvent removal under reduced pressure gave the crude bromide as a viscous paste, which was used for the next step without further purification.
compound 226 [0545] Compound 226 was prepared similarly as Compound 222 in Scheme 33 using 10 the bromide prepared from above. MS: [M+1 ] = 389.
[0546] Compound 227 was prepared in a similar fashion as Compound 226, depicted in Scheme 34. MS: [M+1] = 407.
[0547] Compound 228 was prepared in a similar fashion as Compound 226, depicted in Scheme 34. MS: [M+1] = 407.
Synthesis of Compound 229:
290 [0548] The benzyl analog 229, shown in Scheme 35, was prepared similarly as the benzyl compound 220 in Scheme 32. MS: [M+1] = 411.
Synthesis of Compound 230:
[0549] The ketone analog 230, shown în Scheme 35, was prepared similarly as ketone 218 in Scheme 32. MS: [M+1 ] = 474.
Synthesis of Compound 231:
[0550] The benzyl analog 231, shown in Scheme 35, was prepared similarly as the benzyl compound 220 in Scheme 32. MS: [M+1] = 460.
Scheme 36
(ichemo 16a) DMB dlmethaxy benzyl
Ar · Ph’ compound 232 Ar 5-pyridyl' compound 23S
Ar Ph’ compound 236
Ar 3-pyrkfyl compound 241
(see Scheme 36)
2. NBS. NaHCOi
----------->1. LIOH
[0551] Compound 63 (0.805g, 1.78mmol; from Scheme 18a) was treated with lithium hydroxide (0.128g, 5.34mmol) în a solvent mixture of THF (6ml), water (5ml) and MeOH ( 1 ml) at rt for 16 hrs. The réaction was then concentrated in vacuo, acidîfied to pH 3-4 with dil. HCL Resulting precipitate was collected by filtration, washed with water and dried to give 0.638g acid as a yellow solid. MS: [M+1 ] = 424.
The acid from above (0.638g, L5mmol) was treated with NBS (1.61g. 9mmol) and NaHCO3 (1.51g, 18mmol) at rt for 16hrs. The reaction mixture was cooled to 0°C, sat.
291 sodium thiosulfate (aq.) was carefully and slowly added. This was extracted with EtOAc (2x), washed with sat. NaHCOj, brine, and dried over MgSO*. Silica gel chromatography of the filtered concentrate with a gradient of 0 to 100% EtOAc in hexanes gave 0.580g (72%) of the di-bromo product as a yellowish solid. MS: [M+1] = 538.
[0552] Compound 232 was prepared similarly as Compound 55 in Scheme 18a, using the bromide prepared above. MS: [M+1] = 439.
[0553] Compound 235 was prepared similarly as Compound 55 in Scheme 18a, using the bromide prepared above. MS: [M+1] = 440.
[0554] Compound 236 The alkyne moiety was prepared similarly as Compound 161 in Scheme 21. MS: [M+1] = 384.
[0555] Compound 241 The alkyne moiety was prepared similarly as Compound 161 in
Scheme 21. MS: [M+1] = 385.
(see Scheme 32)
-
1. UOH
2. (CICO)2, DMF; amine
compound 247
Synthesis of Compound 247:
[0556] The bromide ester (13.9mg, 0.0344mmol) was treated with lithium hydroxide (lOmg) in a solvent mixture of THF (0.3ml), water (0.25ml) and MeOH (0.05ml) at rt for 16 hrs. The reaction was then concentrated in vacuo, acidified to pH 3-4 with dil. HCl and cooled to 0°C. Resulting precipitate was collected by filtration, washed with water and dried to give 9.5mg (74%) acid as a light brown solid. MS: [M+1] = 377.
To the acid from above (5.1mg, 0.0136mmol) stirring in DCM (0.15ml) was added oxalyl chloride (8.6mg, 0.0678mmol), and DMF (5ul). After 2hrs stirring, solvent and excess reagent was removed in vacuo. Resulting residue was re-suspended in DCM (0.15ml), cooled ïn an ice-salt bath, and ethanolic methyl amine (lOOul; 33%) wasadded dropwise. After 20 min stirring, the reaction mixture was applied to a prep. TLC plate and product was isolated using 5% MeOH în DCM as eluent. 4.3mg (81%) Compound 247 was
292 obtained as a white solid. MS: [M+1] = 390.
[0557] Compound 248 was prepared similarly as Compound 247, as depicted in
Scheme 32. MS: [M+1] = 430.
[0558] To the acid (108.0mg, 0.335mmol) suspended in DCM (2ml) at 0°C was added oxalyl chloride (170.1 mg, 1.34mmol) slowly, followed by DMF (20ul). After bubbling stopped, ice bath was removed and the reaction was allowed to proceed at rt for 2 hrs. Solvent and excess reagent was removed in vacuo. Resulting light brown solid was cooled to 0°C. NaBH» solution (2.2ml; 1.5M în methoxyethoxy ethane) was added. After 30 min, the reaction was quenched with IN HCl (0.2ml), and stirring continued until bubbling stopped. EtOAc (10ml) and sat. NaHCOj (10ml) was added and this was stirred ovemight. Aq. Layer was separated and extracted with EtOAc (3x); combined organic layer was washed with brine and dried over MgSCh. Filtration and solvent removal gave 97.0mg (94%) of the alcohol as a yellowish solid. MS: [M+1 ] = 309.
[0559] The alcohol from above (97.0mg, 0.315mmol) was treated with Dess-Martin Periodinane (266.9mg, 0.629mmol) in DCM (2ml) for lhr. The reaction mixture was diluted with DCM, washed with sat. NaHCOj. Aq. Layer was separated and extracted with DCM (3x), combined organic layer washed with brine, and dried over MgSO®. Filtration and solvent removal under reduced pressure gave quantitative yield ofthe crude aldéhyde as a brownish solid, which was used without further purification.
[0560] Compound 250 was prepared similarly as compound 48 în Scheme 16 using the aldéhyde from above, as depicted in Scheme 33. MS: [M+1] = 362 [0561] Compound 251 was prepared similarly as compound 250, as depicted in Scheme 33. MS: [M+1] = 376.
[0562] Compound 252 was prepared similarly as compound 250, as depicted in Scheme 33. MS: [M+1] = 364.
[0563] Compound 253 was prepared similarly as compound 250, as depicted in Scheme 33. MS: [M+1] = 452.
293
Scheme 37
R 2-propyl. compound 272
R Ma compound 273 compound 279
[0564] The acid (16 in Scheme 15, X = OMe; 258.1mg, 0.941mmol) was treated with acetic acid (2ml) at 120°C for 5hr. Solvent was then removed in vacuo. Solid residue was treated in water (7ml) with sonication, filtered, washed with water, and dried to give 158.4mg (73%) decarboxylated product as a brownish solid. MS: [M+1] = 231.
compound 257 [0565] Compound 257 was prepared în a similarly fashion as compound 167 in
Scheme 11. MS: [M+1] = 364.
294 [0566] Compound 258 was prepared in a similarly fashion as compound 167 in Scheme 11. MS: [M+1] = 336.
Synthesis of Compound 262:
compound 262 [0567] Benzyl triphenyl phosphonium bromide (29.0mg, 0.0669mmol) was stirred in THF (0.5ml) cooled in a sait-ice bath. Sodium hydride (4.12mg, 0.103mmol; 60% oil suspension) was added. After 20 min stirring, aldéhyde (15.8mg. 0.0515mmol) was added. The reaction was allowed to slowly warm to rt over four hrs, then quenched with sat. NH4CI, extracted with EtOAc (3x), washed with brine, and dried over MgSOj. Compound 262 was isoiated by repeated prep. TLCs using 2% MeOH in DCM. 1.1 mg was isoiated as a white solid. MS: [M+1] = 381.
[0568] The starting ester (76.4mg, 0.235mmol) was treated with N-bromosuccinamide (83.6mg, 0.470mmol) in acetonitrile (2.3ml) at rt for three days. To the reaction mixture was added sat. sodium thiosulfate. After 15 min stirring, aq. Layer was separated and extracted with EtOAc (2x). Combined organic layer was washed with brine and dried over MgSO4. The bromide product was isoiated by prep. TLC using hexanes:EtOAc = 1:3 as the eluting solvent. 50.2mg (52%) was obtained as a light brown foamy solid. MS: [M+1] = 405.
[0569] To the bromide from above (24.1mg, 0.0596mmol) under nitrogen atm. was added phenyl boronic acid (10.3mg, 0.083mmol), tetrakis(triphenylphosphine)pa!ladium(0) (6.9mg, 0.006mmol), dimethoxyethane (0.69mL; degassed), and aq. Na2CO3 solution (77ul; 2M). The réaction was heated at 100°C for 5hrs, cooled to rt, diluted with EtOAc, washed with sat. NaHCO3, brine, and
295 dried over MgSO.). Prep. TLC with hexanes:EtOAc = 1:3 gave 17.2mg (72%) Suzuki coupling product as a yellowish amorphous material. MS: [M+1 ] = 402.
Synthèses of Compound 272,273 and 277:
[0570] Compound 272 was prepared similarly as compound 167 in Scheme 11, starting from the imidazole ester above. MS: [M+1 ] = 440.
[0571] Compound 273 was prepared similarly as compound 167 En Scheme 11, starting from the imidazole ester above. MS: [M+1] = 412.
[0572] Compound 277 was prepared similarly as compound 167 En Scheme 11. MS: 10 [M+l] = 378.
