Title of Invention

COMPOUNDS FOR THE PREVENTION OF CARDIOVASCULAR DISEASES

Abstract The present disclosure relates to compounds, which are useful for regulating the expression of apolipoproteiπ A-I (ApoA-I), and their use for the treatment and prevention of cardiovascular disease and related disease states, including cholesterol- or lipid-related disorders, such as, for example, atherosclerosis.
Full Text COMPOUNDS FOR THE PREVENTION AND TREATMENT OF
CARDIOVASCULAR DISEASES
Technical Field
[001] The present disclosure relates to compounds, which are useful for
regulating the expression of apolipoprotein A-l (ApoA-l), and their use for the
treatment and prevention of cardiovascular disease and related disease states,
including cholesterol- or lipid-related disorders, such as, for example,
atherosclerosis.
BACKGROUND
' [002] Epidemiologic data demonstrate an inverse relationship between
circulating levels of high density lipoprotein cholesterol (HDL-C) and the incidence
of clinically significant atherosclerosis. Each 1 mg/dL increment in the HDL-C
serum level is associated with a 2-3% decrement in cardiovascular risk; a 1 %
reduction in LDL-C reduces coronary heart disease (CHD) risk by 2% (Gordon et
al. (1997) Am. J. Med. 62, 707-714). Experimental evidence further supports the
protective effect of HDL-C against cardiovascular disease. For example, in
subjects with low HDL-C, administration of gemfibrozil results in a 6% increase in
the HDL-C level and a corresponding 22% reduction of the CHD risk (Rubins et al.
(1999) N. Engl. J. Med. 341, 410-418). Observations in genetic disorders
associated with low HDL-C due to reduced ApoA-l expression, also indicate the
link between elevated risk of CHD and low HDL-C.
[003] HDL-C appears to exert its anti-atherogenic effect by mediating
reverse cholesterol transport (RCT), in which cholesterol is recruited from
peripheral tissues and transported to the liver. In addition, HDL-C also exerts anti-

inflammatory and anti-oxidant effects and promotes fibrinolysis. HDL-C particles
protect against oxidation of LDL, an important initial step in promoting cholesterol
uptake by arterial macrophages. HDL-C exists in two main forms, one containing
both apolipoprotein A-l (ApoA-l) and apolipoprotein A-ll (ApoA-ll), and the other
containing ApoA-l without ApoA-ll (Schultz et al. (1993) Nature 365, 762-764).
The cardioprotective effect of HDL-C is mostly, but not exclusively, attributable to
ApoA-l.
[004] Clinical and experimental data suggest that the production of ApoA-l
is a critical determinant of circulating HDL-C. For example, persons with familial
hyperalphalipoproteinemia (elevated ApoA-l) appear to be protected from
atherosclerosis, while those deficient in ApoA-l (hypoalphalipoproteinemia) show
accelerated cardiovascular disease. In addition, various experimental
manipulations to increase production of ApoA-l are associated with reduced
atherogenicity. For example, human ApoA-l is protective in transgenic animal
models (Shah et al. (1998) Circulation 97, 780-785; Rubin et al. (1991) Nature
353, 265-267), and treatment with ApoA-lMilano prevents atherosclerotic lesions
and leads to regression of atherosclerotic plaques in human patients (Nissen ef a/.
(2003) JAMA 290, 2292-2300). Further lines of research demonstrate that ApoA-l
plays a role in enhancing reverse cholesterol transport, attenuating oxidative
stress, increasing paraoxonase activity, enhancing anticoagulant activity, and
increasing anti-inflammatory activity (Andersson (1997) Curr. Opin. Lipidol. 8, 225-
228). Accordingly, ApoA-l is an attractive target for therapeutic intervention.
[005] Currently available therapeutic agents that increase the plasma
concentration of ApoA-l, for example, recombinant ApoA-l or peptides that mimic
ApoA-l, have potential drawbacks with respect to, e.g., stability during storage,

delivery of active product, and in vivo half-life. Thus, small molecule compounds
that up-regulate the production of endogenous ApoA-l, such as, for example, up-
regulators of ApoA-l expression, would be very attractive as new therapeutic
agents for cardiovascular disease. Such small molecule compounds have been
described in WO 2006/045096.
[006] The compounds of the present invention represent a major
improvement over compounds disclosed in WO 2006/045096. Specifically, the
compounds of the present invention are more than an order of magnitude more
potent than the most active compounds described in that publication, such as 2-
(4-hydroxy-phenyl)-pyrano[2,3-b]pyridin-4-one.

2-(4-hydroxy-phenyl)-pyrano[2,3-b]pyridin-4-one
SUMMARY
[007] The present invention includes non-naturally occurring compounds
that are useful for regulating the expression of apolipoprotein A-l (ApoA-l), and
their use in the treatment and prevention of cardiovascular disease and related
disease states, including cholesterol- and lipid-related disorders, such as, for
example, atherosclerosis.
[008] The methods of invention include administering to a mammal (e.g.,
a human) in need thereof a therapeutically effective amount of a compound of
Formula II:


Formula II
wherein:
X is selected from CR11, N and NR11;
Y is selected from CO, CS, and SO2;
R11 is selected from hydrogen, unsubstituted alkyl (preferably C1-3alkyl),
unsubstituted alkenyl (preferably C1-3 alkenyl), and unsubstituted alkynyl
(preferably C1-3 alkynyl);
R1 and R3 are each independently selected from alkoxy (preferably
methoxy), alkyl, amino, halogen (preferably chloride), and hydrogen;
R2 is selected from alkoxy, alkyl, alkenyl, amide, amino, halogen
(preferably bromide or chloride), and hydrogen;
R6 and R8 are each independently selected from alkoxy, alkyl (preferably
methyl), amino, halogen (preferably chloride and fluoride), and hydrogen;
R5 and R9 are each independently selected from halogen (preferably
chloride), and hydrogen;
R7 is selected from alkoxy, alkyl, alkenyl, amide, amino, ether, hydrogen,
and hydroxyl;
R10 is selected from hydrogen and alkyl (preferably methyl); or
two adjacent substituents selected from R1, R2, R3, R6, R7, R8, R10, and R11
are connected to form a group selected from aryl, heteroaryl, cycloalkyl, and
heterocyclyl;

each W is independently selected from C and N, wherein if W is N, then p
is0or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
Z1, Z2 and Z3 are each independently selected from a single bond and a
double bond, wherein at least one of Z1 or Z2 is a double bond;
and pharmaceutically acceptable salts and hydrates thereof.
[009] The invention further includes certain compounds falling within the
scope of Formula II and methods of administering a therapeutically effective
amount of those compounds to a mammal (e.g., a human) in need thereof
wherein:
X is selected from N and CH;
Y is CO;
R1 and R3are each independently selected from alkoxy and hydrogen;
R2 is selected from alkoxy, alkyl, and hydrogen;
R6 and R8 are each independently selected from alkyl, alkoxy, chloride, and
hydrogen;
R5 and R9 are each hydrogen;
R7 is selected from amino, hydroxyl, alkoxy (preferably a substituted ethoxy
group), and alkyl substituted with a heterocyclyl;
R10 is hydrogen; or
two adjacent substituents selected from R6, R7, and R8 are connected to
form a heterocyclyl;
each W is independently selected from C and N, wherein if W is N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R10)P, W is N and p is 1;

for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
Z1 is a double bond, and Z2and Z3 are each a single bond;
with the proviso that if R2 is selected from alkoxy and hydrogen, then at
least one of R1 and R3 is alkoxy;
with the proviso that if R7 is selected from hydroxyl and alkoxy, then at least
one of R6and R8 are independently selected from alkyl, alkoxy, and chloride;
with the proviso that if R7 is an amino, then X is N;
with the proviso that if for W-(R7)P, W is N and p is 0, then at least one of R6
and R8 is chloride;
and pharmaceutically acceptable salts and hydrates thereof.
[010] In some embodiments of the invention, R7 is an amino or an alkoxy
selected from the group represented by Formula III:

wherein:
A is selected from O and N;
n is selected from 0,1, 2, 3, 4 and 5;
B is selected from -C(O)N(Rh)2-, -S(O)2N(Rh)2-, -C(O)-, -S(O)2-, -
C(O)O-, wherein each Rh is selected from alkyl, alkenyl, alkynyl, aryl, arylalkyl,
cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, and hydrogen; and
R20 is selected from (C1-C6) alkyl, (C1-C6) alkenyl, (C1-C6) alkynyl, aryl,
arylalkyl, cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, and hydrogen.

In another embodiment, if A is O and B is -C(O)NH-, then R20 is not an
unsaturated cycloalkyl.
[011] In certain embodiments, the methods, compounds, and
compositions of the invention are useful for the prevention or treatment of
diseases that benefit from raised ApoA-l or HDL, and diseases characterized by
reduced ApoA-l and/or HDL-C, abnormal lipid parameters, or lipid parameters
indicative of high cholesterol. The methods, compounds, and compositions of the
invention can be used to increase expression of ApoA-l. Increasing expression of
ApoA-l may refer to, but is not limited to, transcriptionally modulating the
expression of the ApoA-l gene, thereby affecting the level of the ApoA-l protein

produced (synthesized and secreted). An increase in ApoA-l levels may lead to
an increase the levels of HDL-C and/or increase in the functionality of HDL-C
particles. Thus, the methods, compounds, and compounds of the invention may
further be used to reduce cholesterol levels. Accordingly, the methods,
compounds, and compositions of the invention can be used for treatment and
prevention of cardiovascular disease and related disease states, particularly,
cholesterol- or lipid-related disorders, such as, for example, atherosclerosis.
BRIEF DESCRIPTION OF THE FIGURES
[012] Figure 1 depicts plasma levels of ApoA-l in hApoA-l transgenic
mice receiving 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (Example 7) (10, 30, and 60 mg/kg body weight)
twice daily for 7 days by oral gavage.
[013] Figure 2 depicts plasma levels of HDL cholesterol in hApoA-l
transgenic mice receiving 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-

dimethoxyquinazolin-4(3H)-one (Example 7) (10 and 30 mg/kg body weight) twice
daily for 7 days by oral gavage.
[014] Figure 3 depicts plasma levels of ApoA-l in wild-type C57BL/6 mice
receiving 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one (Example 7) (10, 30, and 60 mg/kg of body weight) twice daily for
3 days by intraperitoneal administration.
[015] Figure 4 depicts plasma levels of HDL cholesterol in wild-type
C57/BI mice receiving 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (Example 7) (10, 30, and 60 mg/kg of body
weight) twice daily for 3 days by oral gavage.
[016] Figure 5 depicts plasma levels of ApoA-l and tissue levels of ApoA-l
mRNA in hApoA-l transgenic mice administered 2-(4-(2-hydroxyethoxy)-3,5-
dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one (Example 7) (30 mg/kg body
weight) twice daily for 7 days by oral gavage.
DETAILED DESCRIPTION
Definitions
[017] The term "aldehyde" or "formyl" as used herein refers to -CHO.
[018] The term "alkenyl" as used herein refers to an unsaturated straight
or branched hydrocarbon having at least one carbon-carbon double bond, such as
a straight or branched group of 2-22, 2-8, or 2-6 carbon atoms, referred to herein
as (C2-C22)alkenyl, (C2-C8)alkenyl, and (C2-C6)alkenyl, respectively. Exemplary
alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl,
hexenyl, butadienyl, pentadienyl, hexadienyl, 2-ethylhexenyl, 2-propyl-2-butenyl,
4-(2-methyl-3-butene)-pentenyl, etc.

[019] The term "alkoxy" as used herein refers to an alkyl group attached to
an oxygen (-O-alkyl-). "Alkoxy" groups also include an alkenyl group attached to
an oxygen ("alkenyloxy") or an alkynyl group attached to an oxygen ("alkynyloxy")
groups. Exemplary alkoxy groups include, but are not limited to, groups with an
alkyl, alkenyl or alkynyl group of 1-22, 1-8, or 1-6 carbon atoms, referred to herein
as (C1-C22)alkoxy, (C1-C8)alkoxy, and (C1-C6)alkoxy, respectively. Exemplary
alkoxy groups include, but are not limited to methoxy, ethoxy, etc.
[020] The term "alkyl" as used herein refers to a saturated straight or
branched hydrocarbon, such as a straight or branched group of 1-22, 1-8, or 1-6
carbon atoms, referred to herein as (C1-C22)alkyl, (C1-C8)alkyl, and (C1-C6)alkyl,
respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl,
propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1 -butyl, 3-methyl-
1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methy-1-pentyl, 3-methyl-1-
pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl,
2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl,
pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc.
[021] The term "alkynyl" as used herein refers to an unsaturated straight
or branched hydrocarbon having at least one carbon-carbon triple bond, such as
a straight or branched group of 2-22, 2-8, or 2-6 carbon atoms, referred to herein
as (C2-C22)alkynyl, (C2-C8)alkynyl, and (C2-C6)alkynyl, respectively. Exemplary
alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl,
hexynyl, methylpropynyl, 4-methyl-1-butynyl, 4-propyl-2-pentynyl, and 4-butyl-2-
hexynyl, etc.
[022] The term "amide" as used herein refers to the form -NRaC(O)(Rb)-
or-C(O)NRbRc, wherein Ra, Rb and RC are each independently selected from

alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, haloalkyl, heteroaryl, heterocyclyl,
hydrogen. The amide can be attached to another group through the carbon, the
nitrogen, Rb, or Rc. The amide also may be cyclic, for example Rb and Rc, may
be joined to form a 3- to 12-membered ring, such as a 3- to 10-membered ring or
a 5- to 6-membered ring. The term "amide" encompasses groups such as
sulfonamide, urea, ureido, carbamate, carbamic acid, and cyclic versions thereof.
The term "amide" also encompasses an amide group attached to a carboxy group,
e.g., -amide-COOH or salts such as -amide-COONa, etc., an amino group
attached to a carboxy group, e.g., -amino-COOH or salts such as -amino-
COONa, etc.
4
[023] The term "amine" or "amino" as used herein refers to the form
-NRdRe or -N(Rd)Re_, where Rd and Re are independently selected from alkyl,
alkenyl, alkynyl, aryl, arylalkyl, carbamate, cycloalkyl, haloalkyl, heteroaryl,
heterocyclyl, hydrogen. The amino can be attached to the parent molecular group
through the nitrogen. The amino also may be cyclic, for example any two of Rd
and Re may be joined together or with the N to form a 3- to 12-membered ring,
e.g., morpholino or piperidinyl. The term amino also includes the corresponding
quaternary ammonium salt of any amino group. Exemplary amino groups include
alkylamino groups, wherein at least one of Rd or Re is an alkyl group.
[024] The term "aryl" as used herein refers to a mono-, bi-, or other multi-
carbocyclic, aromatic ring system. The aryl group can optionally be fused to one
or more rings selected from aryls, cycloalkyls, and heterocyclyls. The aryl groups
of this invention can be substituted with groups selected from alkoxy, aryloxy,
alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano,
cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl,

hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid,
sulfonamide and thioketone. Exemplary aryl groups include, but are not limited to,
phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, and naphthyl, as well as
benzo-fused carbocyclic moieties such as 5,6,7,8-tetrahydronaphthyl. Exemplary
aryl groups also include, but are not limited to a monocyclic aromatic ring system,
wherein the ring comprises 6 carbon atoms, referred to herein as "(C6)aryl."
[025] The term "arylalkyl" as used herein refers to an alkyl group having at
least one aryl substituent, e.g. -aryl-alkyl-. Exemplary arylalkyl groups include, but
are not limited to, arylalkyls having a monocyclic aromatic ring system, wherein
the ring comprises 6 carbon atoms, referred to herein as "(C6)arylalkyl."
[026] The term "aryloxy" as used herein refers to an aryl group attached to
an oxygen atom. Exemplary aryloxy groups include, but are not limited to,
aryloxys having a monocyclic aromatic ring system, wherein the ring comprises 6
carbon atoms, referred to herein as "(C6)aryloxy."
[027] The term "arylthio" as used herein refers to an aryl group attached to
an sulfur atom. Exemplary arylthio groups include, but are not limited to, arylthios
having a monocyclic aromatic ring system, wherein the ring comprises 6 carbon
atoms, referred to herein as "(C6)arylthio."
[028] The term "arylsulfonyl" as used herein refers to an aryl group
attached to a sulfonyl group, e.g., -S(O)2-aryl-. Exemplary arylsulfonyl groups
include, but are not limited to, arylsulfonyls having a monocyclic aromatic ring
system, wherein the ring comprises 6 carbon atoms, referred to herein as
"(C6)arylsulfonyl."
[029] The term "benzyl" as used herein refers to the group -CHh-phenyl.

[030] The term "bicyclic aryl" as used herein refers to an aryl group fused
to another aromatic or non-aromatic carbocylic or heterocyclic ring. Exemplary
bicyclic aryl groups include, but are not limited to, naphthyl or partly reduced forms
thereof, such as di-, tetra-, or hexahydronaphthyl.
[031] The term "bicyclic heteroaryl" as used herein refers to a heteroaryl
group fused to another aromatic or non-aromatic carbocylic or heterocyclic ring.
Exemplary bicyclic heteroaryls include, but are not limited to, 5,6 or 6,6-fused
systems wherein one or both rings contain heteroatoms. The term "bicyclic
heteroaryl" also encompasses reduced or partly reduced forms of fused aromatic
system wherein one or both rings contain ring heteroatoms. The ring system may
contain up to three heteroatoms, independently selected from oxygen, nitrogen, or
sulfur. The bicyclic system may be optionally substituted with one or more groups
selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl,
carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl,
heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl,
sulfonyl, sulfonic acid, sulfonamide and thioketone. Exemplary bicyclic
heteroaryl's include, but are not limited to, quinazolinyl, benzothiophenyl,
benzoxazolyl, benzimidazolyl, benzothiazolyl, benzofuranyl, indolyl, quinolinyl,
isoquinolinyl, phthalazinyl, benzotriazolyl, benzopyridinyl, and benzofuranyl.
[032] The term "carbamate" as used herein refers to the form
-RgOC(O)N(Rh)-, -RgOC(O)N(Rh)Ri-, or -OC(O)NRhRi, wherein Rg, Rh and Ri
are each independently selected from alkyl, alkenyl, alkynyl, aryl, arylalkyl,
cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, hydrogen. Exemplary carbamates
include, but are not limited to, arylcarbamates or heteroaryl carbamates, e.g.,

wherein at least one of Rg, Rh and Ri are independently selected from aryl or
heteroaryl, such as pyridine, pyridazine, pyrimidine, and pyrazine.
[033] The term "carbonyl" as used herein refers to -C(O)-.
[034] The term "carboxy" as used herein refers to -COOH or its
corresponding carboxylate salts, e.g. -COONa, etc. The term carboxy also
includes "carboxycarbonyl," e.g. a carboxy group attached to a carbonyl group,
e.g., -C(O)-COOH or salts such as -C(O)-COONa, etc..
[035] The term "cyano" as used herein refers to -CN.
[036] The term "cycloalkoxy" as used herein refers to a cycloalkyl group
attached to an oxygen.
[037] The term "cycloalkyl" as used herein refers to a saturated or
unsaturated cyclic, bicyclic, or bridged bicyclic hydrocarbon group of 3-12
carbons, or 3-8 carbons, referred to herein as "(C3-C8)cycloalkyl," derived from a
cycloalkane. Exemplary cycloalkyl groups include, but are not limited to,
cyclohexanes, cyclohexenes, cyclopentanes, and cyclopentenes. Cycloalkyl
groups may be substituted with alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide,
amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl,
halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate,
sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone. Cycloalkyl
groups can be fused to other cycloalkyl saturated or unsaturated, aryl, or
heterocyclyl groups.
[038] The term "dicarboxylic acid" as used herein refers to a group
containing at least two carboxylic acid groups such as saturated and unsaturated
hydrocarbon dicarboxylic acids and salts thereof. Exemplary dicarboxylic acids
include alkyl dicarboxylic acids. Dicarboxylic acids may be substituted with

alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate,
carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl,
heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl,
sulfonyl, sulfonic acid, sulfonamide and thioketone. Dicarboxylic acids include,
but are not limited to succinic acid, glutaric acid, adipic acid, suberic acid, sebacic
acid, azelaic acid, maleic acid, phthalic acid, aspartic acid, glutamic acid, malonic
acid, fumaric acid, (+)/(-)-malic acid, (+)/(-) tartaric acid, isophthalic acid, and
terephthalic acid. Dicarboxylic acids further include carboxylic acid derivatives
thereof, such as anhydrides, imides, hydrazides, etc., for example, succinic
anhydride, succinimide, etc.
' [039] The term "ester refers to the structure -C(O)O-, -C(O)O-Rj_,
-RkC(O)O-Rj_, or -RKC(O)O-, where O is not bound to hydrogen, and Rj and Rk
can independently be selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide,
amino, aryl, arylalkyl, cycloalkyl, ether, haloalkyl, heteroaryl, heterocyclyl. R| be a hydrogen, but Rj cannot be hydrogen. The ester may be cyclic, for example
the carbon atom and Rj, the oxygen atom and Rk, or Rj and Rk may be joined to
form a 3- to 12-membered ring. Exemplary esters include, but are not limited to,
alkyl esters wherein at least one of Rj or Rk is alkyl, such as -O-C(O)-alkyl,
-C(O)-O-alkyl-, -alkyl-C(O)-O-alkyl-, etc. Exemplary esters also include aryl or
heteoraryl esters, e.g. wherein at least one of Rj or Rk is a heteroaryl group such
as pyridine, pyridazine, pyrmidine and pyrazine, such as a nicotinate ester.
Exemplary esters also include reverse esters having the structure -RkC(O)O-,
where the oxygen is bound to the parent molecular. Exemplary reverse esters
include succinate, D-argininate, L-argininate, L-lysinate and D-lysinate. Esters
also include carboxylic acid anhydrides and acid halides.

[040] The term "ether" refers to the structure -RjO-Rm_, where Rj and Rm
can independently be alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heterocyclyl, or ether.
The ether can be attached to the parent molecular group through R| or Rm.
Exemplary ethers include, but are not limited to, alkoxyalkyl and alkoxyaryl
groups. Ethers also includes polyethers, e.g., where one or both of R| and Rm are
ethers.
[041] The terms "halo" or "halogen" or "Hal" as used herein refer to F, CI,
Br, or I.
[042] The term "haloalkyl" as used herein refers to an alkyl group
substituted with one or more halogen atoms. "Haloalkyls" also encompass alkenyl
or alkynyl groups substituted with one or more halogen atoms.
[043] The term "heteroaryl" as used herein refers to a mono-, bi-, or multi-
cyclic, aromatic ring system containing one or more heteroatoms, for example 1 to
3 heteroatoms, such as nitrogen, oxygen, and sulfur. Heteroaryls can be
substituted with one or more substituents including alkoxy, aryloxy, alkyl, alkenyl,
alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester,
ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro,
phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
Heteroaryls can also be fused to non-aromatic rings. Illustrative examples of
heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrimidyl,
pyrazyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3)-and (1,2,4)-triazolyl,
pyrazinyl, pyrimidilyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, furyl, phenyl,
isoxazolyl, and oxazolyl. Exemplary heteroaryl groups include, but are not limited
to, a monocyclic aromatic ring, wherein the ring comprises 2 to 5 carbon atoms
and 1 to 3 heteroatoms, referred to herein as "(C2-C5)heteroaryl."

[044] The terms "heterocycle," "heterocyclyl," or "heterocyclic" as used
herein refer to a saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered ring
containing one, two, or three heteroatoms independently selected from nitrogen,
oxygen, and sulfur. Heterocycles can be aromatic (heteroaryls) or non-aromatic.
Heterocycles can be substituted with one or more substituents including alkoxy,
aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy,
cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl,
hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid,
sulfonamide and thioketone. Heterocycles also include bicyclic, tricyclic, and
tetracyclic groups in which any of the above heterocyclic rings is fused to one or
two rings independently selected from aryls, cycloalkyls, and heterocycles.
Exemplary heterocycles include acridinyl, benzimidazolyl, benzofuryl,
benzothiazolyl, benzothienyl, benzoxazolyl, biotinyl, cinnolinyl, dihydrofuryl,
dihydroindolyl, dihydropyranyl, dihydrothienyl, dithiazolyl, furyl, homopiperidinyl,
imidazolidinyl, imidazolinyl, imidazolyl, indolyl, isoquinolyl, isothiazolidinyl,
isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl,
oxazolyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrazinyl, pyrazolyl,
pyrazolinyl, pyridazinyl, pyridyl, pyrimidinyl, pyrimidyl, pyrrolidinyl, pyrrolidin-2-
onyl, pyrrolinyl, pyrrolyl, quinolinyl, quinoxaloyl, tetrahydrofuryl,
tetrahydroisoquinolyl, tetrahydropyranyl, tetrahydroquinolyl, tetrazolyl, thiadiazolyl,
thiazolidinyl, thiazolyl, thienyl, thiomorpholinyl, thiopyranyl, and triazolyl.
[045] The terms "hydroxy" and "hydroxyl" as used herein refers to -OH.
[046] The term "hydroxyalkyl" as used herein refers to a hydroxy attached
to an alkyl group.

[047] The term "hydroxyaryl" as used herein refers to a hydroxy attached
to an aryl group.
[048] The term "ketone" as used herein refers to the structure -C(O)-Rn
(such as acetyl, -C(O)CH3) or -Rn_C(O)-Ro_. The ketone can be attached to
another group through Rn or Ro. Rn or Ro can be alkyl, alkenyl, alkynyl,
cycloalkyl, heterocyclyl or aryl, or Rn or Ro can be joined to form a 3- to 12-
membered ring.
[049] The term "monoester" as used herein refers to an analogue of a
dicarboxylic acid wherein one of the carboxylic acids is functionalized as an ester
and the other carboxylic acid is a free carboxylic acid or salt of a carboxylic acid.
4
Examples of monoesters include, but are not limited to, to monoesters of succinic
acid, glutaric acid, adipic acid, suberic acid, sebacic acid, azelaic acid, oxalic and
maleic acid.
[050] The term "nitro" as used herein refers to the -NO2.
[051] The term "perfluoroalkoxy" as used herein refers to an alkoxy group
in which all of the hydrogen atoms have been replaced by fluorine atoms.
[052] The term "perfluoroalkyl" as used herein refers to an alkyl group in
which all of the hydrogen atoms have been replaced by fluorine atoms.
Exemplary perfluroalkyl groups include, but are not limited to, C1-5 perfluoroalkyl,
such as trifluoromethyl, etc.
[053] The term "perfluorocycloalkyl" as used herein refers to a cycloalkyl
group in which all of the hydrogen atoms have been replaced by fluorine atoms.
[054] The term "phenyl" as used herein refers to a 6-membered
carbocyclic aromatic ring. The phenyl group can also be fused to a cyclohexane
or cyclopentane ring. Phenyl can be substituted with one or more substituents

including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl,
carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl,
heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl,
sulfonyl, sulfonic acid, sulfonamide and thioketone.
[055] The term "phosphate" as used herein refers to the structure
-OP(O)O2-, -RxOP(O)O2-, -OP(O)O2Ry-, or-RxOP(O)O2Ry-, wherein Rxand Ry
can be alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heterocyclyl, hydrogen
[056] The term "sulfide" as used herein refers to the structure -RZS-,
where Rz can be alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, haloalkyl,
heteroaryl, heterocyclyl. The sulfide may be cyclic, forming a 3 to 12-membered
4
ring. The term "alkylsulfide" as used herein refers to an alkyl group attached to a
sulfur atom.
[057] The term "sulfinyl" as used herein refers to the structure -S(O)O-,
-RpS(O)O-, -RpS(O)ORq-, or -S(O)ORq-, wherein RP and Rq can be alkyl, alkenyl,
aryl, arylalkyl, cycloalkyl, , haloalkyl, heteroaryl, heterocyclyl, hydroxyl, Exemplary
sulfinyl groups include, but are not limited to, alkylsulfinyls wherein at least one of
Rp or Rq is alkyl, alkenyl or alkynyl.
[058] The term "sulfonamide" as used herein refers to the structure -(Rr)-
N-S(O)2-Rs- or-Rt(Rr)-N-S(O)2-Rs, where Rt, Rr, and Rs can be, for example,
hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, and heterocyclyl. Exemplary
sulfonamides include alkylsulfonamides (e.g., where Rs is alkyl), arylsulfonamides
(e.g., where Rsis aryl), cycloalkyl sulfonamides (e.g., where Rs is cycloalkyl), and
heterocyclyl sulfonamides (e.g., where Rs is heterocyclyl), etc.