[0573] Compound 279 was prepared via Suzuki coupling in a similar fashion as detailed above (see Scheme 37). MS: [M+1] = 436.
compound 267
Synthesis of Compound 280:
[0574] To compound 267 (11.7mg, 0.0274mmoi) under nitrogen atmosphère was added dicyclohexy![2-(2,4,6-triisopropylphenyl) phenyljphosphane (7.8mg, 0.0l64mmol), césium carbonate (22.3mg, 0.0685mmo!), and acetonitrile (0.30ml). The reaction flask was flushed with nitrogen gas, and dichlorobis(acetonitri!e)pal!adium (11) (1.42mg,
0.0055mol) was added. After stirring at rt for 30 min, trimethylsilyl acetylene (80.7mg,
0.822mmo!) was added, and the reaction was heated at 90°C for 5 hrs, cooled to rt, diluted with EtOAc, and washed with sat. NaHCOj. Aq. Layer was separated and extracted with
EtOAc (2x), combined organic layer was washed with brine and dried over MgSOi. Prep.
TLC ofthe filtered concentrate using 5% MeOH in DCM/EtOAc (1:1) gave 4.1 mg yellowish solid. MS: [M+1] = 417.
trimethylsilyl acetylene dérivative as a yellowish solid. MS: [M+1] =489.
[0575] The trimethylsilyl acetylene (4.1mg, 0.0084mmo!) from above was treated with potassium carbonate (1.2mg, 0.0084mmol) in methanol (0.2ml) at rt for 3hrs. Prep. TLC using 7% MeOH in DCM/EtOAc (1:1) as eluting solvent gave 1.6mg Compound 280 as a
29S
Synthèses of Compound 284,301 and 302:
[0576] Compound 284 was prepared similarly as compound 280, starting from compound 240. MS: [M+1] ® 403.
[0577] Compound 301 was prepared similarly as compound 280 starting from 5 compound 264. MS: [M+1] = 437.
[0578] Compound 302 was prepared similarly as compound 280 starting from compound 245. MS: [M+1] = 435.
(sm Schemes 29 and 30) >
R (S)-P h: compound 289 R (R)-Ph: compound 290 R “ (R>Me: compound 291 R (S>Me: compound 292
Synthèses of Compound 289,290,291 and 292:
[0579] Compound 289 was prepared similarly as compound 263 as depicted in
Scheme 30. MS: [M+1 ] = 399.
[0580] Compound 290 was prepared similarly as compound 263 as depicted în
Scheme 30. MS: [M+1] = 399.
[0581] Compound 291 was prepared similarly as compound 243 as depicted în
Scheme 29. MS: [M+1] = 337.
[0582] Compound 292 was prepared similarly as compound 243 as depicted in
Scheme 29. MS: [M+1] = 337.
(see Scheme 32)
1. UBH4
2. POBr3
3. MeOH, NaH
compound 298
Synthesis of Compound 298:
[0583] The ester (107.9mg, 0.264mmol) în THF (2.4ml) was treated with lithium borohydrîde solution (0.264ml; 2M THF) at 0°C. The reaction was allowed to warm to ambient température over 4hrs, then quenched with sat. NaHCO3 slowly, extracted with EtOAc (4x), washed with brine, and dried over MgSOj. Filtration and solvent removal
297 gave 77.3mg (86%) alcohol as a yellowîsh solid.
[0584] Alcohol from above (16.4mg, 0.0448mmol) was treated with phosphores oxybromide (25.7mg, 0.0895mmol) in 1,4-dioxane (0.5ml) at 95°C for 3hrs. The reaction was then cooled to 0°C, quenched with sat. NaHCOj (5ml) for 20 min, and extracted with EtOAc (3x), washed with brine, and dried over MgSO4. Filtration and drying gave 16.6 mg yellowîsh solid which was dissolved in anhydrous MeOH (18ul) and THF (0.35ml). This was cooled to 0°C, and NaH (9.2mg; 60% suspension) was added. After 2hrs stirring at 0°C, the reaction was quenched with sat. NaHCOj, extracted with EtOAc (3x), washed with brine, and dried over MgSO4. Prep. TLC using 10% MeOH în DCM gave 0.8mg
Compound 298 as a yellowîsh solid. MS: [M+1] = 381.
I.HBr, HOAc (Scheme 21) ---------->► 2. NaOMe, MeOH
[0585] The starting alcohol (616mg) was converted to the corresponding bromide as described earlier (see Scheme 21). The resulting crude bromide was dissolved in anhydrous methanol (23ml), and cooled to 0°C. NaH (932mg; 60% suspension) was added portîonwise. After bubbling stopped, the reaction mixture was heated to reflux for 30 min, then cooled to rt, and treated with 2N HCl (11ml). Resulting precipitate was collected by filtration, and the desired methyl ether was isolated by silica gel chromatography, using a gradient elution of 0 to 10% MeOH in DCM. 217 mg was collected as a yellowîsh solid. MS: [M+1] = 279.
R = Bn: compound 299
R = Me: 300
Synthèses of Compounds 299 and 300 [0586[ Compound 299 was prepared similarly as Compound 289, using the methyl ether intermediate above. MS: [M+1] = 46L [0587] Compound 300 was prepared similarly as Compound 289, using the methyl
298 ether intermediate above. MS: [M+1] = 385.
[0588] Compounds 180-313 were characterized by MS and *H NMR. The MS characterization is summarized below în Table 5.
Table 5. MS characterization of Compounds 180-313,
Cmp No. Structure Observed MS (M+1)
180 460
181 Me0 «A-p n'N f z 460
182 442
183 A-Λ? o—< 502
184 A-Λ? MeO''k'^V-N O-x F 502
299
Cmp No. Structure Observed MS (M+1)
185 .A»? 459
186 (Α__Λ° ix^tTzN \ ' ρχ1χ#ΐΓΜ\ ?0Me /1./-7 396
187 OEt n'N 410
188 (Α-Λ F oAT 476
189 A/?-? Va, 476
190 Α/ΐ χχΓΥ ’kl·» 486
300
Cmp No. Structure Observed MS (M+1)
(91 “ο-Λχ 403
192 C^-CN 441
(93 F CN XX > ÎF’ ΧΧ 453
194 Λ-Λ? MecX^X jO 440
195 MeoX^N-XjCjf 458
196 ιύνΧν MoX^Lp'çr F 403
301
Cmp No. Structure Observed MS (M+1)
197 r\-cN 389
198 /°'N Cl 384
199 Ά jCO Αω 426
200 α κΗΌ 414
201 Α/« XX'T' 450
302
Cmp No. Structure Observed MS (M+1)
202 α 443
203 485
204 α ίΗΌ 436
205 α 388
206 ry-AJU N'N 412
207 î^vcn XX j o n 369
302
303
Cmp No. Structure Observed MS (M+1)
208 Λ-cn jCO O C! 403
209 A-cn h.0OnJjÇ>° 403
210 A-CN 370
211 xçf / 347
212 J3S 423
213 o~ JCQ· **«^Y'X 441
304
Cmp No. Structure Observed MS (M+1)
214 xcf 437
215 fA/0'7 c,MrL^ n'N 360
216 Λ-Βγ N'N 395
217 Λ-βγ n'N 381
218 XX j Ni ./Y N o 400
219 JCQ <5 λ,.νΜ N 0 416
305
Cmp No. Structure Observed MS (M+1)
220 370
221 xx 5 ό MeCT^\A \=/ Q-^ 384
222 Λ-cn “•XXjVAA nîn 403
223 A-cn XX j k MeO^^N-Â O-Z \ nX/-^ v/ m‘N 403
224 λ-™ Μ·οΧΧ£ο-0 n'n 385
225 d*>-« ΓΎ > Br •«•'An-A’-q n''N 464
306
Cmp No. Structure Observed MS (M+1)
226 O~cn 'N 389
227 f%y-cN XX j z=<F Cl ί1 NV 407
228 aX^X°-OF ruN 407
229 ^B, ΡΎ^ΖΝ~\ Z-3 1X3 Q yy NV 411
230 (A-/·? FrrNV NV O F 474
307
Cmp No. Structure Observed MS (M+1)
» 231 χζΓ-ν Ο F 460
232 Λ-Br 439
233 -N Ρ-Ν cl Ν ν·Ν 383
234 ^Γ/>-Λ μ'Ν 383
235 Q-.Br 440
236 Q— Η’“^νχ) 384
308
Cmp No. Structure Observed MS (M+1)
237 n'N 385
238 „N--/ N> 371
239 clAy°s n'N 413
240 XXJ ^0 413
241 HiC0 Q-XO 385
242 r-wX «y ciX^y>_A N'N 447
309
Cmp No. Structure Observed MS (M+1)
243 c,^wo- n'N 385
244 N N'N 383
245 'N 445
246 n'N 411
247 Λ-* N-n'A N zNH 390
310
Cmp No. Structure Observed MS (M+1)
24S A-* O 430
249 A-X XO N n'N 369
250 XCN Me<XAxO N- M'N 362
251 Q-™ MeO'Û'N-l O Kl, z~7 N'N 376
252 ' x 0CN MeO^^N-T_N-/ 364
253 O- Q .XÇfo n'n' 4&
Cmp No. Structure Observed MS (M+1)
254 'N 426
255 ^N /°'N n'N 398
256 'N 326
257 ^-N P'N />-<χ Λ χχ□ N V MeO^— Q 364
258 -N P'N χχΝ:Α W 336
259 XUX N'n 399
312
Cmp No. Structure Observed MS (M+1)
260 λΛ jCO Nu m'N 461
261 n'N 340
262 XCN XXX NV 381
263 Λ CI^^V-N (Ί ί 'N 447
264 P p $ Vi >i tf o 447
265 «Α-/Ί ΓΠ ' a—J. JL· JJ ΌΟ M CI^ÏX 461
313
Cmp No. Structure Observed MS (M+1)
266 467
267 'N 427
268 A0 475
270 Αλ 475
271 'N 461
272 AA MeOxS''Z*%~Z —-. o-çy 440
314
Cmp No. Structure Observed MS (M+1)
273 P'N MeO^^^N-^ /=\ Q-Q 412
274 _cçT° Cl Ip' 409
275 461
276 473
277 •cef^ 378
278 'N 397
315
Cmp No. Structure Observed MS (M+1)
279 Λ-COÆt jQQ ΜβΟ^^Ν-4/==Λ cr 436
280 417
281 “Ar'UY n*N 1 411
282 n‘N 385
283 -N 385
284 ixkV 'N 403
316
Cmp No. Structure Observed MS (M+1)
285 402
286 “''XfVY n'n i 413
287 a-A. jCO N 413
288 o b III 354
289 xVAo Q 399
290 XA MeO'A^N--/ 399
317
Cmp No. Structure Observed MS (M+1)
291 aF W 337
292 Ô-Ai XX J V 337
293 416
294 aax _.n-A n XL0' n'N 385
295 aax /v/A n “FF N'N 385
296 aaï z^AH hr n'N 383
318
Cmp No. Structure Observed MS (M+1)
297 /°Ί °χ n'N 399
298 xx^3 381
299 xHXX) XX n 461
300 A-<Q XXy a>XXo- NX 385
301 A-Ù njO 437
302 αΛ rfX 'N 435
319
Cmp No. Structure Observed MS (M+1)
303 _P-N ci^7TN\ /°^CF3 N '/ N'N 451
304 nrNVNO Œ,CF3 515
305 βχΟψγ 368
306 'X? αζΟ9~Ύ 382
307 Λ-ρ XX > F n'N 434
308 Λ-COîS aXj-V-CF1 n'N 442
320
Cmp No. Structure Observed MS (M+1)
309 Α-φ Cl Ap'·'’ 439
310 ô—b 444
311 ô—b m'N 430
312 ATY jCO ^'N 4SI
313 -.N-A 'o 'N 513 .