[059] The term "sulfonate" as used herein refers to -OSO3-. Sulfonate
includes salts such as -OSO3Na, -OSO3K, etc. and the acid -OSO3 H

[060] The term "sulfonic acid" refers to -SO3H- and its corresponding salts,
e.g. -SO3K-, -SO3Na-.
[061] The term "sulfonyl" as used herein refers to the structure RuSO2-,
where Ru can be alkyl, alkenyl, alkynyl, aryl, cycloalkyl, and heterocyclyl, e.g.,
alkylsulfonyl. The term "alkylsulfonyl" as used herein refers to an alkyl group
attached to a sulfonyl group. "Alkylsulfonyl" groups can optionally contain alkenyl
or alkynyl groups.
[062] The term "thioketone" refers to the structure -RV_C(S)-RW_. The
ketone can be attached to another group through Rv or Rw. Rv or Rw can be
alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl or aryl, or Rv or Rw can be joined to
form a 3- to 12-membered ring.
[063] "Alkyl," "alkenyl," "alkynyl", "alkoxy", "amino" and "amide" groups can
be substituted with or interrupted by or branched with at least one group selected
from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl,
carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl,
heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl,
sulfonyl, sulfonic acid, sulfonamide, thioketone, ureido and N. The substituents
may be branched to form a substituted or unsubstituted heterocycle or cycloalkyl.
[064] As used herein, a "suitable substituent" refers to a group that does
not nullify the synthetic or pharmaceutical utility of the compounds of the invention
or the intermediates useful for preparing them. Examples of suitable substituents
include, but are not limited to: C1-22, C1-8, and C1-6 alkyl, alkenyl or alkynyl; C1-6
aryl, C2-5 heteroaryl; C3-7 cycloalkyl; C1-22, C1-8, and C1-6 alkoxy; C6 aryloxy; -CN;

-OH; oxo; halo, carboxy; amino, such as -NH(C1-22 C1-8, or C1-6 alkyl), -N(C1-22, C1-
8, and C1-6 alkyl)2, -NH((C6)aryl), or -N((C6)aryl)2; formyl; ketones, such as -CO(C1-
22, C1-8, and C1-6 alkyl), -CO((C6.aryl) esters, such as -CO2(C1-22, C1-8, and C1-6
alkyl) and -CO2 (C6 aryl). One of skill in art can readily choose a suitable
substituent based on the stability and pharmacological and synthetic activity of the
compound of the invention.
[065] The term "pharmaceutically acceptable carrier" as used herein refers
to any and all solvents, dispersion media, coatings, isotonic and absorption
delaying agents, and the like, that are compatible with pharmaceutical
administration. The use of such media and agents for pharmaceutically active
substances is well known in the art. The compositions may also contain other
active compounds providing supplemental, additional, or enhanced therapeutic
functions.
[066] The term "pharmaceutically acceptable composition" as used herein
refers to a composition comprising at least one compound as disclosed herein
formulated together with one or more pharmaceutically acceptable carriers.
[067] The term "pharmaceutically acceptable prodrugs" as used herein
represents those prodrugs of the compounds of the present invention that are,
within the scope of sound medical judgment, suitable for use in contact with the
tissues of humans and lower animals without undue toxicity, irritation, allergic
response, commensurate with a reasonable benefit / risk ratio, and effective for
their intended use, as well as the zwitterionic forms, where possible, of the
compounds of the invention. A discussion is provided in Higuchi et al., "Pro-drugs
as Novel Delivery Systems," ACS Symposium Series, Vol. 14, and in Roche, E.B.,

ed. Bioreversible Carriers in Drug Design, American Pharmaceutical Association
and Pergamon Press, 1987, both of which are incorporated herein by reference.
[068] The term "pharmaceutically acceptable salt(s)" refers to salts of
acidic or basic groups that may be present in compounds used in the present
compositions. Compounds included in the present compositions that are basic in
nature are capable of forming a wide variety of salts with various inorganic and
organic acids. The acids that may be used to prepare pharmaceutically
acceptable acid addition salts of such basic compounds are those that form non-
toxic acid addition salts, i.e., salts containing pharmacologically acceptable
anions, including but not limited to sulfate, citrate, matate, acetate, oxalate,
chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate,
isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate,
pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate,
gluconate, glucaronate, saccharate, formate, benzoate, glutamate,
methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and
pamoate (i.e., 1,1'-methylene-bis-(2-hydroxy-3-naphthoate)) salts. Compounds
included in the present compositions that include an amino moiety may form
pharmaceutically acceptable salts with various amino acids, in addition to the
acids mentioned above. Compounds included in the present compositions, that
are acidic in nature are capable of forming base salts with various
pharmacologically acceptable cations. Examples of such salts include alkali metal
or alkaline earth metal salts and, particularly, calcium, magnesium, sodium,
lithium, zinc, potassium, and iron salts.
[069] The compounds of the disclosure may contain one or more chiral
centers and/or double bonds and, therefore, exist as stereoisomers, such as

geometric isomers, enantiomers or diastereomers. The term "stereoisomers"
when used herein consist of all geometric isomers, enantiomers or diastereomers.
These compounds may be designated by the symbols "R" or "S," depending on
the configuration of substituents around the stereogenic carbon atom. The
present invention encompasses various stereoisomers of these compounds and
mixtures thereof. Stereoisomers include enantiomers and diastereomers.
Mixtures of enantiomers or diastereomers may be designated "(±)" in
nomenclature, but the skilled artisan will recognize that a structure may denote a
chiral center implicitly.
[070] Individual stereoisomers of compounds of the present invention can
be prepared synthetically from commercially available starting materials that
contain asymmetric or stereogenic centers, or by preparation of racemic mixtures
followed by resolution methods well known to those of ordinary skill in the art.
These methods of resolution are exemplified by (1) attachment of a mixture of
enantiomers to a chiral auxiliary, separation of the resulting mixture of
diastereomers by recrystallization or chromatography and liberation of the
optically pure product from the auxiliary, (2) salt formation employing an optically
active resolving agent, or (3) direct separation of the mixture of optical
enantiomers on chiral chromatographic columns. Stereoisomeric mixtures can
also be resolved into their component stereoisomers by well known methods,
such as chiral-phase gas chromatography, chiral-phase high performance liquid
chromatography, crystallizing the compound as a chiral salt complex, or
crystallizing the compound in a chiral solvent. Stereoisomers can also be
obtained from stereomerically-pure intermediates, reagents, and catalysts by well
known asymmetric synthetic methods.

[071] Geometric isomers can also exist in the compounds of the present
invention. The present invention encompasses the various geometric isomers
and mixtures thereof resulting from the arrangement of substituents around a
carbon-carbon double bond or arrangement of substituents around a carbocyclic
ring. Substituents around a carbon-carbon double bond are designated as being
in the "Z' or "P' configuration wherein the terms "Z" and "E" are used in
accordance with IUPAC standards. Unless otherwise specified, structures
depicting double bonds encompass both the E and Z isomers.
[072] Substituents around a carbon-carbon double bond alternatively can
be referred to as "cis" or "trans," where "cis" represents substituents on the same
side df the double bond and "trans" represents substituents on opposite sides of
the double bond. The arrangements of substituents around a carbocyclic ring are
designated as "cis" or "trans." The term "cis" represents substituents on the same
side of the plane of the ring and the term "trans" represents substituents on
opposite sides of the plane of the ring. Mixtures of compounds wherein the
substituents are disposed on both the same and opposite sides of plane of the
ring are designated "cis/trans."
Embodiments of the Invention
[073] Disclosed herein are methods for increasing expression of ApoA-l in
a mammal (e.g., a human) comprising administering a therapeutically effective
amount of a compound of Formula II:


wherein:
X is selected from CR11, N and NR11,
Y is selected from CO, CS, and SO2,
R11 is selected from hydrogen, unsubstituted alkyl (preferably C1-3lkyl),
unsubstituted alkenyl (preferably C1-3 alkenyl), and unsubstituted alkynyl
(preferably C1-3 alkynyl);
R1 and R3 are each independently selected from alkoxy (preferably
methoxy), alkyl, amino, halogen (preferably chloride), and hydrogen;
R2 is selected from alkoxy, alkyl, alkenyl, amide, amino, halogen
(preferably bromide or chloride), and hydrogen;
R6 and R8 are each independently selected from alkoxy, alkyl (preferably
methyl), amino, halogen (preferably chloride and fluoride), and hydrogen;
R5 and R9 are each independently selected from halogen (preferably
chloride), and hydrogen;
R7 is selected from alkoxy, alkyl, alkenyl, amide, amino, ether, hydrogen,
and hydroxyl;
R10 is selected from hydrogen and alkyl (preferably methyl); or
two adjacent substituents selected from R1, R2, R3, R6, R7, R8, R10, and R11
are connected to form a group selected from aryl, heteroaryl, cycloalkyl, and
heterocyclyl;

each W is independently selected from C and N, wherein if W is N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
Z1, Z2 and Z3 are each independently selected from a single bond and a
double bond, wherein at least one of Z1 or Z2 is a double bond;
and pharmaceutically acceptable salts and hydrates thereof.
[074] Another embodiment includes methods for increasing expression of
ApoA-l in a mammal (e.g., a human) comprising administering a therapeutically
effective amount of a compound of Formula II:

wherein:
X is selected from N and CH;
R1 and R3are each independently selected from alkoxy and hydrogen;
R2 is selected from alkoxy, alkyl, and hydrogen;
R6 and R8 are each independently selected from alkyl, alkoxy, chloride, and
hydrogen;
R5 is hydrogen;
R7 is selected from amino, hydroxyl, alkoxy (preferably a substituted ethoxy
group), and alkyl substituted with a heterocyclyl, or

two adjacent substituents selected from R6, R7, and R8 are connected to
form a heterocyclyl;
each W is independently selected from C and N, wherein if W is N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
with the proviso that if R2 is selected from alkoxy or hydrogen, then at least
one of R1 and R3 is alkoxy;
with the proviso that if R7 is selected from hydroxyl or alkoxy, then at least
one of R6and R8are independently selected from alkyl, alkoxy, and chloride;
with the proviso that if R7 is an amino, then X is N;
with the proviso that if for W-(R7)P, W is N and p is 0, then at least one of R6
and R8 is chloride;
and pharmaceutically acceptable salts and hydrates thereof.
[075] The following is a list of specific exemplary embodiments that are
encompassed by the invention:
1. A method for increasing expression of ApoA-l in a mammal (e.g. a
human) comprising administering a therapeutically effective amount of a
compound of Formula II:

wherein:

X is selected from N and CH;
R1 and R3are each independently selected from alkoxy and hydrogen;
R2 is selected from alkoxy, alkyl, and hydrogen;
R6 and R8 are each independently selected from alkyl, alkoxy, chloride, and
hydrogen;
R5 is hydrogen;
R7 is selected from amino, hydroxyl, alkoxy (preferably a substituted ethoxy
group), and alkyl substituted with a heterocyclyl, or
two adjacent substituents selected from R6, R7, and R8 are connected to
form a heterocyclyl;
each W is independently selected from C and N, wherein if W is N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
with the proviso that if R2 is selected from alkoxy or hydrogen, then at least
one of R1 and R3 is alkoxy;
with the proviso that if R7 is selected from hydroxyl or alkoxy, then at least
one of R6 and R8 are independently selected from alkyl, alkoxy, and chloride;
with the proviso that if R7 is an amino, then X is N;
with the proviso that if for W-(R7)P W is N and p is 0, then at least one of R6
and R8 is chloride;
and pharmaceutically acceptable salts and hydrates thereof.
2. The method according to embodiment 1, wherein at least one of R6 and
R8 is selected from alkyl, alkoxy, and chloride.
3. The method according to embodiment 1, wherein R6 and R8 are each
hydrogen, and W-(R7)P is C-(R7)1.

4. The method according to embodiment 1, wherein both R6 and R8 are
not hydrogen.
5. The method according to embodiment 1, wherein X is CH;
R1 and R3 are alkoxy;
R6 and R8 are alkyl; and
R7 is hydroxyI.
6. The method according to embodiment 1, wherein X is N;
R1 and R3 are alkoxy;
R6 and R8 are alkyl; and
R7 is alkoxy substituted with a hydroxyl.
7. The method according to embodiment 1, wherein R7 is not
diethylamino or an alkoxy substituted with a carboxylate group.
8. The method according to embodiment 1, wherein R7 is selected from
hydroxyl, amino, and alkoxy.
9. The method according to embodiment 1, wherein the compound of
Formula II is 3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-
one (Example 1).
10. The method according to embodiment 1, wherein the compound of
Formula II is 7-(4-hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-
5(6H)-one (Example 6).
11. The method according to embodiment 1, wherein the compound of
Formula II is 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one (Example 7).
12. The method according to embodiment 1, wherein the compound of
Formula II is selected from:

3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one
(Example 1);
3-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-
1(2H)-one (Example 2);
3-(4-hydroxy-3,5-dimethylphenyl)-7-(morpholinomethyl)isoquinolin-1(2H)-
one (Example 3);
2-(4-hydroxy-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 4);
3-(4-(2-hydroxy-2-methylpropoxy)-3,5-dimethylphenyl)-6,8-
dimethoxyisoquinolin-1(2H)-one (Example 5);
,7-(4-hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-5(6H)-
one (Example 6);
2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one (Example 7);
3-(3,5-dimethyl-4-(2-(4-methylpiperazin-1-yl)ethoxy)phenyl)-6,8-
dimethoxyisoquinolin-1(2H)-one (Example 8);
2-(4-hydroxy-3-methoxyphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 9);
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 10);
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-6,7-dimethoxyquinazolin-4(3H)-one
(Example 11);
2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxyquinazolin-4(3H)-one
(Example 12);

2-(4-((4-ethylpiperazin-1-yl)methyl)phenyl)-5,7-dimethoxyquinazolin-4(3H)-
one (Example 13);
2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxypyrido[2,3-
d]pyrimidin-4(3H)-one (Example 14);
2-(2-chloro-6-methylpyridin-4-yl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 15);
5,7-dimethoxy-2-(4-methoxy-3,5-dimethylphenyl)quinazolin-4(3H)-one
(Example 16);
2-(4-amino-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 17);
N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide (Example 18);
2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-
one (Example 18); and
4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide (Example 20).
13. The method according to embodiment 1, wherein R7 is an amino or
an alkoxy selected from the group represented by Formula III:

wherein:
A is selected from O and N;
n is selected from 0, 1, 2, 3, 4 and 5;

B is selected from-C(O)N(Rh)2-, -S(O)2N(Rh)2-, -C(O)-, -S(O)2-, -
C(O)O-, wherein each Rn is selected from alkyl, alkenyl, alkynyl, aryl, arylalkyl,
cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, and hydrogen; and
R20 is selected from (C1-C6) alkyl, (C1-C6) alkenyl, (C1-C6) alkynyl,
aryl, arylalkyl, cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, and hydrogen.
In another embodiment, if A is O and B is -C(O)NH-, then R20 is not an
unsaturated cycloalkyl.
14. The method of embodiment 13, wherein the compound of Formula II
is selected from:
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzenesulfonamide (Example 19);
N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide (Example 18);
4-chloro-A/-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide (Example 20);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)methanesulfonamide (Example 21);
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl propylcarbamate (Example 22);
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl methylcarbamate (Example 23);
N-(2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzamide (Example 24);
2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyI cyclohexylcarbamate (Example 25);

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide (Example 26);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzenesulfonamide (Example 27);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzamide (Example 28);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)acetamide (Example 29);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzamide (Example 30);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)isobutyramide (Example 31);
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-methylurea (Example 32);
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-(4-methoxyphenyl)urea (Example 33);
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-phenylurea (Example 34); and
3-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-1,1-dimethylurea (Example 35).
15. The method of embodiment 1, wherein the therapeutically effective
amount of the compound of Formula II is administered with a pharmaceutically
acceptable carrier in a pharmaceutically acceptable composition.
16. The method of embodiment 1, further comprising treating or
preventing a cardiovascular, cholesterol or lipid related disorder.