[0589[ Implementing reactions similar and analogous to those shown in Schemes 1 through 37, the following compounds are also specifically contemplated in this application
321
322
323
324
325
jCfV 11 rWl
i^N>—4°'V ciMtVA cXX X^-Q-
Λ-CN xo CIX-^
mn-o rXX
ck03X
fXX a-u
ciO9^
326
327
328
Example 105: Assessing a5-contalning GABAa Receptor (GABAaR) positive allosteric modulator activity [0590] Step 1: Establish clones of GABAaR subunits(a5, β3, γ2, al, a2 and a3) and préparé the corresponding cRNAs: Human clones of GABAa-R α5, β3, γ2, α I, α2 and α3 subunits are obtained from commercial resources (e.g., OriGene, http://www.origene.com and Genescript, http://www.genescript.com). These clones are engineered into pRC, pCDM, pcDNA, and pBluescript KSM vector (for oocyte expression) or other équivalent expression vectors. Conventîonal transfection agents (e.g., FuGene, Lipofectamine 2000,
329 or others) are used to transiently transfect host cells.
[0591] Step 2 - Functional GABAffl Assay of α5β3γ2, α!β3γ2, α2β3γ2, and α3β3γ2, subtypes inXenopus oocyte expression system: cRNAs encoding α5, β3, γ2, al, α2 and α3 subunits are transcribed in vitro using T3 mMESSAGE mMACHINE Kit (Ambion) and injected (in a ratio of α:β:γ = 2:2:1 or other optimîzed conditions) into oocytes freshly prepared from Xenopus laevis. After two days of culturing, GABA-gated Clcurrents from oocytes are performed using TEVC setups (Wamer Instruments, Inc., Foster City, CA). GABA, benzodiazépine, and diazepam are used as reference compounds to validate the system.
[0592] Step 3 - Evaluate test compounds for positive allosteric modulator activity on the α5β3γ2 subtype and test off-target activity on the al to a3 coupledβ3γ2 subtypes when the EC50=5pMselectivity cut-offis reached: The GABA-gated Cl- current from oocytes are measured in the TEVC setup in the presence of the test compounds. The positive allosteric modulator activity of each the test compounds is tested în a 5-poînt dose15 response assay. The test compounds include some reference compounds (literature EC50 values for the α5β3γ2 subtype are in the range of 3-10 μΜ). EC50s in the α5β3γ2 subtype are obtained for each compound. If the EC50 in α5β3γ2 is < 5μΜ, then the EC50 of the other three subtypes (alβ2γ2, α2β3γ2, and α3β3γ2) is further determined individually in order to test for selectivity of the compounds in the α5β3γ2 subtype over 20 other subtypes.
[0593] Step 4 - Evaluate further test compounds on the α5β3γ2 subtype and test offtarget activities when the EC50=0.5pMselectivity cut-offis reached: The second batch of test compounds are tested using the same strategy, but with a lower EC50 cutofï (0.5 μΜ). Agaîn, the EC50s of the α5β3γ2 subtype for each of the compounds is determined.
The al to a3 coupled β3γ2 subtypes are tested only if the EC50 for the a5-containing receptor is < 0.5 μΜ.
330
Example 106: Evaluating Compounds for Binding and Positive Allosteric Modulator Activity on the GABAa “5 Receptors
64) Binding activity of test compounds on GABAaR [0594] Tissue culture and Membrane Préparation: The binding was performed on Ltk ceils stably expressing GABAa receptors: α!βΐγ2, α2β3γ2, α3β3γ2 and α5β3γ2 (provided by Merck Co., NJ, USA). Ceils were seeded in 100 mm culture plates in DMEM/FI2 medium containing 10% sérum and antibiotics in 5% CO2 and ailowed to grow for 1-2 days. GABAaR expression was then induced by dexamethasone as follows: 0.5 μΜ for I day for a5 containing and 2 μΜ for 3 days for ai, a2 and a3 containing GABAaRs. After 10 induction, ceils were collected by scraping into Dulbecco’s Phosphate buffered saline (DPBS, pH 7.4, Invitrogen, Carisbad, CA, USA) and centrifuged at 150 x g for 10 min. The pellet was washed twice by re-suspension and centrifugation. The celi pellets from at ieast 5 different preps were combined, suspended in the binding assay buffer (50 mM KH2PO4; 1 mM EDTA; 0.2 M KCI, pH 7.4) and membranes prepared by sonication (3-5 15 times, 30 sec) using Branson Sonifier 150 (G.Heinmann, Germany). Protein content was determined using BCA assay (Βΐο-Rad Labs, Reinach, Switzerland) with Bovine Sérum Aibumin (Sigma Aldrich, St. Louis, MO, USA) as the standard. Aliquots were prepared and stored at -20°C for further use in binding assays.
[0595] Ligand Binding: Saturation binding curves were obtained by incubating membranes with încreasing concentrations (0.01 -8 nM) of [3H]Rol5-1788 (Flumazepil, 75-85 Ci/mmol, PerkinElmer, MA, USA), with nonspecific binding measured in the presence of 10 μΜ diazepam. Inhibition of [3H]Roi5-1788 binding ofthe test compounds was performed at concentrations ofthe radioligand at or lower than the Kd values for al, a2, a3 and a5 containing GABAaRs determined from the saturation curves.
[0596] Ail binding assays were performed for I h at 4°C in assay buffer. The total assay volume was 0.5 ml containing 0.2 mg/mi protein for a5 and 0.4 mg/ml for al, a2, and a3 containing GABAaR membranes. Incubations were terminated by filtration through GF/B filters using a 24-Ceil Harvestor (Brandel, Gaithersburg, MD, USA) followed by 3 washes with jce-cold assay buffer. Filters were transferred to scintillation vials, 5 ml scintillation liquid added. vortex-mixed and kept in dark. Next day, radioactivity was obtained using a scintillation counter (Beckman Coulter, Brea, CA, USA). Ail assays were performed in triplicate.
331 [0597] Data Analyses: Saturation and inhibition curves were obtained using GraphPad Prism software (GraphPad Software, Inc., CA, USA). The equilibrium dissociation constants (K| values) of the unlabeled ligand were determined using Cheng-Prusoff équation Kj= ICso/(I+S/K<j), where ICso is the concentration of unlabeled ligand that 5 inhibits 50% of [3H] ligand binding, S is the concentration of radioligand and K<j is the equilibrium dissociation constant ofthe radioactive ligand. A log range ofthe compounds (1 nM -10 μΜ) was used to détermine the Kj values which are presented as Mean ± SD from triplicate assays.
(B) positive allosteric modulator activity of test compounds on oSB2y2 suhtvpe G ABAaR 10 [0598] Compounds of the présent invention were initially screened at 100 nM for their ability to potentiate an EC2o concentration of GABA in oocytes containing GABAa receptors (α5β2γ2), using a protocol essentially similar to the one presented above.
[0599] On day 1, 1 ng/32nL of GABAa α5β2γ2 cDNA was injected into one oocyte. Test starts on day 2. The cDNA injected to the oocytes was a mix ofalpha, beta and gamma, their ratio is 1 : l : 10 (by weight) and the total weight of the mixed 3 subunits to be injected in one oocyte was lng în 32 ni volume. The injected oocytes can also be tested on day 3. In such case, the cDNA amount injected to the oocytes should be reduced by 20%.
[0600] Compounds of the présent invention were tested using the following procedures.
106011 GABA dose-resnonse
1) . 8 oocytes were placed în 8 chambers of OpusXpress and superfused with Modified Barth’s Saline (MBS) at 3mL/min. Glass électrodes back-filled with 3M KCI (0.5-3 megaohms) were used. Membrane potential ofoocytes was voltage-clamped at -60mV.
2) . Average EC2q GABA obtained from previous tests were applied for five-six times to 25 stabilize oocytes. Oocytes were washed with MBS for 5-10 min between each GABA applications.
3) . Run GABA dose-response to obtain EC2o GABA value.
[06021 Control test (Diazepam or methyl 3.5-dÎDhenvlDvridazine-4-carboxylate)
1). New oocytes was used to run new test.
332
2) . EC20 GABA was applied for five-six times to stabilize oocytes. Oocytes were washed with MBS for 5-10 min between each GABA applications.
3) . EC20 GABA was applied to obtain current (Igaba). Oocytes were washed with MBS for 5-10 min.