17. A compound of Formula II:

wherein:
X is selected from N and CH;
R1 and R3are each independently selected from alkoxy and hydrogen;
R2 is selected from alkoxy, alkyl, and hydrogen;
R6 and R8are each independently selected from alkyl, alkoxy, chloride, and
hydrogen;
R5 is hydrogen;
R7 is selected from amino, hydroxyl, alkoxy (preferably a substituted ethoxy
group), and alkyl substituted with a heterocyclyl, or
two adjacent substituents selected from R6, R7, and R8 are connected to
form a heterocyclyl;
each W is independently selected from C and N, wherein if W is N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
with the proviso that if R2 is selected from alkoxy or hydrogen, then at least
one of R1 and R3 is alkoxy;
with the proviso that if R7 is selected from hydroxyl or alkoxy, then at least
one of R6 and R8 are independently selected from alkyl, alkoxy, and chloride;

with the proviso that if R7 is an amino, then X is N;
with the proviso that if for W-(R7)P, W is N and p is 0, then at least one of R6
and R8 is chloride;
and pharmaceutically acceptable salts and hydrates thereof.
18. The compound according to embodiment 17, wherein the compound
is 3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1 (2H)-one (Example
1).
19. The compound according to embodiment 17, wherein the compound
is 7-(4-hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-5(6H)-one
(Example 6).
20. The compound according to embodiment 17, wherein the compound
is 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 7).
21. The compound according to embodiment 17, wherein the compound
of Formula II is selected from:
3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one
(Example 1);
3-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-
1(2H)-one (Example 2);
3-(4-hydroxy-3,5-dimethylphenyl)-7-(morpholinomethyl)isoquinolin-1(2H)-
one (Example 3);
2-(4-hydroxy-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 4);
3-(4-(2-hydroxy-2-methylpropoxy)-3,5-dimethylphenyl)-6,8-
dimethoxyisoquinolin-1(2H)-one (Example 5);

7-(4-hydroxy-3,5-dimethylphenyl)2,4-dimethoxy-1,6-naphthyridin-5(6H)-
one (Example 6);
2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one (Example 7);
3-(3,5-dimethyl-4-(2-(4-methylpiperazin-1-yl)ethoxy)phenyl)-6,8-
dimethoxyisoquinolin-1(2H)-one (Example 8);
2-(4-hydroxy-3-methoxyphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 9);
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 10);
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-6,7-dimethoxyquinazolin-4(3H)-one
(Example 11);
2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxyquinazolin-4(3H)-one
(Example 12);
2-(4-((4-ethylpiperazin-1-yl)methyl)phenyl)-5,7-dimethoxyquinazolin-4(3H)-
one (Example 13);
2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxypyrido[2,3-
d]pyrimidin-4(3H)-one (Example 14);
2-(2-chloro-6-methylpyridin-4-yl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 15);
5,7-dimethoxy-2-(4-methoxy-3,5-dimethylphenyl)quinazolin-4(3H)-one
(Example 16);
2-(4-amino-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
(Example 17);

N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yi)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide (Example 18);
2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-
one (Example 18); and
4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide (Example 20).
22. The compound according to embodiment 17, wherein R7 is an amino
or an alkoxy selected from the group represented by Formula III:

wherein:
A is selected from O and N;
n is selected from 0, 1, 2, 3, 4 and 5;
B is selected from -C(O)N(Rn)2-, -S(O)2N(Rh)2-, -C(O)-, -S(O)2-, -
C(O)O-, wherein each Rn is selected from alkyl, alkenyl, alkynyl, aryl,arylalkyl,
cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, and hydrogen; and
R20 is selected from (C1-C6) alkyl, (C1-C6) alkenyl, (C1-C6) alkynyl,
aryl, arylalkyl, cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, and hydrogen.
In another embodiment, if A is O and B is -C(O)NH-, then R20 is not an
unsaturated cycloalkyl.
23. The compound according to embodiment 22, wherein the compound
of Formula II is selected from

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzenesulfonamide (Example 19);
N1-(2-(4-(5,7-dimeth.oxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide (Example 18);
4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-
2,6-dimethylphenoxy)ethyl)benzenesulfonamide (Example 20);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)methanesulfonamide (Example 21);
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl propylcarbamate (Example 22);
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)2,6-dimethyl-
phenoxy)ethyl methylcarbamate (Example 23);
N-(2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzamide (Example 24);
2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl cyclohexylcarbamate (Example 25);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide (Example 26);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzenesulfonamide (Example 27);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzamide (Example 28);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)acetamide (Example 29);

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzamide (Example 30);
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)isobutyramide (Example 31);
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-methylurea (Example 32);
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-(4-methoxyphenyl)urea (Example 33);
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-phenylurea (Example 34); and
3-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-1,1-dimethylurea (Example 35).
24. A pharmaceutical composition comprising a compound according to
embodiment 17 and a pharmaceutically acceptable carrier.
25. A method of treating cardiovascular, cholesterol or lipid related
disorders comprising administering a therapeutically effective amount of a
compound according to embodiment 17.
26. A method of increasing expression of ApoA-l in a mammal
comprising administering a therapeutically effective amount of a compound
according to embodiment 17.
27. A method for increasing expression of ApoA-l in a mammal (e.g. a
human) comprising administering a therapeutically effective amount of a
compound selected from 6,8-dimethoxy-3-(4-hydroxy-3,5-dimethylphenyl)-2H-1,2-
benzothiazine-1,1-dioxide and 3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxy-7-
(morpholinomethyl)isoquinolin-1(2H)-one.

28. A compound selected from 6,8-dimethoxy-3-(4-hydroxy-3,5-
dimethylphenyl)-2H-1,2-benzothiazine-1,1 -dioxide and 3-(4-hydroxy-3,5-
dimethylphenyl)-6,8-dimethoxy-7-(morpholinomethyl)isoquinolin-1(2H)-one.
Pharmaceutical Formulations and Methods of Treatment
[076] The present disclosure also provides pharmaceutical compositions
comprising compounds as disclosed herein formulated together with one or more
pharmaceutically acceptable carriers. These formulations include those suitable
for oral, rectal, topical, buccal and parenteral (e.g. subcutaneous, intramuscular,
intradermal, or intravenous) administration, although the most suitable form of
administration in any given case will depend on the degree and severity of the
condition being treated and on the nature of the particular compound being used.
[077] Formulations suitable for oral administration may be presented in
discrete units, such as capsules, cachets, lozenges, or tablets, each containing a
predetermined amount of the compound as powder or granules; as a solution or a
suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-
oil emulsion. As indicated, such formulations may be prepared by any suitable
method of pharmacy which includes the step of bringing into association the active
compound and the carrier or excipient (which may constitute one or more
accessory ingredients). The carrier must be acceptable in the sense of being
compatible with the other ingredients of the formulation and must not be
deleterious to the recipient. The carrier may be a solid or a liquid, or both, and
may be formulated with the compound as a unit-dose formulation, for example, a
tablet, which may contain from about 0.05% to about 95% by weight of the active
compound. Other pharmacologically active substances may also be present
including other compounds. The formulations of the invention may be prepared by

any of the well known techniques of pharmacy consisting essentially of admixing
the components.
[078] For solid compositions, conventional nontoxic solid carriers include,
for example, pharmaceutical grades of mannitol, lactose, starch, magnesium
stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium
carbonate, and the like. Liquid pharmacologically administrable compositions can,
for example, be prepared by dissolving, dispersing, etc., an active compound as
described herein and optional pharmaceutical adjuvants in an excipient, such as,
for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to
thereby form a solution or suspension. In general, suitable formulations may be
prepared by uniformly and intimately admixing the active compound with a liquid
or finely divided solid carrier, or both, and then, if necessary, shaping the product.
For example, a tablet may be prepared by compressing or molding a powder or
granules of the compound, optionally with one or more accessory ingredients.
Compressed tablets may be prepared by compressing, in a suitable machine, the
compound in a free-flowing form, such as a powder or granules optionally mixed
with a binder, lubricant, inert diluent and/or surface active/dispersing agent(s).
Molded tablets may be made by molding, in a suitable machine, the powdered
compound moistened with an inert liquid diluent.
[079] Formulations suitable for buccal (sub-lingual) administration include
lozenges comprising a compound in a flavored base, usually sucrose and acacia
or tragacanth, and pastilles comprising the compound in an inert base such as
gelatin and glycerin or sucrose and acacia.
[080] Formulations of the present invention suitable for parenteral
administration comprise sterile aqueous preparations of the compounds, which

are approximately isotonic with the blood of the intended recipient. These
preparations are administered intravenously, although administration may also be
effected by means of subcutaneous, intramuscular, or intradermal injection. Such
preparations may conveniently be prepared by admixing the compound with water
and rendering the resulting solution sterile and isotonic with the blood. Injectable
compositions according to the invention may contain from about 0.1 to about 5%
w/w of the active compound.
[081] Formulations suitable for rectal administration are presented as unit-
dose suppositories. These may be prepared by admixing the compound with one
or more conventional solid carriers, for example, cocoa butter, and then shaping
the resulting mixture.
[082] Formulations suitable for topical application to the skin may take the
form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers and
excipients which may be used include Vaseline, lanoline, polyethylene glycols,
alcohols, and combinations of two or more thereof. The active compound is
generally present at a concentration of from about 0.1% to about 15% w/w of the
composition, for example, from about 0.5 to about 2%.
[083] The amount of active compound administered may be dependent on
the subject being treated, the subject's weight, the manner of administration and
the judgment of the prescribing physician. For example, a dosing schedule may
involve the daily or semi-daily administration of the encapsulated compound at a
perceived dosage of about 1 ug to about 1000 mg. In another embodiment,
intermittent administration, such as on a monthly or yearly basis, of a dose of the
encapsulated compound may be employed. Encapsulation facilitates access to
the site of action and allows the administration of the active ingredients

simultaneously, in theory producing a synergistic effect. In accordance with
standard dosing regimens, physicians will readily determine optimum dosages
and will be able to readily modify administration to achieve such dosages.
[084] A therapeutically effective amount of a compound or composition
disclosed herein can be measured by the therapeutic effectiveness of the
compound. The dosages, however, may be varied depending upon the
requirements of the patient, the severity of the condition being treated, and the
compound being used. In one embodiment, the therapeutically effective amount
of a disclosed compound is sufficient to establish a maximal plasma
concentration. Preliminary doses as, for example, determined according to animal
tests,' and the scaling of dosages for human administration is performed according
to art-accepted practices.
[085] Toxicity and therapeutic efficacy can be determined by standard
pharmaceutical procedures in cell cultures or experimental animals, e.g., for
determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the
dose therapeutically effective in 50% of the population). The dose ratio between
toxic and therapeutic effects is the therapeutic index and it can be expressed as
the ratio LD50/ED50. Compositions that exhibit large therapeutic indices are
preferable.
[086] Data obtained from the cell culture assays or animal studies can be
used in formulating a range of dosage for use in humans. Therapeutically
effective dosages achieved in one animal model may be converted for use in
another animal, including humans, using conversion factors known in the art (see,
e.g., Freireich et al., Cancer Chemother. Reports 50(4):219-244 (1966) and Table
1 for Equivalent Surface Area Dosage Factors).


[087] The dosage of such compounds lies preferably within a range of
circulating concentrations that include the ED50 with little or no toxicity. The
dosage may vary within this range depending upon the dosage form employed
and the route of administration utilized. Generally, a therapeutically effective
amount may vary with the subject's age, condition, and sex, as well as the
severity of the medical condition in the subject. The dosage may be determined
by a physician and adjusted, as necessary, to suit observed effects of the
treatment.
[088] In one embodiment, a compound as disclosed herein, or a
pharmaceutically acceptable salt or hydrate thereof, is administered in
combination with another therapeutic agent. The other therapeutic agent can
provide additive or synergistic value relative to the administration of a compound
of the present invention alone. The therapeutic agent can be, for example, a
statin; a PPAR agonist, e.g., a thiazolidinedione or fibrate; a niacin, a RVX, FXR
or LXR agonist; a bile-acid reuptake inhibitor; a cholesterol absorption inhibitor; a
cholesterol synthesis inhibitor; an ion-exchange resin; an antioxidant; an inhibitor
of AcylCoA cholesterol acyltransferase (ACAT inhibitor); a tyrophostine; a
sulfonylurea-based drug; a biguanide; an alpha-glucosidase inhibitor, an
apolipoprotein E regulator; a HMG-CoA reductase inhibitor, a microsomal

triglyceride transfer protein; an LDL-lowing drug; an HDL-raising drug; an HDL
enhancer; a regulator of the apolipoprotein A-IV and/or apolipoprotein genes; or
any cardiovascular drug.
[089] In one embodiment, a method of treating or preventing
cardiovascular disease, cholesterol- or lipid-related disorders, comprises
administering to a mammal (e.g., a human) a therapeutically effective amount of a
disclosed compound. The disclosed compound may be administered as a
pharmaceutically acceptable composition, comprising a disclosed compound and
a pharmaceutically acceptable carrier.
[090] As used herein, the term "cardiovascular disease" refers to diseases
and disorders of the heart and circulatory system. Exemplary cardiovascular
diseases, including cholesterol- or lipid-related disorders, include, but are not
limited to acute coronary syndrome, angina, arteriosclerosis, atherosclerosis,
carotid atherosclerosis, cerebrovascular disease, cerebral infarction, congestive
heart failure, congenital heart disease, coronary heart disease, coronary artery
disease, coronary plaque stabilization, dyslipidemias, dyslipoproteinemias,
endothelium dysfunctions, familial hypercholeasterolemia, familial combined
hyperlipidemia, hypoalphalipoproteinemia, hypertriglyceridemia,
hyperbetalipoproteinemia, hypercholesterolemia, hypertension, hyperlipidemia,
intermittent claudication, ischemia, ischemia reperfusion injury, ischemic heart
diseases, cardiac ischemia, metabolic syndrome, multi-infarct dementia,
myocardial infarction, obesity, peripheral vascular disease, reperfusion injury,
restenosis, renal artery atherosclerosis, rheumatic heart disease, stroke,
thrombotic disorder, transitory ischemic attacks, and lipoprotein abnormalities
associated with Alzheimer's disease, obesity, diabetes mellitus, syndrome X,

impotence, multiple sclerosis, Parkinson's diseases and an inflammatory
diseases.
[091] One embodiment provides methods for altering lipid metabolism in a
patient, e.g., increasing the ratio of HDL to LDL or ApoA-l to ApoB in the blood of
a patient, comprising administering to the patient a composition of the invention in
an amount effective to alter lipid metabolism.
[092] One embodiment provides methods for elevating the levels of ApoA-
I associated molecules, such as HDL, in the blood of a mammal, comprising
administering to the mammal a composition comprising a disclosed compound or
composition in an amount effective to elevate levels of ApoA-l and HDL
associated proteins in the mammal.
[093] In one embodiment, "treatment" or "treating" refers to an amelioration
of a disease or disorder, or at least one discernible symptom thereof. In another
embodiment, "treatment" or "treating" refers to an amelioration of at least one
measurable physical parameter, not necessarily discernible by the patient. In yet
another embodiment, "treatment" or "treating" refers to inhibiting the progression
of a disease or disorder, either physically, e.g., stabilization of a discernible
symptom, physiologically, e.g., stabilization of a physical parameter, or both. In
yet another embodiment, "treatment" or "treating" refers to delaying the onset of a
disease or disorder. For example, treating a cholesterol disorder may comprise
decreasing blood cholesterol levels.
[094] One embodiment provides a compound for administration to a
patient, such as a human, as a preventative measure against cardiovascular
diseases, including cholesterol- or lipid-related disorders. As used herein,
"prevention" or "preventing" refers to a reduction of the risk of acquiring a given

disease or disorder. An additional aspect provides a method for prevention of
arteriosclerosis lesion development in a mammal, including the development of
new arteriosclerotic lesions. In another aspect, the present invention provides a
method for regressing arteriosclerosis lesions.
[095] In another embodiment, the present compositions are administered
as a preventative measure to a patient, such as a human having a genetic
predisposition to a cardiovascular disease, including cholesterol- or lipid-related
disorders, for example familial hypercholeasterolemia, familial combined
hyperlipidemia, atherosclerosis, a dyslipidemia, a dyslipoproteinemia, or
Alzheimer's disease.
(096] In another embodiment, the compositions of the invention are
administered as a preventative measure to a patient having a non-genetic
predisposition to a cardiovascular disease, including cholesterol- or lipid-related
disorders, Examples of such non-genetic predispositions include, but are not
limited to, cardiac bypass surgery and percutaneous transluminal coronary
angioplasty, which often leads to restenosis, an accelerated form of
atherosclerosis; diabetes in women, which often leads to polycystic ovarian
disease; and cardiovascular disease, which often leads to impotence.
[097] Angioplasty and open heart surgery, such as coronary bypass
surgery, may be required to treat cardiovascular diseases, such as
atherosclerosis. These surgical procedures entail using invasive surgical devices
and/or implants, and are associated with a high risk of restenosis and thrombosis.
Accordingly, the compounds of the invention may be used as coatings on surgical
devices (e.g., catheters) and implants (e.g., stents) to reduce the risk of restenosis

and thrombosis associated with invasive procedures used in the treatment of
cardiovascular diseases.
[098] . In another embodiment, the present compositions may be used for
the prevention of one disease or disorder and concurrently treating another (e.g.,
prevention of polycystic ovarian disease while treating diabetes; prevention of
impotence while treating a cardiovascular disease).
[099] Diseases and conditions associated with "diabetes mellitus" as
defined herein refer to chronic metabolic disorder(s) caused by absolute or
relative insulin deficiency including, but not limited to hyperglycemia,
hyperinsulinemia, hyperlipidemia, insulin resistance, impaired glucose
metabolism, obesity, diabetic retinopathy, macular degeneration, cataracts,
diabetic nephropathy, glomerulosclerosis, diabetic neuropathy, erectile
dysfunction, premenstrual syndrome, vascular restenosis, ulcerative colitis, skin
and connective tissue disorders, foot ulcerations, metabolic acidosis, arthritis,
osteoporosis and impaired glucose tolerance.
PREPARATION OF COMPOUNDS
[0100] Exemplary compounds of the invention represented by the general
formula A:

wherein:
Ra may be selected from groups including, but not limited to, alkoxy, alkyl, alkenyl,
alkynyl, amide, amino, aryl, arylalkyl, carbamate, cycloalkyl, ether, halogen,

haloalkyl, heteroaryl, heterocyclyl, hydrogen and hydroxyl; Rb may be selected
from groups including, but not limited to, alkyl and hydrogen; X may be selected
from, e.g., CRC, IM and NRC, where Rc represents substituents such as alkyl,
alkenyl, alkynyl, and hydrogen; Y may be selected from, e.g., CO, CS, and SO2;
and Z3 may be a single or double bond; may be synthesized from readily available
starting materials as outlined in the exemplary schemes below. It should be
appreciated that these designations are non-limiting examples.