4) . 1 μΜ dîazepam or methyl 3,5-diphenylpyridazine-4-carboxy late was pre-applied for 40 sec, followed by co-application of ΙμΜ dîazepam or methyl 3,5-diphenylpyridazine-4carboxylate and EC20 GABA to obtain It«t- ït«t was divided by Igaba to obtain potentîation (%).
106031 Test compounds at multiple doses
1) . Repeat the above steps 1), 2) and 3) in the control test.
2) . The first concentration ofa test compound was pre-applied for 40 sec followed by coapplication of the test compound of the same concentration and EC20 GABA to obtain
Im. Divîde Itest by Igaba to obtain potentîation (%).
3) . Discard ail tested oocytes, new oocytes were used and the above steps I ) and 2) were repeated to test second concentration of the same compound. Each oocyte was used for only one concentration test for a single test compound. The steps were repeated for other test compounds.
[0604] In some embodiments, the compounds of this application hâve a binding affinity (as represented by Kj) at a5-containing GABAaRs of less than 200 nM, less than 180 nM, less than 150 nM, or less than 100 nM. In some embodiments, the compounds ofthis application hâve a binding affinity (as represented by K,) at a5-containing GABAaRs of less than 50 nM. In some embodiments, the compounds of this application hâve a binding affinity (as represented by K() at a5-containîng GABAaRs of less than 10 nM.
[0605[ In some embodiments, the compounds ofthis application are sélective for a5containing GABAaRs over al -containing GABAaRs. In some embodiments, the compounds ofthis application are more than 50-fold, more than IOO-fold, more than 500fold or more than 1000-fold sélective for a5-containing GABAaRs over al-containing GABAaRs.
[0606] In some embodiments, the compounds of this application hâve an EC50 at the a5-containing GABAaRs of less than 500 nM, less than 100 nM or less than 50 nM. In
333 some embodiments, the compounds ofthis application hâve an ECjo atthe a5-containing GABAaRs of less than 25 nM.
[0607] In some embodiments, the compounds of this application potentiate a5containing GABAaRs for more than 10%, more than 25%, more than 50%, or more than
75% at 100 nM. In some embodiments, the compounds ofthis application potentiate a5containing GABAaRs for more than 10%, more than 25%, more than 50%, or more than 75% at 1000 nM.
[0608] Screening results of the binding and PAM functional activity tests are summarized in Tables 1 and 2 below.
[0609] The following Table 1 illustrâtes the ranges of GABA a5 binding Ki’s associated with compounds ofthis invention: Table 1
GABA a5 Binding Ki Values (nM)
< 99 nM 100-1000 nM > 1000 nM
Compounds 1,2,3, 4,6,7,8,9,10,11, 12,44,55, 101,103, 105,107,108,114, 128,153,158,162, 163,164,166,169, 171,172,173,174, 175,177,179,5,47, 48,49,51,52,53, 54,56,102,104, 106,111,112,118, 120,126,127,130, 133,137,145,147, 148,149,155, 156, 157, 160,165,168, 178,45,46,109, 122,129,132,150, 151,159,161,167, 176,180-190, 194-199,202,203, 205-210,216,217, 218,222,223-227, 230,232,233,235, Compounds 50, 110,113,115,119, 124,125,134,136, 138,139,141,143, 144, 146,170,191, 200,201,219,220, 237,240,246,247, 248,265-267,273, 274,281,283,284, 286,287,292,297300,303 Compounds 116, 117,121,123,131, 135,140,142,143, 152,154,192,193, 204,221,229,231, 234,239,250-253, 262,272, 279
334
236,238,241-245, 249,254-261,263, 264,268-271,275278,280,282,285, 288-291,293-296, 301,302,304
[0610] The following Table 2 illustrâtes the ranges of GABA a5 functional potentiation associated with compounds of this invention:
Table 2
GABA a5 Functional Data
20-49% @ 100 nM >50%@ !00nM
Compounds 1,2,9,11, 48,45,55,109,110, 111, 118, 120,126,127, 128, 130, 132,137,147, 148, 153, 155,158,162, 163, 175,180-184,187-189, 191,195,196,198,199, 202,203,205,207,210, 212, 213,222,224,225, 226,238,243,249,254257,264,290,293 Compounds 113, 114,145, 149,160, 171,172,173,174, 176,177,178,179, 185,186, 194,, 271
[0611] Selected compounds of this invention demonstrate > 10-fold binding selectivîty versus GABA al, GABA a2, or GABA a3.
Example 107: Effect of Methyl 3^-diphenylpyridazine-4-carboxylate in Aged10 Impaired (AI) Rats [0612] Methyl 3,5-diphenylpyridazine-4-carboxylate, corresponding to compound number 6 in van Niel et al. J. Med. Chem. 48:6004-6011 (2005), is a sélective a5containing GABAa R agonist. It has an a5 in vitro efficacy of +27 (EC20). The effect of methyl 3,5-diphenylpyridazine-4-carboxylate in aged-impaired rats was studied using a
RAM task. Moreover, receptor occupancy by methyl 3,5-dîphenylpyridazine-4carboxylate in a5-containing GABAa receptor was also studied.
335 (A) Effect of Methyl 3.5-diphenvlnvridazine-4-carboxvlate in Aeed-Impaired Rats Usine a Radial Arm Maze (RAM) Behavioral Task [0613] The effects of methyl 3,5-diphenyIpyridazine-4-carboxy!ate on the in vivo spatial memory rétention ofaged-impaired (Al) rats were assessed in a Radial Arm Maze (RAM) behavioral task using vehicle control and four different dosage levels of methyl 3,5-diphenylpyridazine-4-carboxylate (0.1 mg/kg, 0.3 mg/kg, 1 mg/kg and 3 mg/kg, ip). RAM behavioral tasks were performed on eight Al rats. Ali five treatment conditions (vehicle and four dosage levels) were tested on ail eight rats.
[0614] The RAM apparatus used consisted of eight equidistantly-spaced arms. An elevated maze arm (7 cm width x 75 cm length) projected from each facet of an octagonal center platform (30 cm diameter, 51.5 cm height). Clear side walls on the arms were 10 cm high and were angled at 65° to form a trough. A food well (4 cm diameter, 2 cm deep) was located at the distal end of each arm. Froot Loops™ (Kellogg Company) were used as rewards. Blocks constructed of Plexiglas™ (30 cm height x 12 cm width) could be positioned to prevent entry to any arm. Numerous extra maze eues surrounding the apparatus were also provided.
[0615] The AI rats were initîally subjected to a pre-training test (Chappell et al. Neuropharmacology 37:481-487, 1998). The pre-training test consisted of a habituation phase (4 days), a training phase on the standard win-shift task (18 days) and another 20 training phase ( 14 days) in which a brief delay was imposed between présentation of a subset ofarms desîgnated by the expérimenter (e.g., 5 arms available and 3 arms blocked) and completion of the eight-arm win-shift task (i.e., with ail eight arms available).
[0616] ln the habituation phase, rats were familiarized to the maze for an 8-minute session on four consecutive days. In each of these sessions, food rewards were scattered 25 on the RAM, initially on the center platform and arms and then progressively confined to the arms. After this habituation phase, a standard training protocol was used, in which a food pellet was located at the end ofeach arm. Rats received one trial each day for 18 days. Each daily trial terminated when ali eight food pellets had been obtained or when either 16 choices were made or 15 minutes had elapsed. After completion of this training phase, a second training phase was carried out in which the memory demand was increased by imposing a brief delay during the trial. At the beginning of each trial, three
336 arms of the eight-arm maze were blocked. Rats were allowed to obtain food on the five arms to which access was permitted during this initial “information phase” of the trial. Rats were then removed from the maze for 60 seconds, during which time the barriers on the maze were removed, thus allowing access to ail eight arms. Rats were then placed back onto the center platform and allowed to obtain the remaining food rewards during this “rétention test” phase ofthe trial. The identity and configuration ofthe blocked arms varied across trials.
[0617] The number of “errors” the AI rats made during the rétention test phase was tracked. An error occurred in the trial if the rats entered an arm from which food had already been retrieved in the pre-delay component ofthe trial, or ifthe rat re-visited an arm in the post-delay session that it had already vïsited.
[0618] After completion of the pre-training test, rats were subjected to trials with more extended delay intervals. Le., a two-hour delay, between the information phase (présentation with some blocked arms) and the rétention test (présentation ofall arms). During the delay interval, rats remained off to the side ofthe maze in the testing room, on caris in their individual home cages. AI rats were pretreated 30-40 minutes before daily trials with a one-time shot ofthe following five conditions: 1) vehicle control - 5% dimethyl sulfoxide, 25% polyethylene glycol 300 and 70% distïlled water; 2) methyl 3,5diphenylpyridazine-4-carboxylate at 0.1 mg/kg; 3) methyl 3,5-diphenylpyridazine-4carboxylate at 0.3 mg/kg; 4) methyl 3,5-diphenylpyridazine-4-carboxylate at 1 mg/kg); and 5) methyl 3,5-diphenylpyridazîne-4-carboxylate at 3 mg/kg; through intraperitoneal (i.p.) injection. Injections were given every other day with intervening washout days. Each Al rat was treated with ail five conditions within the testing period. To counterbalance any potential bias, drug effect was assessed using ascending-descending dose sériés, i.e., the dose sériés was given first in an ascending order and then repeated in a descending order. Therefore, each dose had two déterminations.
[0619] Parametric statistics (paired t-tests) was used to compare the rétention test performance ofthe AI rats in the two-hour delay version ofthe RAM task in the context of different doses of methyl 3,5-diphenylpyridazine-4-carboxylate and vehicle control (see Figure 1). The average numbers of errors that occurred in the trials were significantly fewer with methyl 3,5-diphenylpyridazine-4-carboxylate treatment of 3
337 mg/kg (average no. of errors ± standard errer of the mean (SEM) = 1.31 ± 0.40) than using vehicle control (average no. of errors ± SEM = 3.13 ± 0.62). Relative to vehicle control treatment, methyl 3,5-diphenylpyridazine-4-carboxylate significantly improved memory performance at 3 mg/kg (t(7) = 4.233, p = 0.004).
[0620] The therapeutîc dose of 3 mg/kg became ineffective when the AI rats were concurrently treated with 0.3 mg/kg of TB21007, a a5-containing GABAa R inverse agonist. The average numbers of errors made by rats with the combined TB21007/ methyl 3,5-diphenylpyridazine-4-carboxylate treatment (0.3 mg/kg TB21007 with 3 mg/kg methyl 3,5-diphenylpyridazine-4-carboxylate) was 2.88 ± 1.32, and was no different from rats treated with vehicle control (3.13 ±1.17 average errors). Thus, the effect of methyl 3,5-diphenylpyridazine-4-carboxylate on spatial memory îs a GABAa a5 receptor-dependent effect (see Figure 1).
(B) Effect ofMethyl 3.5-diphenvlpvridazine-4-carboxvlate on a5-containin£ GABAa Receptor Occupancy
Animais [0621] Adult male Long Evans rats (265-295 g, Charles River, Portage, MI, n=4/group) were used for GABAacc5 receptor occupancy studies. Rats were individually housed in ventiiated stainless-steel racks on a 12:12 light/dark cycle. Food and water were available ad libitum. In additional studies to evaluate compound exposures at behavioraily active doses, young or aged Long Evan rats (n= 2-4/group) were used for these studies.
Compounds [0622] Ro 15-4513 was used as a receptor occupancy (RO) tracer for GABAacxS receptor sites in the hippocampus and cerebellum. Ro 15-4513 was chosen as the tracer based on its seiectivity for GABAaoiS receptors relative to other alpha subunit containing GABAa receptors and because it has been successfuliy used for GABAAa5 RO studies in animais and humans (see, e.g., Lingford-Hughes et al., J. Cereb. BloodFlow Metab. 22:878-89 (2002); Pym et al, Br. J. Pharmacol. 146: 817-825 (2005); and Maeda et ai., Synapse 47:200-208 (2003)). Ro 15-4513 (1 pg/kg), was dissolved in 25% hydroxylpropyl beta-cyclodextrin and administered i.v. 20’ prior to the RO évaluations. Methyl 3,5-diphenylpyridazine-4-carboxylate (0.1 - 10 mg/kg) was synthesized by Nox Pharmaceuticals (India) and was dissolved in 25% hydroxyl-propyl beta-cyclodextrin and
338 administered i.v. 15’ prior to tracer injection. Compounds were administered in a volume of 0.5 ml/kg except for the highest dose of methyl 3,5-diphenylpyridazine-4-carboxylate (10 mg/kg) which was administered in a volume of 1 ml/kg due to solubility limitations.
Tissue préparation and analysis {0623] The rats were sacrificed by cervical dislocation 20’ post tracer injection. The whole brain was rapidly removed, and lightly rinsed with stérile water. Trunk blood was collected in EDTA coated eppendorf tubes and stored on wet ice until study completion. Hippocampus and cerebellum were dissected and stored in 1.5 ml eppendorf tubes, and placed on wet tce until tissue extraction. In a drug naïve rat, six cortical brain tissues samples were collected for use in generating blank and standard curve samples.
[0624] Acetonitrile containing 0.1% formîc acid was added to each sample at a volume of four times the weight of the tissue sample. For the standard curve (0.1-30 ng/g) samples, a calculated volume of standard reduced the volume of acetonitrile. The sample was homogenîzed (FastPrep-24, Lysing Matrix D; 5.5 m/s, for 60 seconds or 7-8 watts power using sonie probe dismembrator; Fisher Scïentifie) and centrifuged for 16-minutes at 14,000 rpm. The (100 μΐ) supematant solution was diluted by 300 μΙ of stérile water (pH 6.5). This solution was then mixed thoroughly and analyzed via LC/MS/MS for Ro 15-4513 (tracer) and methyl 3,5-diphenylpyridazine-4-carboxylate.
[0625] For plasma exposures, blood samples were centrifuged at 14000 rpm for 16 minutes. After centrifuging, 50uI of supematant (plasma) from each sample was added to 200 μΐ of acetonitrile plus 0.1% formic acid. For standard curve (1-1000 ng/ml) samples, a calculated volume of standard reduced the volume of acetonitrile. Samples were sonicated for 5 minutes in an ultrasonic water bath, followed by centrifugation for 30 minutes, at 16000 RPM. lOOul of supematant was removed from each sample vial and placed in a new glass auto sample vial, followed by the addition of 300 μΙ of stérile water (pH 6.5). This solution was then mixed thoroughly and analyzed via LC/MS/MS for methyl 3,5-dipheny1pyridazine-4-carboxylate.
[0626] Receptor occupancy was determined by the ratio method which compared occupancy in the hippocampus (a région of high GABAa<x5 receptor density) with occupancy in the cerebellum (a région with low GABAa<x5 receptor density) and
339 additionally by a high dose ofthe GABAacc5 négative allosteric modulator L-655,708 (10 mg/kg, i.v.) to define full occupancy.
[0627] Vehicle administration followed by tracer administration of 1 pg/kg, i.v., of Ro 15-4513 resulted in > 5-fold higher levels of Ro 15-4513 ln hippocampus (1.93 ± 0.05 ng/g) compared with cerebellum (0.36 ± 0.02 ng/g). Methyl 3,5-diphenylpyridazine-4carboxylate (0.01 -10 mg/kg, i.v. ) dose-dependently reduced Ro 15-4513 binding in hippocampus, without affecting cerebellum levels of Ro 15-4513 (Figure 2) with a dose of 10 mg/kg, i.v., demonstrating >90% occupancy (Figure 3). Both methods of calculating RO yieiding very similar results with ED50 values for methyl 3,5diphenylpyridazine-4-carboxylate as 1.8 mg/kg or 1.1 mg/kg based on the ratio method or using L-755,608 to define occupancy.
[0628] Methyl 3,5-diphenylpyridazine-4-carboxylate exposure was below the quantification limits (BQL) at 0.01 mg/kg, i.v., in both plasma and hippocampus and but was détectable at low levels ln hippocampus at 0.1 mg/kg, i.v. (see Table 3). Hippocampal exposure was linear as a 10-fold increase in dose from 0.1 to I mg/kg, i.v., resulted in a 12-fold increase in exposure. Increasing the dose from 1 to 10 mg/kg, i.v., only Increased the exposure by ~5-fold. Plasma exposure increased 12-fold as the dose increased from 1 to 10 mg/kg, i.v.
Table 3: % GABAa a5 Receptor Occupancy by methyl 3,5-diphenylpyridazine-4carboxylate (0.01-10 mg/kg, i.v.). Hippocampus and Plasma Exposure of methyl 3,5diphenylpyridazine-4-carboxylate by Treatment Group in young Long Evans rats.
Dose (mg/kg, i.v.) %RO (L-655,708 Method) (SEM) %RO (Ratio Method) (SEM) Plasma ng/mL (SEM) Hippocampus ng/g (SEM)
0.01 19.2(11.1) 15.7 (9.1) BQL BQL
0.1 16.4 (4.9) 13.4(4.0) BQL 14.6 (3.5)
1 38.5(11.2) 31.5 (9.1) 62.8(6.1) 180.0(10.3)
10 110.0(6.6) 90.2 (5.4) 763.5 (85.7) 947.2 (51.3)
340 [0629] Additional studies were conducted in aged Long-Evans rats in order to détermine the exposures at the behaviorally relevant doses in the cognition studies. Exposure in young Long-Evans rats was also determined to bridge with the receptor occupancy studies that were conducted in young Long-Evans rats. Exposures in young and aged Long-Evans rats were relatively similar (Table 4, Figure 4). Increasing the dose 3-fold from I to 3 mg/kg, ip resulted in a greater than dose-proportional increase in exposure in young and aged rats in both hippocampus and plasma with increases ranging from 4.5 to 6.6-fold.
Table 4: Hippocampus and Plasma Exposure of methyl 3,5-diphenylpyridazine-4carboxylate in Young Long Evans Rats by Treatment Group
Young Young Aged Aged
Dose (mg/kg, Ip) Hippocampus ng/g (SEM) Plasma ng/mL (SEM) Hippocampus ng/g (SEM) Plasma ng/mL (SEM)
1 25.9(1.7) 20.0(1.4) 38.8(21.7) 45.2 (29.6)
3 129.1 (22.4) 132.9 (19.5) 177.5 (19.5) 196(18.2)
[0630] In the RO studies, an exposure of 180 ng/g in hippocampus ( 1 mg/kg, i.v.) represented 32-39% receptor occupancy depending on method used to détermine RO. This exposure is comparable to that observed in aged rats at 3 mg/kg, i.p., suggesting that 30-40% RO is required for cognitive efficacy in this model.
[0631] These studies demonstrated that methyl 3,5-diphenylpyridazine-4-carboxylate produced dose-dependent increase in GABAa a5 receptor occupancy. Methyl 3,5diphenylpyridazine-4-carboxylate also demonstrated good brain exposure with brain/plasma ratios>l. The studies further demonstrated that methyl 3,5diphenylpyridazine-4-carboxylate was producing its cognitive enhancing effects by positive allosteric modulation at the GABAa a5 subtype receptor.
Exempte 108: Effect of Ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5a][l^,4]triazolo[43-d][l,4]diazepine-10-carboxylate in Aged-Impaired (AI) Rats [0632] Ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3d][l,4]diazepîne-10-carboxylate, corresponding to compound number 49 in Achermann et
341 al. Bioorg. Med. Chem. Lett., 19:5746-5752 (2009), is a sélective a5-containing GABAa R agonist.