[0101] Scheme 1 illustrates that condensation followed by oxidation of
amide 1 and aldehyde 2 can provide quinazolinone 3. Condensation can occur
under a variety of conditions, such as NaHSO3 and p-TsOH in dimethylacetamide,
l2 in the presence of K2CO3, and treatment with catalytic trifluoroacetic acid
followed by DDQ oxidation.

[0102] Condensation of amide 4 with nitrile 5 in the presence of n-BuLi can
afford isoquinolinone 6, as shown in Scheme 2.


Scheme 3
[0103] Scheme 3 provides a method for synthesizing benzothiazine-1,1-
dioxide 9. Amide coupling of sulfonamide 7 with carboxylic acid 8 can be followed
by treatment with n-BuLi to afford 9.
EXAMPLES
[0104] Abbreviations used herein denote the following compounds,
reagents and substituents: acetic acid (AcOH); 2,2'-azobisisobutyronitrile (AIBN);
N-bromosuccinimide (NBS); N-tert-butoxycarbonyl (Boc); t-butyldimethylsilyl
(TBDMS); m-chloroperoxybenzoic acid (mCPBA); dimethylaminopyridine (DMAP);
dichloromethane (DCM); dimethylformamide (DMF); dimethylsulfoxide (DMSO);
ethanol (EtOH); ethyl acetate (EtOAc); 1-ethyl-3-(3-
dimethylaminopropyl)carbodiimide (EDCI); 1-hydroxybenzotriazole (HOBt);
iodomethane (Mel); lithium hexamethyldisilazide (LHMDS); methanol (MeOH);
methoxymethyl (MOM); tetrahydrofuran (THF); triethylamine (Et3N); lithium
aluminum hydride (LAH); p-toluenesulfonic acid (p-TSA); tetrabutylammonium
fluoride (TBAF); N-Methyl morpholine (NMM); N,N-dimethylacetamide (DMA);
twice daily (BID), once daily (QD).


3-(4-hydroxy-3.5-dimethylphenyl)-6.8-dimethoxyisoquinolin-1(2H)-one
[0105] To a suspension of 2-methyl-4,6-dimethoxy benzoic acid (2.61 g,
13.1 mmol) in CH2CI2 (50 mL), oxalyl chloride (3.38 g, 26.6 mmol) was added and
the mixture was stirred at room temperature for 16 h. The solvent and excess
oxalyl chloride were removed at reduced pressure. The solid was dissolved in
CH2GI2 (10 mL) and methyl amine (1.24 g, 39.9 mmol) with cooling and was
stirred at room temperature for 4 h. The solvent was removed and crude product
was purified by chromatography by using 5% methanol in CH2CI2 to give the
amide (2.27 g, 82%). To a solution of the above amide (2.27 g, 10.9 mmol) in
THF (50 mL), n-butyl lithium (9.98 mL, 25.0 mmol, 2.5 M solution in hexane) was
added slowly under nitrogen with cooling, maintaining the temperature below
20°C. The mixture was stirred for 1 h at 0°C, then cooled to -50°C, and a solution
of 4-O-TBDMS-3,5-dimethyl benzonitrile (2.97 g, 11.39 mmol) in THF (10 mL) was
added quickly, the cooling bath was removed and the mixture was stirred for 16 h
at room temperature. A saturated aqueous NH4CI solution was added with
cooling, and the layers were separated. The organic layer was washed with water,
brine, dried over Na2SO4and concentrated to give 3.9 g of the crude product
mixture. A suspension of the crude product mixture (3.9 g) in ethanol (20 mL) was
heated with cone. HCI (2 mL) at 80°C for 2 h. The reaction mixture was cooled to
room temperature and the solvent was removed. The solid was dissolved in water
and neutralized by NaHCO3, followed by extraction with CH2CI2. The product was

purified by chromatography to give two products: 3-(4-hydroxy-3,5-
dimethylphenyi)-6,8-dimethoxy-2-methylisoquinolin-1(2H)-one (128 mg, 5%) and
3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethpxyisoquinolin-1 (2H)-one (340 mg,
9%). Selected data for 3-(4-hydroxy-3,5-dimethylphenyl)-6,8-
dimethoxyisoquinolin-1(2H)-one: MS (ES) m/z: 326.00; MP 226-227°C.

3-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one
[0106] To a solution of 3,5-dimethyl-4-hydroxy benzonitrile (1.0 g,
6.79 mmol) in DMF (100 mL), were added a NaH (1.065 g, 26.63 mmol) and (2-
bromoethoxy)-tert-butyl dimethyl silane (1.95 g, 8.15 mmol). The reaction mixture
was stirred for 10 d at room temperature under nitrogen The reaction mixture was
poured into ice-water and the products were extracted with ethyl acetate. The
organic layer was separated, washed with water, dried and concentrated to give
crude product, which was purified by column chromatography to give 1.9 g of the
B-ring building block in 92% yield.
[0107] n-Butyl lithium (2.84 mL, 7.1 mmol, 2.5 M solution in hexane) was
added slowly to a solution of 2,4-dimethoxy-6-methyl benzamide (650 mg,
3.1 mmol) in THF (30 mL), under nitrogen with cooling (ice-salt bath), maintaining
the temperature below 20°C. After completion of addition, the mixture was stirred
for 1 h at 0°C, and then cooled to -50°C and a solution of 4-(2-tert-butyldimethyl
silanyloxy) ethoxy)-3,5-dimethyl benzonitrile (the B-ring building block, above)
(996 mg, 3.26 mmol) in THF (10 mL) was added quickly. The cooling bath was

removed and the reaction mixture was allowed to warm to room temperature and
was stirred for 16 h at room temperature. A saturated NH4CI solution was added
with cooling, and the layers were separated. The organic layer was washed with
water, brine, dried over Na2SO4 and concentrated to give 1.2 g of crude product.
[0108] The above crude product (1.2 g) was treated with ethanol (10 mL)
and cone. HCI (2 mL) at 80°C for 1 h. The solvent was removed and the residue
was dissolved in methanol and neutralized by NaHCO3. The solvent was
evaporated and crude product was purified by column chromatography to give 3-
(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one
(100 mg, 11%). Selected data: MP 193-195°C.

3-(4-hydroxy-3,5-dimethylphenyl)-7-(morpholinomethyl)isoauinolin-1(2H)-one
[0109] Hydrogen bromide in acetic acid (13 mL, 33 wt%) was added to a
mixture of 2-methyl benzoic acid (4.08 g, 30 mmol), paraformaldehyde (2.50 g,
83.0 mmol), and o-phosphoric acid (7 mL, 85%). The reaction mixture was stirred
at 115°C for 15 h. It was cooled to room temperature and poured into ice-cold
water. A white precipitate was formed. The mixture was extracted with ethyl
acetate (300 mL). The organic layer was washed with water (100 mL), brine
(100 mL) and dried over anhydrous Na2SO4. Removal of solvent gave 6.84 g of a
white solid, which was used in the next step without further purification. The above
compound (6.8 g) was dissolved in anhydrous dichloromethane (150 mL). Oxalyl

chloride (7.8 mL) was added drop-wise. After the addition was complete, 3 drops
of anhydrous DMF were added. A vigorous reaction occurred and the stirring was
continued overnight. Solvent and excess oxalylchloride were removed under
reduced pressure and the residue was dried under vacuum to give 7.02 g of
brown liquid, which was used in the next step without further purification. The
above compound (7.02 g, 28.36 mmol) was dissolved in anhydrous THF (60 mL)
and cooled to 0°C. A solution of N-methylamine (2.0 M in THF, 19 mL,
38.03 mmol) was added drop-wise under nitrogen. The stirring was continued for
15 min at 0°C. The ice-bath was removed, and the stirring was continued at room
temperature for 3 h. A white precipitate was formed. Water (100 mL) was added
and the mixture was extracted with ethyl acetate (150 mL). The organic layer was
separated, washed with water (50 mL), saturated NaHCO3 solution (2x50 mL),
water (50 mL), and brine (50 mL), and dried over anhydrous Na2SO4. Removal of
solvent gave 5.64 g of 5-bromomethyl-2,N-dimethylbenzamide as a white solid
which was used in the next step without further purification. To a solution of the
above compound (2.42 g, 10 mmol) in anhydrous THF was added morpholine
(1.92 g, 22 mmol) at room temperature under nitrogen. A white precipitate was
formed. Stirring continued overnight. Water (100 mL) was added and the mixture
was extracted with ethyl acetate (150 mL). The organic layer was separated,
washed with water (50 mL) and brine (50 mL) and dried (Na2SO4). Removal of
solvent gave a colorless oil, which was purified by column chromatography
(silica gel 230-400 mesh; 0-5% methanol in CH2CI2 as eluent) to give the desired
benzamide intermediate (yield 0.50 g, 20%). N-Butyl lithium (1.6 M solution in
hexanes, 4.1 mL, 6.6 mmol) was added drop-wise to a solution of the benzamide
(0.5 g, 2.0 mmol) in anhydrous THF (4 mL) at -10°C over a period of 10 min under

nitrogen. Stirring was continued at 0°C for 1 h. The reaction mixture was cooled to
-50°C. A solution of 4-(tert-butyldimethylsilanyloxy)-3,5-dimethylbenzonitrile
(0.653 g, 2.5 mmol) in anhydrous THF (3 mL) was quickly added. The cooling
bath was removed and the reaction mixture was allowed to warm to room
temperature. Stirring was continued at room temperature for 1 h. An aqueous
ammonium chloride solution (5 mL) was added followed by ethyl acetate (50 mL).
The organic layer was separated, washed with water (5 mL) and dried (Na2SO4).
Removal of the solvent gave 1.23 g pale yellow gummy material, which was used
in next step without further purification. The above compound (1.2 g) was
dissolved in 10 mL anhydrous ethanol. Cone. HCI (1 mL) was added and the
mixture was refluxed for 15 min, then cooled to room temperature. The solvent
was removed under reduced pressure. The crude compound was basified with
methanolic ammonia and purified by column chromatography (silica gel 230-400
mesh; 0-5% methanol in CH2CI2 as eluent) to give 3-(4-hydroxy-3,5-
dimethylphenyl)-7-morpholin-4-ylmethyl-2H-isoquinolin-1-one (35 mg) as a white
solid (the free base). To a solution of the above compound (35 mg) in CH2CI2
(5 mL) and MeOH (1 mL) was added drop-wise hydrogen chloride in ether
(0.5 mL, 1.0 M) under nitrogen. The reaction mixture was stirred at room
temperature for 1 h. The solvent was removed under reduced pressure and dried
under vacuum to give the hydrochloride of 3-(4-hydroxy-3,5-dimethylphenyl)-7-
(morpholinomethyl)isoquinolin-1(2H)-one (36 mg, 93%) as a yellow solid. Selected
data: MP 281-283°C (hydrochloride).


2-(4-hydroxy-3,5-dimethylDhenyl)-5.7-dimethoxyquinazolin-4(3H)-one
[0110] A solution of 3,5-dimethoxyaniline (199 g, 1.30 mol) in ether (5.0 L)
in a 5 L 3-necked flask was cooled to 0°C. HCI gas (227 g) was bubbled through
the solution over 45 min. After 45 min at 10°C, the mixture was filtered, washed
with isopropylacetate (4 L), and dried overnight on high vacuum at 45°C to give
the hydrochloride (242.3 g, 98%), as a white solid. A mixture of the hydrochloride
above (20 g, 0.105 mol) and oxalyl chloride (33 mL) in a 3-necked flask equipped
with a reflux condenser was heated for 2 h with stirring (170°C external
temperature), and the oxalyl chloride was distilled from the reaction mixture. The
flask was cooled to 0°C and methanol (40 mL) was added. The reaction mixture
was heated to reflux for 45 min, filtered while hot, and washed with methanol
(80 mL) to give the 4,6-dimethoxyisatin (17.2 g, 79%) as a yellow-green solid. To
a heated solution (external temp 70°C) of the isatin (162 g, 0.78 mol) in
aqueous NaOH (40%, 1.5 L) was added H2O2 (35%, 405 mL) slowly over 2 h.
After the addition of each portion of H2O2, the internal reaction temperature
(initially 64°C) increased (to a maximum temp of 80°C). After the addition was
complete, the foaming reaction mixture was then stirred for an additional 2 h at
70°C, and the mixture was allowed to stir overnight while cooling to RT. The
mixture was heated to 70°C. Additional H2O2 (75 mL) was added, and the mixture
was stirred at 70°C for a further 2 h until the reaction was complete. After cooling
to 10°C (bath temperature), aqueous Na2S2O3 (150 mL, saturated) was added.