[06331 The effect of ethyl 3-methoxy-7-methy l-9H-benzo[f]imidazo[ 1,5a][l,2,4]trîazolo[4,3-d][l,4]dîazepine-10-carboxylate on the in vivo spatial memory rétention of aged-impaîred (Al) rats was assessed în a Radial Arm Maze (RAM) behavioral task that is essentially similar to the task as described in Example 107 (A), using vehicle control (25% cyclodextrin, which was tested 3 times: at the beginning, middle and end of ascending/descending sériés) and six different doses levels (0.1 mg/kg, 0.3 mg/kg, 1 mg/kg, 3 mg/kg, 10 mg/kg and 30 mg/kg. each dose was tested twice) of ethyl 3-methoxy-7-methyl-9H-benzo[f]imÎdazo[ 1,5-a][ 1,2,4]triazolo[4,3-
d][l,4]diazepine-10-carboxylate. The same experiment was repeated using the same vehicle control and doses of ethyl 3-methoxy-7-methyl-9H-benzo[f]îmidazo[l,5a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate, where the vehicle control was tested 5 times, the 3 mg/kg dose of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-
a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate was tested 4 tîmes, and the other doses of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3d][l,4]diazepine-10-carboxylate were tested twice.
[0634] Parametric statistics (paired t-tests) was used to compare the rétention test performance of the Al rats in the four-hour delay version of the RAM task in the context of different doses of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5a][l,2,4]triazolo[4,3-d][I,4]dîazepine-10-carboxylate and vehicle control (see Figure 5). Relative to vehicle control treatment, ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5a][l,2,4]triazo!o[4,3-d][l,4]diazepine-IO-carboxylate significantly improved memory performance at 3 mg/kg (t(7) = 4.13, p = 0.004, or t(7) = 3.08, p = 0.018) and at 10 mg/kg (t(7) = 2.82, p=0.026).
[0635] The effect of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5a][l,2,4]triazolo[4,3-d][l,4]diazep!ne-10-carboxylate on a5-containïng GABAa receptor occupancy was also studied following a procedure that is essentially similar to the one as described în Example 107(B) (see above). This study demonstrated that ethyl 3-methoxy30 7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-l0-carboxylate (0.01 - 10 mg/kg, i.v. ) reduced Ro 15-4513 binding în hippocampus, without affecting
342 cerebellum levels of Ro 15-4513 (Figure 6) with a dose of 10 mg/kg, i.v., demonstrating >90% occupancy (Figure 7).
Example 109: Effect of 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7dihydro-2-benzothlophen-4(5H)-one ln Aged-Impalred Rats Using a Morris Water 5 Maze Behavioral Task [0636] 6,6dimethyi-3-(3-hydroxypropyi)thio-l-(thiazol-2-yi)-6,7-dihydro-2benzothiophen-4(5H)-one, corresponding to compound 44 in Chambers et ai. J. Med.
Chem. 46:2227-2240 (2003) îs a sélective a5-containîng GABAa R agonist.
[0637] The effects of 6,6 dimethyi-3-(3-hydroxypropy l)thio-1 -(thiazol-2-yl)-6,710 dihydro-2-benzothiophen-4(5H)-one on the in vivo spatial memory rétention of agedimpaîred (AI) rats were assessed în a Morris water maze behavioral task. A water maze is a pool surrounded with a novel set of patterns relative to the maze. The training protocol for the water maze may be based on a modified water maze task that has been shown to be hippocampai-dependent (de Hoz et al., Eur. J. Neurosci., 22:745-54,2005;
Steele and Morris, Hippocampus 9:118-36, 1999).
[0638] Cognitîveiy impaired aged rats were implanted unilaterally with a cannula into the latéral ventricle. Stereotaxic coordinates were 1.0 mm posterior to bregma, 1.5 mm latéral to midline, and 3.5 mm ventral to the skuli surface. After about a week of recovery, the rats were pre-trained in a water maze for 2 days (6 triais per day) to locate a 20 submerged escape platform hidden undemeath the surface of the pool, in which the escape platform location varied from day to day. No intracerebroventricuiar (ICV) infusion was given during pre-training.
[0639] After pre-training, rats received ICV infusion of either 100 pg 6,6 dimethyi-3(3-hydroxypropyl)thio-I-(thiazol-2-yl)-6,7-dihydro-2-benzothîophen-4(5H)-one (n = 6) in 25 5 pl DMSO or vehicle DMSO (n = 5) 40 min prior to water maze training and testing.
Training consisted of 8 triais per day for 2 days where the hidden escape platform remained in the same location. Rats were given 60 seconds to locate the platform with a 60 seconds inter-trial intervai. The rats were given a probe test (120 seconds) 24 hr. after the end of training where the escape platform was removed. During the training, there 30 were 4 blocks, where each block had 4 training trials.
343
[06401 Rats treated with vehicle and 6,6 dimethyl-3-(3-hydroxypropy I) thio-1 -(thiazol2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one found the escape piatform about the same time at the beginning oftraining (block 1 ). In this block oftraining, rats treated with vehicle and 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-25 benzothiophen-4(5H)-one both spent about 24 seconds to find the escape piatform. However, rats treated with 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-y 1)-6,7dihydro-2-benzothiophen-4(5H)-one were able to find the piatform more proficiently (i.e., quicker) at the end oftraining (block 4) than those treated with vehicle alone. In block 4, rats treated with 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-210 benzothiophen-4(5H)-one spent about 9.6 seconds to find the escape piatform, while rats treated with vehicle spent about 19.69 seconds. These results suggest that 6,6 dimethyl-3(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one improved the learning ofthe water maze task în rats (see Figure 8(A)).
[0641] During a test trial 24 hr. after training, the escape piatform was removed. The search/swim pattern of the rats was used to measure whether the rats remember where the escape piatform was located during pre-trial training in order to test for the long-term memory of the rats. In this trial, “target annulus” is a designated area 1.5 times the size of the escape piatform around the area where the piatform was located during pre-trial training. “Opposite annulus*’ is a control area ofthe same size as the size ofthe target annulus, which is located opposite to the target annulus in the pool. If the rats had good long term memory, they would tend to search in the area surrounding the location where the piatform was during lhe pre-trial training (i.e., the “target” annulus; and not the “opposite” annulus). “Time in annulus” is the amount of time in seconds that the rat spent in the target or opposite annulus area. “Number (#) of crossings” in annulus is the number of times the rat swam across the target or opposite annulus area.
[0642] Rats received vehicle spent the same amount of time in the target annulus and opposite annulus, indicating that these rats did not seem to remember where the piatform was during the pre-trial training. By contrast, rats treated with 6,6 dimethyl-3-(3hy droxypropy l)th io-1 -(thiazol-2-y l)-6,7-dîhy dro-2-be nzoth iophen-4(5 H)-one spent significantly more time in the target annulus, and crossed the “target annulus” more often, as compared to the time they spent in, or the number of times they crossed the “opposite annulus”. These results suggest that 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-218506
yl)-6,7-dihydro-2-benzothiophen-4(5H)-one improved the long-term memory of rats în the water maze task (see, Figures 8(B) and 8(C)).
[0643] Compounds of the présent invention demonstrated positive allosteric modulatory effect on the GABAa a5 receptor (See, e.g., Example 106). These compounds will enhance the effects of GABA at the GABAa a5 receptor. Therefore, compounds of the présent invention should produce cognitive enhancing effects in aged-impaired animais (such as rats), similar to the effects produced by other GABAa a5 receptor sélective agonists, such as methyl 3,5-diphenylpyridazine-4-carboxylate, ethyl 3-methoxy-7methyl-9H-benzo[f]imidazo[ 1,5-a][l,2,4]triazolo[4,3-d][ 1,4]diazepine-l O-carboxylate, [0 and 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen4(5H)-one (See, e.g., Examples 28-30).