The mixture was brought to pH 8 with HCI (37%, 1.6 L) and pH 6 with acetic acid
(glacial, 75 mL), without allowing the reaction mixture to warm to greater than
40°C. Filtration of the reaction mixture and washing with water (4 L) gave the
expected amino acid as a tan solid (83.7 g, 55%). To a solution of the amino acid
(82.7 g, 0.42 mol) in anhydrous THF (4.2 L) was added EDCI (89.2 g, 0.48 mol),
HOBT (65 g, 0.48 mol), and NMM (51.3 mL), and the mixture was allowed to stir
at RT for 3 h. Aqueous NH3 (83 mL, 50%) was added, and the mixture was stirred
at RT for 16 h. Water (1.25 L) was added, and the mixture was extracted with
DCM (2x250 mL). The combined extracts were then washed with water
(2x500 mL). Concentration, formation of a slurry with ether (550 mL), filtration,
and drying under high vacuum gave 2-amino-4,6-dimethoxybenzamide (46.7 g,
57%) as a brown solid.
[0111]2-Amino-4,6-dimethoxy-benzamide (1.06 g, 5.4 mmol), 3,5-dimethyl-
4-hydroxybenzaldehyde (0.810 g, 5.4 mmol), K2CO3 (0.747 g, 5.4 mmol) and l2
(1.645 g, 6.5 mmol) were mixed in DMF (20 mL) and the reaction mixture was
heated at 80°C for 12 h. It was cooled to RT and poured into crushed ice. The
solid was collected and purified by column chromatography to give 2-(4-hydroxy-
3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one (0.9 g, 51%) as a white
solid. Selected data: MP 291-293°C.

3-(4-(2-hydroxy-2-methylpropoxy)-3,5-dimethylphenyl)-
6,8-dimethoxyisoquinolin-1(2H)-one

[0112] To a solution of 4-hydroxy-3,5-dimethylbenzonitrile (2.00 g,
13.5 mmol) and 1-chloro-2-methyl propan-2-ol (8.85 g, 81.5 mmol) in ethanol
(50 mL) was added potassium carbonate (7.5 g, 54 mmol) and water (5 mL). The
reaction mixture was stirred at reflux for 24 h and cooled to RT. The precipitated
solid was filtered off and washed with water. The solid was dissolved in ethyl
acetate (100 mL), washed with water (50 mL), brine (50 mL), and dried over
anhydrous Na2SO4. Removal of solvent gave 4-(2-hydroxy-2-methylpropoxy)-3,5-
dimethyl benzonitrile (2.9 g, 97%) as a white solid.
[0113] To a solution of 4-(2-hydroxy-2-methylpropoxy)-3,5-dimethyl
benzonitrile (2.90 g, 13.2 mmol) in anhydrous DMF (20 mL) was added imidazole
(2.7 g, 40 mmol) and tert-butyldimethylsilylchloride (2.19 g, 14.6 mmol). The
reaction mixture was stirred at RT under nitrogen for 3 d. Water (200 mL) was
added and the mixture was extracted with ethyl acetate (200 mL). The organic
layer was washed with water (2x100 mL) and brine (100 mL), and dried over
anhydrous Na2SO4. The solvent was removed under reduced pressure and the
crude compound was purified by column chromatography to give 4-[2-(tert-
butyldimethylsilanyloxy)-2-methylpropoxy]-3,5-dimethylbenzonitrile (2.24 g, 54%).
n-Butyl lithium (6.2 mL, 6.6 mmol, 1.6 M solution in hexanes) was added to a
solution of 2,4-dimethoxy-6-N-dimethylbenzamide (0.9 g, 4.3 mmol) in anhydrous
THF (10 mL) drop-wise at -10°C over a period of 10 min under nitrogen. The
stirring was continued at 0°C for 1 h. The reaction mixture was cooled to -50°C. A
solution of 4-[2-(tert-butyldimethylsilanyloxy)-2-methylpropoxy]-3,5-
dimethylbenzonitrile (1.58 g, 4.73 mmol) in anhydrous THF (5 mL) was quickly
added. The cooling bath was removed and the reaction mixture was allowed to
warm to RT. The stirring was continued at RT for 1 h. An aqueous ammonium

chloride solution (10 mL) was added followed by ethyl acetate (100 mL). The
organic layer was separated, washed with water (10 mL) and dried (Na2SO4). The
solvent was removed under reduced pressure and the. crude compound was
purified by column chromatography (silica gel 230-400 mesh; 0-5% methanol in
CH2CI2 as eluent) to give 3-{4-[2-(tert-butyldimethylsilanyloxy)-2-methylpropoxy]-
3,5-dimethylphenyl}-6,8-dimethoxy-2H-isoquinolin-1-one (0.82 g, 37%), as a white
solid.
[0114] The above compound (0.42 g, 0.82 mmol) was dissolved in
anhydrous THF (20 mL). Tetrabutylammonium fluoride (4.1 mL, 1.0 M solution in
THF) was added at 0°C. The reaction mixture was stirred at 0°C for 10 min, then
at RT'for 2 h and then stirred at 70°C for 24 h. The mixture was cooled to RT.
Saturated aqueous ammonium chloride (30 mL) was added. The organic layer
was separated, washed with water, brine, and dried over anhydrous Na2SO4. The
solvent was removed under reduced pressure. The crude product was purified by
column chromatography (silica gel 230-400 mesh; 0-4% methanol in CH2CI2 as
eluent) to give 3-(4-(2-hydroxy-2-methylpropoxy)-3,5-dimethylphenyl)-
6,8-dimethoxyisoquinolin-1(2H)-one (0.15-g, 46%), as a white solid. Selected
data: MS (ES) m/z. 397.98; MP 252-254 °C at decomposition.

7-(4-hydroxy-3.5-dimethylphenyn-2.4-dimethoxy-1.6-naphthyridin-5(6H)-one
[0115] A mixture of malonic acid (20 g, 192 mmol), 2,4,6-trichlorophenol
(72 g, 365 mmol), and phosphorus oxychloride (38 mL, 403.2 mmol) was stirred at

reflux for 12 h. The reaction mixture was cooled to 70°C and poured into ice
water. The solid was collected by filtration, washed with water, and dried to give
malonic acid bis-(2,4,6-trichloro-phenyl) ester (85 g, 95%). A solution of malonic
acid bis-(2,4,6-trichloro-phenyl) ester (85 g, 184 mmol) and ethyl 3-
aminocrotonate (26.08 g, 201.9 mmol) in bromobenzene (100 mL) was stirred at
reflux for 50 min. The reaction mixture was cooled to 50°C and diluted with EtOAc
(260 mL). The solid was collected by filtration, washed with water, and dried
to give 4,6-dihydroxy-2-methyl nicotinic acid ethyl ester (31 g, 86%). A solution of
4,6-dihydroxy-2-methyl nicotinic acid ethyl ester (31 g, 157 mmol) in phosphorus
oxychloride (60 mL, 629 mmol) was stirred at reflux for 1.5 h. The extra
phosphorus oxychloride was removed and the reaction mixture was poured into
ice water. The solid was removed by filtration. The filtrate was extracted with
dichloromethane (3x100 mL) and concentrated. The residue was further purified
by column chromatography, to yield 4,6-dichloro-2-methyl nicotinic acid ethyl ester
(16.9 g, 46%). A solution of 4,6-dichloro-2-methyl nicotinic acid ethyl ester
(16.9 g, 71.3 mmol) in MeOH (60 mL) was mixed with sodium methoxide (58 mL,
256.68 mmol) and stirred at reflux for 12 h. The reaction was quenched by adding
HOAc (50 mL). The mixture was diluted with water (200 mL), extracted with
dichloromethane (3x100 mL), and concentrated. The residue was purified by
column chromatography (SiO2, hexanes/EtOAc = 6:1), to yield 4,6-dimethoxy-2-
methyl nicotinic acid methyl ester (10 g, 67%). A solution of 4,6-dimethoxy-2-
methyl nicotinic acid methyl ester (2.6 g, 12.3 mmol), lithium hydroxide (1.06 g,
44.08 mmol) in water (40 mL), MeOH (30 mL) and THF (20 mL) was stirred at
reflux for 4 h. The reaction mixture was concentrated to dryness. The residue was
mixed with HCI (cone, 20 mL) and was concentrated again on high vacuum to

dryness to yield crude 4,6-dimethoxy-2-methyl nicotinic acid (quantitative yield).
To a solution of 4,6-dimethoxy-2-methyl nicotinic acid (2.5 g, 12.0 mmol) in
dichloromethane (50 mL) and THF (50 mL) at room temperature was added oxalyl
chloride (2.57 mL, 29.4 mmol) and DMF (3 drops). The reaction mixture was
stirred at room temperature for 0.5 h, concentrated to dryness using a rotary
evaporator to afford crude 4,6-dimethoxy-2-methyl nicotinic acid chloride HCI salt
(2.8 g, quantitative). A solution of 4,6-dimethoxy-2-methyl nicotinic acid chloride
HCI salt (4.8 g, 23.5 mmol) in dichloromethane (100 mL) at room temperature was
poured into a beaker of ammonium hydroxide (200 mL). The reaction mixture was
stirred at room temperature for 1 h, extracted with dichloromethane (3x100 mL),
and concentrated using a rotary evaporator to yield 4,6-dimethoxy-2-methyl-
nicotinamide (2.4 g, 52%) as a light yellow solid. A solution of 4-hydroxy-3,5-
dimethyibenzonitrile (2.00 g, 13.59 mmol) in DMF (20 mL) at room temperature
was mixed with sodium hydride (0.706 g, 17.6 mmol) and stirred for 0.5 h. Benzyl
bromide (1.62 mL, 13.59 mmol) was added and the reaction mixture was stirred at
room temperature for 24 h. The reaction was quenched by adding water (200 mL),
extracted with EtOAc (3x100 mL), and concentrated. The residue was purified by
column chromatography to yield 4-benzyloxy-3,5-dimethylbenzonitrile (3.25 g,
100%) as a white solid. To a solution of 4,6-dimethoxy-2-methyl-nicotinamide
(1 g, 5.1 mmol) in THF (120 mL) at -20°C was added n-BuLi (9.6 mL, 15.3 mmol).
The reaction was stirred at -20-0°C for 2.5 h and then was cooled to -78°C. 4-
Benzyloxy-3,5-dimethylbenzonitrile (1.21 g, 5.1 mmol) was added, the cooling
bath was removed, and the reaction was allowed to warm up gradually to room
temperature. After stirring at room temperature for 20 h the reaction was
quenched by adding water (100 mL), extracted with dichloromethane (3x100 mL),

and concentrated using a rotary evaporator. The residue was further purified by
column (SiO2, Hexanes/EtOAc/MeOH = 3:2:1) to yield 7-(4-benzyloxy-3,5-
dimethyl-phenyl)-2,4-dimethoxy-[1,6]naphthyridin-5-ylamine (0.4 g, 19%) and 7-(4-
benzyloxy-3,5-dimethyl-phenyl)-2,4-dimethoxy-6H-[1,6]naphthyridin-5-one (0.34 g,
16%). A solution of 7-(4-benzyloxy-3,5-dimethyl-phenyl)-2,4-dimethoxy-6H-
[1,6]naphthyridin-5-one (0.34 g, 0.82 mmol) in DMF (100 mL) and MeOH (100 mL)
was mixed with palladium/carbon (0.1 g) and subjected to hydrogenation (50 psi)
for 2 h. The mixture was filtered through a Celite-pad. The filtrate was
concentrated on high vacuum to afford 7-(4-hydroxy-3,5-dimethyl-phenyl)-2,4-
dimethoxy-6H-[1,6]naphthyridin-5-one (0.23 g, 88%). A solution of 7-(4-hydroxy-
3,5-dimethyl-phenyl)-2,4-dimethoxy-6H-[1,6]naphthyridin-5-one (0.23 g, 0.7 mmol)
in MeOH (20 mL) and DCM (20 mL) was mixed with HCI in ether (7 mL, 7 mmol)
and stirred for 0.5 h. The reaction was concentrated using a rotary evaporator
to get a solid residue. The solid was rinsed with DCM, collected by filtration,
washed with DCM to yield the HCI salt of 7-(4-hydroxy-3,5-dimethylphenyl)-2,4-
dimethoxy-1,6-naphthyridin-5(6H)-one (0.15 g, 59%) as a light yellow solid.
Selected data: MS (ES) m/z: 327.06; MP >324°C at decomposition (HCI salt).

2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5.7-dimethoxyauinazolin-4(3H)-one
[0116] A solution of 2-amino-4,6-dimethoxybenzamide (0.60 g, 3.06 mmol)
and 4-[2-(tert-butyldimethylsilanoxy)ethoxy]-3,5-dimethylbenzaldehyde (0.856 g,
2.78 mmol) in N,N-dimethyl formamide (20 mL) was stirred at 70°C for 1 h. Iodine

(0.846 g, 3.33 mmol) and potassium carbonate (0.384 g, 2.78 mmol) were added
and the reaction mixture was stirred at 70°C for 16 h. The reaction mixture was
poured into ice, and extracted with ethyl acetate. The organic layer was washed
with water, brine, and dried over anhydrous Na2SO4. Removal of the solvent gave
the crude product which was purified by column chromatography to give 2-(4-(2-
hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one(444 mg,
39%) as a white solid. Selected data: 229-231 °C.
[0117] Alternatively, 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one can be synthesized by the following method. In a
2 L dry round-bottom flask with a reflux condenser and magnetic stirrer was
placed 3, 5-dimethyl-4-hydroxy benzaldehyde (26.9 g, 0.179 mol) in ethanol
(350 mL). 2-chloroethanol (87.6 g, 1.074 mol) and K2CO3 (99 g, 0.716 mol) were
added and the reaction mixture was heated to reflux for 24 h. The reaction mixture
was cooled to room temperature and filtered. The solvent was removed under
reduced pressure. The crude product was diluted with ethyl acetate and the
organic layer was washed with water, brine, and dried over Na2SO4. Upon
removal of solvent it gave 45 g of crude product. The crude product was purified
by column chromatography (silica gel 230-400 mesh; 50% ethyl acetate in hexane
as eluent) to give 33.3 g (95%) of product. To a solution of 2-amino-4, 6-
dimethoxy-benzamide (33.45 g, 0.170 mol) and 4-(2-hydroxy ethoxy)-3, 5-
dimethyl benzaldehyde (33.3 g, 0.170 mol) in N,N-dimethyl acetamide (300 mL),
NaHSO3 (33.3 g, 0.187 mol) and p-TSA (3.2 g, 17.1 mmol) were added and the
reaction mixture was heated at 150°C for 14 h. The reaction was cooled to room
temperature. The solvent was removed under reduced pressure. The residue was
diluted with water and stirred for 30 min at room temperature. The solids

separated were filtered and dried to give crude product. The crude product was
purified by column chromatography (silica gel 230-400 mesh; 5 % methanol in
CH2CI2 as eluent) to give 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (33 g, 52%).

3-(3.5-dimethyl-4-(2-(4-methylpiperazin-1-yl)ethoxy)phenyl)-6,8-
dimethoxyisoquinolin-1 (2H)-one
[0118] The compound 3-[4-(2-chloro-ethoxy)-3,5-dimethyl-phenyl]-6,8-
dimethoxy-isochromen-1-one (298 mg, 0.767 mmol) was dissolved in DMSO
(5 mL) and N-methyl piperazine (388 mg, 3.83 mmol) and Et3N (392 mg,
3.83 mmol) were added. The reaction mixture was heated at 110°C for 16 h
before being cooled to room temperature. Water was added and the mixture was
extracted with ethyl acetate. The solvent was evaporated in vacuo to leave a
residue which was purified by column chromatography. The yield was 60 mg
(17%). The compound 3-[3,5-dimethyl-4-(2-(4-methyl piperazin-1-yl-ethoxy)-
phenyl]-6,8-dimethoxy-isochromen-1-one (60 mg, 0.13 mmol) and NH3 (2.0 M
solution in ethanol, 20 mL) were mixed in a steel bomb and heated at 130°C for
16 h. The solvent was removed and the crude compound was purified by column
chromatography. The compound was then converted to the hydrochloride salt of
3-(3,5-dimethyl-4-(2-(4-methylpiperazin-1-yl)ethoxy)phenyl)-6,8-
dimethoxyisoquinolin-1(2H)-one (40 mg, 62%), an off-white solid. Selected data:
MS (ES) m/z: 452.1; MP 195-198°C (HCI salt).


2-(4-hydroxy-3-methoxyphenyl)-5.7-dimethoxyquinazolin-4(3H)-one
[0119] 2-(4-Hydroxy-3-methoxyphenyl)-5,7-dimethoxyquinazolin-4(3H)-one
was synthesized from 2-amino-4,6-dimethoxybenzamide and 4-hydroxy-3-
methoxybenzaldehyde, using the method described for 5,7-dimethoxy-2-(pyridin-
2-yl)quinazoiin-4(3H)-one. 2-(4-Hydroxy-3-methoxyphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (90 mg, 36%) was isolated as a white solid.
Selected data: MS (m/z): 329.06; MP 294-296°C.

2-(4-(bis(2-hydroxyethyl)amino)phenyl)-5,7-dimethoxyquinazolin-4(3H)-one
[0120]2-(4-(Bis(2-hydroxyethyl)amino)phenyl)-5,7-dimethoxyquinazolin-
4(3H)-one was synthesized from 2-amino-4,6-dimethoxybenzamide and 4-[bis-(2-
hydroxy-ethyl)-amino]-benzaldehyde, using the method described for 5,7-
dimethoxy-2-(pyridin-2-yl)quinazolin-4(3H)-one. 2-(4-(bis(2-
hydroxyethyl)amino)phenyl)-5,7-dimethoxy-quinazolin-4(3H)-one (120 mg, 41%)
was isolated as a yellow solid. Selected data: MS (m/z): 386.15; MP 249-251 °C.


2-(4-(bis(2-hydroxyethyl)amino)phenyl)-6.7-dimethoxyquinazolin-4(3H)-one
[0121]2-(4-(Bis(2-hydroxyethyl)amino)phenyl)-6,7-dimethoxyquinazolin-
4(3H)-one was synthesized from 2-amino-4,5-dimethoxy-benzamide and 4-(N,N-
bis(2-hydroxyethyl)amino)benzaldehyde, using the method described for 5,7-
dimethoxy-2-(pyridin-2-yl)quinazolin-4(3H)-one. 2-(4-(Bis(2-
hydroxyethyl)amino)phenyl)-6,7-dimethoxyquinazolin-4(3H)-one (72 mg, 24%)
was isolated as a yellow solid. Selected data: MS (m/z): 386.15; MP 268-270°C.

2-(2,3-dihydrobenzo[b][1M,4]dioxin-6-yl)-6,7-dimethoxyquinazolin-4(3H)-one
[0122]2-(2,3-Dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxyquinazolin-
4(3H)-one was synthesized from 2-amino-4,5-dimethoxybenzamide and 2,3-
dihydro-benzo[1,4]dioxine-6-carbaldehyde, using the method described for 5,7-
dimethoxy-2-(pyridin-2-yl)quinazolin-4(3H)-one. 2-(2,3-
Dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxy-quinazolin-4(3H)-one (180 mg,
69%) was isolated as a light yellow solid. Selected data: MS (m/z): 341.03; MP
316.4-318.2°C.


2-(4-((4-ethylpiperazin-1-yl)methyl)phenyl)-5.7-dimethoxyquinazolin-4(3H)-one
[0123] To a solution of 4-bromoethyl-benzoic acid ethyl ester (4.0 g,
16.46 mmol) in THF (30 mL), /V-ethy! piperazine (3.76 g, 32.92 mmol) was added
and the reaction mixture was stirred for 16 h at room temperature. The reaction
mixture was diluted with water and the product was extracted with ethyl acetate.
The combined organic layers were washed with water, brine, and dried over
Na2SO4. The solvent was removed to give 4.61 g of crude 4-(4-ethyl piperazin-1 -
ylmethyl)-benzoic acid ethyl ester (100% yield). LAH (0.792 g, 20.86 mmol) was
taken up in a 3-neck dry flask and THF (60 mL) was added on cooling. A solution
of 4-(4-ethyl piperazin-1-ylmethyl)-benzoic acid ethyl ester (4.61 g, 16.69 mmol) in
THF (10 mL) was added slowly on cooling. After completion of addition, the
reaction mixture was heated at reflux for 2 h. The reaction mixture was cooled to
0°C, 10% NaOH solution was added, and then water was added. The organic
layer was separated and the aqueous layer was extracted with ethyl acetate. The
combined organic layers were washed with water, brine and dried over Na2SO4.
The solvent was removed to give 2.78 g of crude (4-(4-ethyl piperazin-1-ylmethyl)
phenyl)-methanol in 78% yield. To a 3-neck flask containing anhydrous CH2CI2
(100 mL) cooled to the -78°C oxalyl chloride (1.8 g, 14.25 mmol) and DMSO
(1.85 g, 23.76 mmol) were added and the mixture was stirred for 15 min at -78°C.
The solution of (4-(4-ethyl piperazin-1-ylmethyl) phenyl)-methanol (2.78 g,
11.88 mmol) in CH2CI2 (10 mL) was added at -78°C and stirred at -78°C for 1 h.

Then Et3N (4.8 g, 47.52 mmol) was added at -78°C. The reaction mixture was
allowed to come to room temperature. Water was added and the organic layer
was separated. The aqueous layer was extracted with CH2CI2. The combined
organic layers were washed with water, brine and dried over Na2SO4. Then,
solvent was removed to give crude 4-(4-ethyl piperazin-1-ylmethyl)benzaldehyde
(2.5 g, 91%).
[0124] To a solution of 2-amino-4,6-dimethoxy-benzamide (150 mg,
0.76 mmol) and 4-(4-ethyl piperazin-1-ylmethyl)benzaldehyde (177 mg,
0.76 mmol) in N,/V-dimethyl acetamide (10 mL), NaHSO3 (150 mg, 0.84 mmol)
and p-TSA (319 mg, 1.68 mmol) were added and the reaction mixture was heated
at 150°C for 5 h. The reaction mixture was cooled to room temperature, water was
added and the mixture was neutralized with NaHCO3. The solvent was removed
under reduced pressure to give the crude product, which was purified by column
chromatography to give 2-(4-((4-ethylpiperazin-1-yl)methyl)phenyl)-5,7-dimethoxy-
quinazolin-4(3H)-one (87 mg, 27 %), which was converted to the hydrochloride
salt. Selected data: MS (ES) m/z: 409.11; MP 278-280°C (at decomposition).

2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxypyrido[2,3-d]pyrimidin-4(3H)-one
[0125] To a solution of 2-amino-4, 6-dimethoxy-nicotinamide (1.07 g,
5.42 mmol) and 4-[2-(tert-butyldimethylsilanoxy) ethoxy]-3, 5-
dimethylbenzaldehyde (1.67 g, 5.42 mmol) in N,N-dimethyl acetamide (25 mL),

NaHSO3 (1.06 g, 5.97 mmol) and p-TSA (1.14 g, 5.97 mmol) were added and the
reaction mixture was heated at 150°C for 16 h, cooled to room temperature and
poured into water. The solid was collected to give 3.25 g of crude product. To a
solution of the crude product (3.25 g, 6.70 mmol) in THF (50 mL), TBAF (3.5 g,
13.4 mmol) was added at 0°C and the mixture was stirred at room temperature for
1 h. The reaction mixture was quenched with water. The organic layer was
separated and the aqueous layer was extracted with ethyl acetate. The combined
organic layers were washed with water, brine and dried over Na2SO4. The solvent
was removed, and the crude was purified by column chromatography (silica gel
230-400 mesh; 2% methanol in CH2CI2 as eluent) to give 2-(4-(2-hydroxyethoxy)-
3,5-dimethylphenyl)-5,7-dimethoxypyrido[2,3-d]pyrimidin-4(3H)-one (132 mg, 6%).
Selected data: MS (ES) m/z: 371.99; MP 255-256°C.

2-(2-chloro-6-methylpyridin-4-yl)-5,7-dimethoxyquinazolin-4(3H)-one
[0126] Following the method described for 5,7-dimethoxy-2-(4-methoxy-3,5-
dimethylpheny!)quinazolin-4(3H)-one, 2-(2-chloro-6-methylpyridin-4-yl)-5,7-
dimethoxyquinazolin-4(3H)-one was synthesized from 2-amino-4,6-
dimethoxybenzamide and 2-chloro-6-methylisonicotinoyl chloride in 75% yield as
a white solid. Selected data: 1H NMR (300 MHz, CDCI3) δ 10.95 (s, 1H), 7.90 (s,
2H), 6.74 (d, J = 2.33 Hz, 1H), 6.51 (d, J = 2.32 Hz, 1H), 3.88 (s, 3H), 3.86 (s, 3H),
2.29 (s, 3H); MS (APCI) m/z 332 [M+H]+.


5,7-dimethoxy-2-(4-methoxy-3,5-dimethylphenvnauinazolin-4(3H)-one
[0127] To a solution of 4-methoxy-3,5-dimethylbenzoic acid (0.100 g,
0.555 mmol) in CH2CI2 (2.77 mL) cooled to 0-5°C was added oxalyl chloride (67.8
µL, 0.777 mmol) followed by drop-wise addition of DMF (4.3 µL, 0.056 mmol).
The mixture was stirred for 50 min, the volatiles were removed under vacuum, and
the crude acid chloride was used immediately without further purification.
[0128] To a mixture of 2-amino-4,6-dimethoxybenzamide (0.0990 g,
0.555 mmol) and pyridine (44.9 µL, 0.555 mmol) in THF (2.02 mL) was added
drop-wise a solution of the acid chloride (crude residue described above) in THF
(925 µL). After 16 h, the mixture was diluted with EtOAc (300 mL), washed with
saturated aqueous NH4CI (3x75 mL), saturated aqueous NaHCO3 (3x75 mL), and
brine (75 mL). The insoluble yellow solid was isolated by filtration to provide the
amide (0.150 g, 83%). A mixture of the amide (0.148 g, 0.413 mmol) and
2 M NaOH (7.00 mL) was heated at 85°C for 19 h, cooled to 5°C, and neutralized
with 4 M HCI in dioxanes. The white solid was filtered and rinsed with acetone to
provide 5,7-dimethoxy-2-(4-methoxy-3,5-dimethylphenyl)quinazolin-4(3H)-one
(0.144 g, 100%). Selected data: 1H NMR (300 MHz, CDCI3) δ 11.00 (s, 1H), 7.90
(s, 2H), 6.74 (d, J = 2.33 Hz, 1H), 6.51 (d, J = 2.32 Hz, 1H), 3.88 (s, 3H), 3.86 (s,
3H), 3.72 (s, 3H), 2.29 (s, 6H); MS (APCI) m/z 341 [M+H]+.


2-(4-amino-3.5-dimethylphenyl)-5,7-dimethoxyauinazolin-4(3H)-one
[0129] To a solution of 3,5-dimethyl-4-nitrobenzoic acid (1.00 g, 5.12 mmol)
in CH2CI2 (25.6 mL) cooled to 0-5°C was added oxalyl chloride (0.626 mL,
7.17 mmol) followed by drop-wise addition of DMF (39.8 µL). The mixture was
stirred for 2 h, the volatiles were removed under vacuum, and the crude acid
chloride was used immediately without further purification. To a mixture of 2-
amino-4,6-dimethoxybenzamide (0.913 g, 4.65 mmol) and pyridine (414 µL,
5.12 mmol) in THF (18.6 mL) was added drop-wise a solution of the acid chloride
(crude residue described above) in THF (8.53 mL). After 16 h, the mixture was
diluted with EtOAc (500 mL), washed with saturated aqueous NH4CI (3x100 mL),
saturated aqueous NaHCO3 (3x100 mL), and brine (100 mL). The insoluble
yellow solid was isolated by filtration to provide the amide (1.51 g, 87%). A
mixture of the amide (1.50 g, 4.03 mmol) and 2 M aqueous NaOH (25.0 mL) was
heated at 85°C for 17 h, then added THF (50 mL) and stirred at reflux for 25 h.
The volatiles were removed under vacuum, the mixture was cooled to 5°C, and
neutralized with 4 M HCI in dioxanes. After stirring for 30 min, the white solid was
filtered and lyophilized from MeCN/H2O to afford the cyclized compound (1.36 g,
95%). A mixture of the cyclized compound (0.200 g, 0.563 mmol), Na2S2O4
(0.980 g, 5.63 mmol), water (5.00 mL) and MeOH (15.0 mL) was stirred at 70°C
for 2 h. The volatiles were removed under vacuum, then diluted with EtOAc

(200 mL), washed with saturated NaHCO3 (2x100 mL) and brine (75 mL). The
organic layer was dried over sodium sulfate, filtered, and the volatiles were
removed under vacuum to provide 2-(4-amino-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (0.062 g, 34%) as a yellow solid. Selected data:
1H NMR (300 MHz, DMSO-d6) δ 11.45 (s, 1H), 7.78 (s, 2H), 6.66 (d, J = 2.25 Hz,
1H), 6.42 (d, J = 2.24 Hz, 1H), 5.26 (s, 2H), 3.88 (s, 3H), 3.86 (s, 3H), 2.14 (s,
6H); MS (APCI) m/z 326 [M+H]+.

N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide (left)
and
2-(4-(2-aminoethoxy)-3.5-dimethylphenyl)-5.7-dimethoxyquinazolin-4(3H)-one
(right)
[0130] A mixture of 3,5-dimethyl-4-hydroxybenzaldehyde (0.600 g,
4.00 mmol), /V-(2-bromoethyl)-phthalimide (1.22 g, 4.80 mmol), K2CO3 (0.829 g,
6.00 mmol), Nal (3.00 g, 20.0 mmol) in DMF (40.0 mL) was heated at 80°C for 2.5
h. The reaction was cooled to room temperature, diluted with EtOAc (200 mL),
washed with 1 M NaOH (2x100 mL), 1 M HCI (2x100 mL), brine (75 mL), dried
over sodium sulfate, filtered and concentrated under vacuum. The residue was
chromatographed on silica gel (40 g, hexanes/EtOAc) to provide the expected
ether (0.300 g, 23%) as a yellow solid. A mixture of the above ether (0.293 g,

0.907 mmol), 2-amino-4,6-dimethoxybenzamide (0.178 g, 0.907 mmol), NaHSO3
(94%, 0.100 g, 0.907 mmol), and p-TsOH.H2O (0.0173 g, 0.0907 mmol) in DMA
(11.3 mL) was stirred at reflux for 1.5 h then cooled to room temperature. The
mixture was diluted with EtOAc (250 mL), washed with saturated aqueous
ammonium chloride (3x75 mL) and brine (75 mL), dried over sodium sulfate,
filtered and concentrated under vacuum. The residue was chromatographed on
silica gel (40 g, CH2CI2/CH3OH) to provide the expected product (0.075 g, 17%)
as a light yellow solid. A mixture of the above compound (0.213 g, 0.426 mmol)
and 2 M methylamine in THF (25.0 mL) was stirred at room temperature for 17 h.
The volatiles were removed under vacuum and the residue was chromatographed
on silica gel to provide compound N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-
dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl)-N2-methylphthalamide
(0.0493 g, 22%) and compound 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (0.0360 g, 23%) as white solids. Selected data
for N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide: 1H NMR (300 MHz, DMSO-d6) δ
11.80 (s, 1H), 8.51 (t, J = 5.57 Hz, 1H), 8.18 (q, J = 4.57 Hz, 1H), 7.89 (s, 2H),
7.53-7.42 (m, 4H), 6.74 (d, J = 2.31 Hz, 1H), 6.52 (d, J = 2.29 Hz, 1H), 3.96-3.80
(m, 8H), 3.61 (q, J = 5.73 Hz, 2H), 2.71 (d, J = 4.62 Hz, 3H), 2.32 (s, 6H); MS
(APCI) m/z 531 [M+H]+. Selected data for 2-(4-(2-aminoethoxy)-3,5-
dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one: 1H NMR (300 MHz, DMSO-
d6) δ 7.90 (s, 2H), 6.74 (d, J = 2.31 Hz, 1H), 6.51 (d, J = 2.32 Hz, 1H), 3.88 (s,
3H), 3.85 (s, 3H), 3.77 (t, J = 5.76 Hz, 2H), 2.91 (t, J = 5.75 Hz, 2H), 2.30 (s, 6H);
MS (APCI) m/z 370 [M+H]+.