Claims (23)

  1. or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
    U and the two carbon atoms designated by a and β together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
    A is C, CR6, or N;
    B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
    D is N, NR7,0, CR6 or C(R6)2;
    E is N, NR7, CR6 or C(R6)2;
    W is N, NR7, CR6 or C(R6)2;
    X is N, NR7.0, CR6 or C(R6)2;
    Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
    Vis Cor CR6, or when Z is C or CR6, V is C, CR6, or N;
    wherein when the ring formed by X, Y, Z, V and W is
    -SR8, -(CH2)nOR8, -(CH2)nO(CH2)nR8, -(CH2)pR8 and -(CH2)nN(R)R10; and wherein R2 is independently substituted with 0-5 R';
    m and n are independently integers selected from 0-4;
    p is an integer selected from 2-4;
    346 each occurrence of the bond “ ” is either a single bond or a double bond;
    each occurrence of R1, R2, R4, and R5 are each independently selected from:
    halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -S1R3, -N(R)2, -SR, -SOR,
    -SO2R, -SO2N(R)2, -SO3R, -(CR2)i.3R, -(CR2)i.3-OR, -(CR2)o-3-C(0)NR(CR2)o-3R,
    5 -(CR2)o.3-C(0)NR(CR2)o.30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R,
    -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)o-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2, -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R
    10 )2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2. -P(O)(OR)2, and -P(O)(H)(OR);
    R3 is absent or is selected from:
    halogen, -R, -OR, -NO2, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR,
    -SO2R, -SO2N(R)2, -SO3R, -(CR2)t.3R, -(CR2)i-3-OR, -(CR2)o-3-C(0)NR(CR2)o.3R,
    15 -(CR2)o-3-C(0)NR(CR2)o-30R, -C(O)R, -C(O)C(O)R, -C(O)CH2C(O)R, -C(S)R,
    -C(S)OR, -C(O)OR, -C(O)C(O)OR, -C(O)C(O)N(R)2, -OC(O)R, -C(O)N(R)2, -OC(O)N(R)2, -C(S)N(R)2, -(CR2)o-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(O)OR, -N(R)N(R)CON(R)2t -N(R)SO2R, -N(R)SO2N(R)2, -N(R)C(O)OR, -N(R)C(O)R, -N(R)C(S)R, -N(R)C(O)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH)N(R
    20 )2, -C(O)N(OR)R, -C(=NOR)R, -OP(O)(OR)2, -P(O)(R)2, -P(O)(OR)2, and -P(O)(H)(OR);
    each R6 is independently-H or -(Cl-C6)alkyl;
    each R7 is independently-H or -(Cl-C6)alkyl;
    each R8 is independently -(Cl-C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to 1025 membered heteroaryl, wherein each occurrence of R8 is independently substituted with 05 R’;
    each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’;
    30 each R is independently selected from:
    H-, (Cl-C12)-aliphatic-, (C3-C10)-cycloalkyl-,
    347 (C3 -C 10)-cycloalkenyl-, [(C3-C10)-cycloalkyl]-(C 1 -C 12)-aliphatîc-, [(C3-C10)-cycloalkenyl]-(Cl-C12)-aliphatic-, [(C3-C10)-cycloalkyl]-O-(C 1-C12)-al ïphatic-,
    5 [ (C3 -C 10)-cycloalkenyl ]-O-(C 1 -C 12)-al iphatic-, (C6-C10)-aryl-, (C6-C10)-aryl-(C 1 -C 12)al iphat ic-, (C6-C10)-aryl-O-(C 1 -C 12)al iphat i c-, (C6-C 10)-aryl-N(R”)-(C 1 -C 12)al iphat i c-,
    10 3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)aliphatic-,
    5- to 10- membered heteroaryl-,
    15 (5- to 10- membered heteroaryl)-(Cl-C12)-aliphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aliphatic-;
    wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, 20 and S;
    wherein each occurrence of R is independently substituted with 0-5 R’;
    or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or non-aromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2,
    25 wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3-C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
    wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CFj, -OCF3 and 30 N(R)2;
    wherein each occurrence of R” is independently selected from H, -(Cl-C6)-alkyl, -(C1-C6)aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)
    348 aryl-(Cl-C6)-a!kyl-, (5- to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-CI0)aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independentiy substituted with 0-3 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR0, -CFhNfR0)
  2. 2, -C(O)N(R°)2, -C(O)OR°, -NO2, -NCS, -CN,
    -CF3, -OCF3 and -N(R°)2, wherein each occurrence of R° is independentiy selected from: ~(Cl-C6)-al iphatic, (C3-C6)-cycloa!kyl, 3- to 6- membered heterocyclyl, 5- to 10membered heteroaryl-, and (C6-CI0)-aryI-.
    Il, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
    m îs 0-3;
    each R1 is independentiy selected from: halogen, -H, -(Cl-C6)alkyl, -OH, -O((Cl-C6)alkyl), -NO2, -CN, -CF3, and -OCF3, wherein R* is independentiy substituted with 0-5 R’;
    R2 is selected from:
    -H, halogen, -(Cl-C6)alkyl, -OH, -O((Cl-C6)alkyl), -C(O)O((Cl-C6)alkyl), -C(O)NR2, (C6-C10)-aryl-, (C6-CI0)-aryl-(CI-C12)aliphatic-, (C6-C10)-aryl-O-(C 1 -C 12)al iphatic-, (C6-C10)-aryI-N(R”)-(C 1 -C 12)aliphatic-, (5- to 10- membered heteroaryl)-(CI-C12)aliphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)aliphatic-, (5- to 10- membered heteroaryl)-N(R,’)-(Cl-C12)aliphatic-,
    349 (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-O-(Cl-C12)aliphatic-, and (3- to 10- membered hetcrocyclyl)-N(R”)-(Cl-C12)aliphatic-, wherein R2 is independently substituted with 0-5 R’;
    5 R3 is selected from:
    -(Cl-C6)alkyl, -(C2-C6)alkenyl, -OCH, -CN, halogen, -SO2((C6-C10)-aryl), -SO2((C1C6)alkyl), -C(O)N((C1-C6)alkyl)2, -C(0)NH2, -C(0)0((Cl-C6)aIkyl), -C(O)((C1C6)alkyl), -(C6-C10)aryl, 5- to 10- membered heteroaryl, 5- to 10- membered heterocyclyl, wherein R3 is independently substituted with 0-5 R’;
    10 R4 and Rs are each independently selected from -H, halogen and -(Cl-C6)alkyl;
    R6 is selected from -H and -(Cl-C6)alkyl;
    each R is independently selected from:
    H-, (C 1-C12)-aliphatic-,
    15 (C3-C10)-cycloaIkyl-, (C3-C10)-cycloalkenyl-, [(C3-C10)-cycloalkyl]-(Cl-C12)-aliphatic-, [(C3-C10)-cycloalkenylF(Cl*C12)-aliphatic-, [(C3-C10)-cycloalkyl]-O-(C 1 -Cl 2)-aliphatic-,
    20 [(C3-CI0)-cycloalkenyl]-O-(Cl-C12)-aliphatic-, (C6-C10)-aryl-, (C6-C10)-aryl-(Cl-C12)aliphatic-, (C6-C 10)-aryl-O-(C 1 -C 12)aliphatic-, (C6-C10)-aryl-N(R)-(Cl-C12)aliphatic-,
    25 3- to 10- membered heterocyclyl-, (3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-0-(Cl-C12)aliphatic-, (3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)aliphatic-,
    5- to 10- membered heteroaryl-,
    30 (5- to 10- membered heteroaryl)-(Cl-C12)-aliphatic-, (5- to 10- membered heteroaryl)-O-(Cl-C12)-aliphatic-; and (5- to 10- membered heteroaryl)-N(R)-(Cl-C12)-alîphatic-;
    wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O,
    350 and S;
    wherein each occurrence of R is independently substituted with 0-5 R’;
    or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or non-aromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3-C10)cycloalkyl, or a 3- to 10- membered heterocyclyl;
    wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR”, -NO2, -NCS, -CN, -CF3, -OCF3 and N(R”h;
    wherein each occurrence of R” is independently selected from H, -(Cl-C6)-aIkyl, (C3-C6)cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)-alkyl-, (5to 10- membered heteroaryl)-O-(Cl-C6)-aIkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-.
  3. 3. A compound of formula IV:
    IV, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, wherein:
    m is 0-3;
    each R1 is independently selected from: halogen, -H, -(Cl-C6)alkyl, -C^CH, -OH, -O((C1C6)alkyl), -NO21 -CN, -CF3, -OCF3, wherein R1 is independently substituted with 0-5 R’;
    R2 is selected from -OR8, -SR8, -(CH2)nOR8, -(CH2)nO(CH2)„R8, -(CH2)PR8 and
    351
    -(CH2)nN(R”)R10, wherein n is an integer selected from 0-4; p is an integer selected from 2-4; each R8 is independently -(Cl-C6)alkyl, -(C3-C10)-cycloaIkyI, (C6-C10)-aryl, or 5to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’; each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered
    5 heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’; and wherein R2 is independently substituted with 0-5 R’;
    R3 is selected from;
    -H, -CN, halogen (e.g., Br), -(Cl-C6)alkyl, -C^CH, -SO2((Cl-C6)alkyl), -C(O)N((C110 C6)alkyl)2, ), -C(O)NH((C1-C6)aliphatic)2, (C6-C10)-aryl-(ClC12)aliphatic-, -C(O)((Cl-C6)alkyl), -C(O)O((Cl-C6)alkyl), 5- or 6-membered heterocyclyl, and 5- or 6-membered heteroaryl; and wherein R3 is independently substituted with 0-5 R’;
    R4 and R3 are each independently selected from -H, halogen and -(C 1-C6)alkyl;
    15 R6 is selected from -H and -(C 1 -C6)alky 1 ;
    wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CHiOR”, -CH2NR”2, -C(O)N(R”)2, -C(O)OR, -NO2, -NCS, -CN, -CF3, -OCF3 and N(R”)2;
    wherein each occurrence of R” is independently selected from H, -(Cl-C6)-alkyl, (C3-C6)20 cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)aryl-, (5- to 10- membered heteroaryl )-(Cl-C6)-alkyl-, (C6-C10)-aryl-(Cl-C6)-alkyl-, (5to 10- membered heteroaryl)-O-(Cl-C6)-alkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-.
  4. 4. A compound selected from:
    Cmp No. Structure 180 n-'n
    352
    353
    Cmp No. Structure 187 XWN~\ 'n*»- F'+»-N OEt 1-+-7 188 αΛ 189 Α_Λ? fUR1f 190 y k U- 191 192 v zy-cN
    354
    Cmp No. Structure 193 Λ-CN 194 195 λ/2 ^Xn'-V ΜβθΌ^Ν-210Γ ^'N 196 i^CN Meo kAaF 197 cl Α°ΰ 198 XI P'N n'N
    355
    Cmp No. Structure 199 ci^x°O 200 XT' ’Xo 201 A-/'i Me° tp’t 202 ° ιχ°Ό 203 A-A xxX1' '—N 204 <x Χό
    356
    Cmp No. Structure 205 α 0-¾ 206 P'N n'N 207 Λ-cn jCO o 208 λ-™ jCO o 209 A-CN MeO'CÇAZY' i/ 210 îA-gn Λ ) n MeO-AAN^
    357
    358
    Cmp No. Structure 216 ci^^A-n CX / n'N 217 Γ >-Br Ά ci^^A-n cl n'N 218 N-f Λ χχ 5 fX MeCr^A-ZA/ N 'o 219 yNy__= M p XX 5 <5 MeO'”X' ^=/ N'hf q 220 AJ mo MeCT^^N -f y=/ A 221 / <νΝΛ /V MeCT^Al-/ y/ **!>
    359
    Cmp No. Structure 222 -N Γ >-CN ΓΎ T U'N 223 Λ-cn ___ jTY ) S-λ MeO^^N-Z O-Z\ 224 î^Z-cn ΓΎ ) 225 Λ-cn j3Q MeO^^S-Z o-Z) jV>-/ V/ 'N 226 if-X™ Λ N'N 227 Λ-ον jGQ /Ύ Λ.·Λ 'N
    360
    361
    Cmp No. Structure 233 /O’N c,^Ç/>-^0x n-N 234 (A_A AA > “JT'A n'N 235 Γ />-Br 236 H3CoXAlH^q 237 αλ LAw a'N^\,u 0 Xa 238 a_Ù N-A N CI^T n'N
    362
    Cmp No. Structure 239 N-7 N'N 240 0,χ>ΧίίΓΝ\ 241 HjCO-VA^n V 242 O c,xI^Ç->~yOx n'N 243 P-i ιχΓΛ 244 Λ-<γ n'N
    363
    Cmp No. Structure 245 CT Cl 246 A CT N'N 247 Λ-Βγ χχ ί Me° Ύ 248 jîH XX 5 MeCr^^N-X .0 ‘n Ό 249 rWji χχΤN h'N 250 A-cn μλΧ^ν-^,Ο NV
    364
    Cmp No. Structure 251 Λ-» Λ7O N- '~~' Π 252 x XCN _N-/ NV-7 253 /VCN \ > jCO o 254 ^N P'N jcdV n'N 255 P'N n'N 256 Λ-= χ~χ^νλ Π
    365
    366
    Cmp No. Structure 263 [A-TF 264 AAo n'N 265 AvFx Fl jCO N “Afp- ”*N 266 Fy-?1 Fl jCO N n'N 267 aa^· 268 XXo αΛΛκτ
    367
    Cmp No. Structure 270 ° Ax 271 aAr'x°^ n'N 272 1 //--Ν' JL ' jCQ^ MeO^^N-i /=\ Q-o 273 MeO^X^N-X /=^ 274 “ΛτΧ0' n'N 275 aVao m'N
    368
    Cmp No. Structure 276 °Av 277 1 V___ n*n 278 279 n-r N-wV Cl 280 nA 281 Άνγ n'N 1
    369
    Cmp No. Structure 282 n'N 283 hr'/ y-U n'N 284 285 n'N 286 ολή^ n'N 1 287 Λ-<ο3 jOCj αΛύργ
    370
    Cmp No. Structure 288 clXrVY N'N 1 289 ΠνΤΛ0 MeO-X^N-/ N-V 290 XX MeoX^N-/ Q 291 XX MecX^XX X 292 ax Χχ MeCT^XX ”X 293 xX5 ο,Χχρ.
    371
    Cmp No. Structure 294 ruy jCO n n'N 295 N<7 N ci^SrL° ”'N 296 ÆV n'N 297 N 0^*» n'N 298 a-ù jCCj M-. Z-7 N'N 299 Aj j3 «'''A/ ΑγΛ N'NZ
    372
    Cmp No. Structure 300 JM /θ'! xfT ' 301 (Α/°Ί χχV Ό 302 303 P-N O-'vX C|X^lTNV 7°^CF3 x/-' 304 χχΤ^Ό c,yX^°-cFN'N 305 qrt—
    373
    Cmp No. Structure 306 jîyZ Cl tpr 307 yw XX > F 308 ÎS-CQîEt ciA>yN o^cf3 N N'N 309 Λ-0 N-Y 0 °ArV^ n'N 310 α ξΥΥ 311 Ύ'
    374
    Cmp No. Structure 312 'N 313 cr oArcF· m'N
    or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
  5. 5. A compound selected from:
    Compound Structure 180 ιΛ_Λ? N 181 Α-Λι Me0 'N f/ 182 rf Avq n''N
    375
    Compound Structure 183 I ι A-Λ? MeO'^^V-N 0^ 184 Α_Λϊ Ά^Ά, Λ-Α Q 185 Α-Λ .. ν<7 νΑζ f ΟΑΟ1 186 _^Ν<7 fUA“ 'N 187 (ΑΛΟ fJCÇL°e· η 188 A-/'? <^ζΝ \ 'Ν<γ^ F AAy
    376
    Compound Structure • · . 189 fXXhuf * 190 λ/? |XyN \ NV 191 « Q-cn f 192 Λ-cn Ά 193 r />-cn XX > ?Fa “VrH'ô 194 Æ_?'î iYV Nif ΜθΟ^ΝΧΠ
    377
    Compound Structure 195 rx 5 i 'Y- NV k»/ _xrF 196 r ,>-cn 197 Q-™ 198 -N P N ,ιχΓΛ N'N I 199 rpF ΌΟ 200 ,F kl' “Λα»
    378
    Compound Structure 201 A_/'° |x^zN~\ ''Y' MeCr^V-N CL s Λ-Λ-' t 202 203 jXV-V 204 205 Cl QAJ 206 ^.N__O'N I 'N 207 Γ />-CN XXf O
    379
    Compound Structure 208 A-cn jCO O N.y-' ci 209 Λ-°ν 210 ffcn MeoF^Îp4 211 AÇf /— 212 0- ‘F 213 * ,XQ 1 >—< X F
    380
    Compound Structure 214 Q
    or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof.
  6. 6. A pharmaceutical composition comprising a compound according to any one of claims 1-
    5 5, or a pharmaceutically acceptable sait, hydrate, solvaté, polymorph, isomer, or combination thereof, in a therapeutically effective amount; and an acceptable carrier, adjuvant or vehicle.
  7. 7. The pharmaceutical composition according to claim 6, wherein said composition further comprises a second therapeutic agent.
    10
  8. 8. The pharmaceutical composition according to daim 7, wherein the second therapeutic agent Is selected from an antipsychotic, memantine and an acétylcholine esterase
    I inhibitor (AChE-l), or wherein the second therapeutic agent is an antipsychotic selected
    I - from aripîprazole, olanzapine and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvatés, and polymorphe thereof, or wherein the second therapeutic
    15 agent is memantine, a pharmaceutically acceptable sait, hydrate, solvaté, or polymorph thereof, or wherein the second therapeutic agent is an AChE-l selected from Donepezil, Galantamine, and Rivastigmine, and the pharmaceutically acceptable salts, hydrates, solvatés, and polymorphs thereof.
  9. 9. A compound according to any one of claims 1-5 for use as a médicament.
    20
  10. 10. A compound according to any one of claims 1-5 for use in treating cognitive impairment associated with a central nervous system (CNS) disorder in a subject in need thereof.
    381
  11. 11. The compound for use of claim 10, wherein the CNS disorder Is age-related cognitive impairment.
  12. 12. The compound for use of claim 11, wherein the age-related cognitive impairment is Mild Cognitive Impairment (MCI).
  13. 13. The compound for use of claim 12, wherein the Mild Cognitive impairment Is amnestic Mild Cognitive impairment (AMCI).
  14. 14. The compound for use according to claim 10, wherein the CNS disorder is dementia.
  15. 15. The compound for use of claim 14, wherein the dementia is Alzheimeris disease.
  16. 16. The compound for use of claim 10, wherein the CNS disorder is schizophrénie or bipolar disorder, or wherein the CNS disorder is amyotrophie latéral sclerosis (ALS), or wherein the CNS disorder is post traumatic stress disorder (PTSD), or wherein the CNS disorder is associated with cancer therapy, or wherein the CNS disorder 1$ mental retardation, or
    I wherein the CNS disorder is Parkinson's disease (PD), or wherein the CNS disorder is !
    autism, or wherein the CNS disorder is compulsive behaviour, or wherein the CNS disorder 1$ substance addiction.
  17. 17. Use of a compound according to any one of claims 1-5 or a pharmaceutical composition according to any one of claims 6-8 in the préparation of a médicament for the treatment of cognitive impairment associated with a central nervous system (CNS) disorder ln a subject in need thereof.
  18. 18. The use of claim 17, wherein the CNS disorder is age-related cognitive Impairment.
  19. 19. The use of claim 18, wherein the age-related cognitive Impairment Is Mild Cognitive Impairment (MCI).
  20. 20. The use of claim 19, wherein the Mild Cognitive Impairment is amnestic Mild Cognitive Impairment (AMCI).
  21. 21. The use according to claim 17, wherein the CNS disorder is dementia.
  22. 22. The use of claim 21, wherein the dementia is Alzheimeris disease.
    382
  23. 23. The use of claim 17, wherein the CNS disorder is schizophrenia or bipolar disorder, or
    I wherein the CNS disorder is amyotrophie latéral sclerosis (ALS), or wherein the CNS
    I disorder Is post traumatic stress disorder (PTSD), or wherein the CNS disorder is associated with cancer therapy, or wherein the CNS disorder is mental retardation, or I
    5 wherein the CNS disorder is Parkinson’s disease (PD), or wherein the CNS disorder is autism, or wherein the CNS disorder is compulsive behaviour, or wherein the CNS disorder s substance addiction.
OA1201700496 2015-06-19 2016-06-17 Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment. OA18506A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US62/182,336 2015-06-19

Publications (1)

Publication Number Publication Date
OA18506A true OA18506A (en) 2018-12-04

Family

ID=

Similar Documents

Publication Publication Date Title
AU2014368961B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
AU2017393082B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
AU2021200164B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
IL278053B1 (en) Peptide macrocycles against acinetobacter baumannii
CA3104478A1 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US11505555B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
OA18506A (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment.
OA20303A (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment.
OA19667A (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment.
BR122023024430A2 (en) COMPOUNDS DERIVED FROM BENZODIAZEPINE, PHARMACEUTICAL COMPOSITION COMPRISING THE SAME AND USE THEREOF FOR THE TREATMENT OF COGNITIVE DEFICIT ASSOCIATED WITH A DISORDER OF THE CENTRAL NERVOUS SYSTEM (CNS)
BR112016014564B1 (en) COMPOUNDS DERIVED FROM BENZODIAZEPINE, PHARMACEUTICAL COMPOSITION COMPRISING SAID COMPOUNDS AND USE THEREOF FOR THE TREATMENT OF COGNITIVE DEFICIENCY ASSOCIATED WITH A DISORDER OF THE CENTRAL NERVOUS SYSTEM