N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxv)ethyl)-4-methoxybenzenesulfonamide
[0131] A mixture of 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (0.060 g, 0.162 mmol), 4-methoxybenzenesulfonyl
chloride (0.044 mg, 0.211 mmol), and triethylamine (29.4 µL, 0.211 mmol) in
CH2CI2 (812 (µL) was stirred at room temperature for 3 h. The mixture was
chromatographed directly on silica gel to yield N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-
dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl)-4-
methoxybenzenesulfonamide (0.046 g, 53%) as a white solid after lyophilization
from MeCN/H20. Selected data: 1H NMR (300 MHz, DMSO-d6) δ ppm 11.81 (s,
1H), 7.88 (s, 2H), 7.83-7.73 (m, 3H), 7.17-7.07 (m, 2H), 6.73 (d, J = 2.31 Hz, 1H).
6.52 (d, J = 2.29 Hz, 1H), 3.91-3.75 (m, 11H), 3.12 (q, J = 5.75 Hz, 2H), 2.24 (s,
6H); MS (APCI) m/z 540 [M+H]+.

4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl):
2,6-dimethylphenoxy)ethyl)benzenesulfonamide

[0132] Following the method described for N-(2-(4-(5,7-dimethoxy-4-oxo-
3,4-dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl)-4-
methoxybenzenesulfonamide, compound 4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-
3,4-dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl)benzene-sulfonamide was
made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one in 51% yield and isolated as a white solid after lyophilization from
MeCN/H2O. Selected data: 1H NMR (300 MHz, DMSO-d6) δ ppm 11.8 (s, 1H),
8.1 (s, 1H), 7.9 - 7.6 (m, pH), 6.75 (1H), 6.5 (1H), 3.9 - 3.7 (m, 8H), 3.15 (m, 2H),
2.1 (s, 6H); MS (APCI) m\z 544 [M+H]+.

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)methanesulfonamide
i
[0133] Following the method described for N-(2-(4-(5,7-dimethoxy-4-oxo-

3,4-dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl)-4-
methoxybenzenesulfonamitie, compound N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-
i
j
dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl)methanesulfonamide was
i
made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one in 42% yield and isolated as a white solid after lyophilization from

MeCN/H2O. Selected data: 1H NMR (300 MHz, DMSO-d6) δ ppm 11.82 (s, 1H),

7.90 (s, 2H), 7.33 (t, J = 5.94 Hz, 1H), 6.74 (d, J = 2.31 Hz, 1H), 6.52 (d, J = 2.30


Hz, 1H), 3.92-3.81 (m, 8H), 3.41-3.34 (m, 2H), 2.97 (s, 3H), 2.32 (s, 6H); MS
(APCI) m/z 448 [M+H]+.


2-(4-(5.7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2.6-dimethyl-phenoxy)ethyl
propylcarbamate
[0134] A mixture of 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (0.070 g, 0.19 mmol), propyl isocyanate (0.088
mL, 0.94 mmol), and TEA (0.14 g, 1.1 mmol) in THF (4.0 mL) was stirred at 70°C
for 16 h. The mixture was filtered, washed with THF, and the solvent was
removed under reduced pressure. The residue was dissolved in EtOAc (50 mL)
and washed with saturated aqueous sodium bicarbonate (50 mL), dried and the
solvent was removed under reduced pressure. The resulting solid was
chromatographed on silica gel to yield 2-(4-(5,7-Dimethoxy-4-oxo-3,4-
dihydroquinazolin-2-yl)-2,6-dimethyl-phenoxy)ethyl propylcarbamate (0.035 g,
41%) as an off-white solid: Selected data: 1H NMR (300 MHz, DMSO-d6) δ 11.82
(s, 1H), 7.90 (s, 2H), 7.23 (t, J = 5.27 Hz, 1H), 6.74 (d, J = 2.32 Hz, 1H), 6.52 (d, J
= 2.31 Hz, 1H), 4.27 (t, J = 4.29 Hz, 2H), 3.99 (t, J = 4.29 Hz, 2H), 3.89 (s, 3H),
3.84 (s, 3H), 3.02-2.86 (m, 2H), 2.29 (s, 6H), 1.50-1.30 (m, 2H), 0.84 (t, J = 7.33
Hz, 3H); MS (APCI) m/z 456 [M+H]+.


2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2.6-dimethyl-Dhenoxy)ethyl
methylcarbamate
[0135] Following the method described for 2-(4-(5,7-Dimethoxy-4-oxo-3,4-
dihydroquina2olin-2-yl)-2,6-dimethyl-phenoxy)ethyl propylcarbamate, compound
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-phenoxy)ethyl
methylcarbamate was made from 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 11 % yield and isolated as an off-white solid: 1H
NMR (300 MHz, DMSO-d6) δ 11.82 (s, 1H), 7.90 (s, 2H), 7.08 (m, 1H), 6.74 (d, J =
2.29 Hz, 1H), 6.52 (d, J = 2.27 Hz, 1H), 4.27 (t, J = 4.55 Hz, 2H), 3.99 (t, J = 4.55
Hz, 2H), 3.89 (s, 3H), 3.84 (s, 3H), 2.60 (d, J = 4.57 Hz, 3H), 2.29 (s, 6H); MS
(APCI)m/z 428[M+H]+.

N-(2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzamide
[0136] A mixture of compound 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-
5,7-dimethoxyquinazolin-4(3H)-one (0.060 g, 0.16 mmol), p-toluoyl chloride (0.028

mL, 0.21 mmol), and PS-DIEA (0.057 g, 0.21 mmol) in CH2CI2 (4.0 mL) was
stirred at room temperature for 16 h. The mixture was filtered, washed with
CH2CI2 and the solvent was removed under reduced pressure. The resulting
residue was chromatographed on silica gel to yield N-(2-(4-(5,7-Dimethoxy-4-oxo-
3,4-dihydroquina2olin-2-yl)-2,6-dimethylphenoxy)ethyl)-4-methylbenzamide (0.037
g, 51%) as an off-white solid: 1H NMR (300 MHz, DMSO-d6) δ 11.80-11.00 (s,
1H), 8.69 (t, J = 5.43 Hz, 1H), 7.88 (s, 2H), 7.79 (d, J = 8.19 Hz, 2H), 7.28 (d, J =
8.00 Hz, 2H), 6.73 (d, J = 2.31 Hz, 1H), 6.51 (d, J = 2.31 Hz, 1H), 3.94 (t, J = 5.59
Hz, 2H), 3.88 (s, 3H), 3.84 (s, 3H), 3.72-3.60 (m, 2H), 2.36 (s, 3H), 2.27 (s, 6H);
MS (APCI) m/z 488 [M+H]+.

2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl
cyclohexylcarbamate
[0137] A mixture of 4-(6,8-dimethoxyisoquinolin-3-yl)-2,6-dimethylphenol
(0.100 g, 0.270 mmol), cyclohexylisocyanate (172 µL, 1.35 mmol), and Et3N (263
µL, 1.89 mmol) in THF (1.00 mL) was stirred at reflux for 4 h then diluted with
EtOAc (200 mL) and washed with saturated aqueous ammonium chloride (3 x 75
mL) and brine (75 mL). The organic layer was dried over sodium sulfate, filtered
and concentrated under vacuum. The residue was chromatographed on silica gel
(12 g, CH2CI2/CH3OH) and the product freeze dried from MeCN/H20 to provide 2-
(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethylphenoxy)ethyl
cyclohexylcarbamate (0.0981 g, 73%) as a white solid. 1H NMR (300 MHz,

DMSO-d6δ) 11.82 (s, 1H), 7.90 (s, 2H), 7.24-7.05 (m, 1H), 6.73 (d, J = 2.30 Hz,
1H), 6.52 (d, J = 2.31 Hz, 1H), 4.30-4.22 (m, 1H), 4.03-3.95 (m, 1H), 3.88 (s, 3H),
3.85 (s, 3H), 2.29 (s, 6H), 1.82-1.46 (m, 5H), 1.18 (m, 5H); MS (APCI) m/z496
[M+H]+

4-(2-(4-(6,8-dimethoxyisoquinolin-3-yl)-2,6-dimethylphenoxy)ethy)morpholine
[0138] To a solution of 4-(6, 8-dimethoxyisoquinolin-3-yl)-2, 6-
dimethylphenol (0.309 g, 1.0 mol) in anhydrous THF (20 mL), triphenyl phosphene
(0.52 g, 2.0 mmol), 4-(2-hydroxyethyl) morpholine (0.262 g, 2.0 mmol) and N,N-
diisopropylethylamine (0.387 g, 3.0 mmol) were added. To this stirred solution
was added diethylazodicarboxylate (0.348 g, 2.0 mmol). The reaction mixture was
stirred at room temperature overnight under nitrogen, then diluted with ethyl
acetate (100 mL). The organic layer was washed with water and brine, and dried
over anhydrous Na2SO4. The solvent was removed under reduced pressure. The
crude material was purified by column chromatography to give 3-[3,5-dimethyl-4-
(2-morpholin-4-ylethoxy) phenyl]-6,8-dimethoxyisoquinoline (0.54 g) as a white
solid.
[0139] To a solution of the above compound (0.54 g, impure) in 1:1
ether-CH2CI2 (10 mL), was added 1.0 M solution of hydrogen chloride in ether
(2 mL) and the reaction mixture was stirred at room temperature for 30 min.
Solvent was removed under reduced pressure. The residue was triturated with

10% methanol in ether to give 4-(2-(4-(6,8-dimethoxyisoquinolin-3-yl)-2,6-
dimethylphenoxy)ethyl)morpholine (0.323 g, 70% over two steps) as a yellow
solid. Selected data: MS (ES) m/z: 423.1; MP 239-240°C (HCI salt).

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide
[0140] Following the methodology described for Reference Example A,
the title compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 41% yield and isolated as an off-white solid:
MS(APCI)m/z 510[M+H]+.

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxv)ethyl)-4-methylbenzenesulfonamide
[0141] Following the methodology described for Reference Example A,
the title compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 50% yield and isolated as an off-white solid: MS
(APCI)m/z 524[M+H]+


5,7-dimethoxy-2-(pyridin-2-yl)quinazolin-4(3H)-one
[0142] To a solution of 2-amino-4,6-dimethoxybenzamide (0.15 g,
0.764 mmol) in N,/V-dimethyl acetamide (5 mL) were added 2-pyridine
carboxaldehyde (0.082 g, 0.764 mmol), sodium hydrogen sulphite (58.5%, 0.15 g,
0.84 mmol), and p-toluenesulfonic acid (15 mg, 0.0764 mmol). The reaction
mixture was stirred at 150°C overnight. The mixture was cooled to room
temperature. Water (40 mL) was added and the reaction mixture was extracted
with dichloromethane (2 x 50 mL). The combined organic layers were washed with

water and dried over anhydrous Na2SO4. The solvent was removed and the crude
compound was purified by column chromatography (silica gel 230-400 mesh; 1%
methanol in CH2CI2 as eluent) to give 5,7-dimethoxy-2-(pyridin-2-yl)quinazolin-
4(3H)-one (0.077 g, 36%) as a white solid. 5,7-dimethoxy-2-(pyridin-2-
yl)quinazolin-4(3H)-one was converted to the corresponding hydrochloride.
Selected data: MS (m/z): 284.0; MP 215-217°C (hydrochloride).

5,7-dimethoxy-2-(pyridin-3-yl)quinazolin-4(3H)-one
[0143] 5,7-Dimethoxy-2-(pyridin-3-yl)quinazolin-4(3H)-one was
synthesized from 2-amino-4,6-dimethoxybenzamide and 3-pyridine

carboxaldehyde, using the method described for Reference Example B. 5,7-
Dimethoxy-2-(pyridin-3-yl)quinazolin-4(3H)-one (105 mg, 48%) was isolated as a
white solid. Selected data: MS (m/z): 284.0; MP 257-259°C (hydrochloride).

N-(2-(4-(5,7-dimethoxy-4-oxo-3.4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzamide
[0144] Following the methodology described for Reference Example C,
the title compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 46% yield and isolated as a white solid: MS
(APCI) m/z 526 [M+Na]+.

N-(2-(4-(5,7-dimethoxy-4-oxo-3.4-dihydroauinazolin-2-yl)-2,6-
dimethylphenoxytethyl)acetamide
[0145] Following the methodology described for Example 27, the title
compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 40% yield and isolated as a white solid: MS
(APCI) m/z 412 [M+H]+.


N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-clihydroauinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzamide
[0146] Following the methodology described for Example 27, the title
compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 66% yield and isolated as a white solid: MS
(APCI) m/z 474 [M+H]+.

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroauinazolin-2-yl)-2,6-
dimethylphenoxv)ethynisobutyramide
[0147] Following the methodology described for Example 27, the title
compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 59% yield and isolated as a white solid: MS
(APCI) mlz 440 [M+H]+.


1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroauinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-methylurea
[0148] A mixture of compound 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-
5,7-dimethoxyquinazolin-4(3H)-one (0.10 g, 0.27 mmol), methylisocyanate (0.020
g, 0.35 mmol), and Et3N (0.034 g, 0.35 mmol) in THF (4.0 mL) was stirred at room
temperature for 16 hours. The mixture was filtered, washed with CH2CI2 and the
solvent was removed under reduced pressure. The resulting residue was
chromatographed on silica gel to yield the title compound (0.082 g, 71%) as a
white solid: MS (APCI) m/z 449 [M+Na]+.

1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-(4-methoxyphenyl)urea
[0149] Following the methodology described for Example 35, the title
compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazo!in-4(3H)-one in 57% yield and isolated as a white solid: MS
(APCI) m/z 541 [M+Na]+.


1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-phenylurea
[0150] Following the methodology described for Example 35, the title
compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 59% yield and isolated as a light yellow solid:
MS.(APCI) m/z 489 [M+H]+.

3-(2-(4-(57-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyn-1,1 -dimethylurea
[0151] Following the methodology described for Example 35, the title
compound was made from 2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one in 59% yield and isolated as a white solid: MS
(APCI) m/z 441 [M+H]+.


6,8-dimethoxy-3-(4-hydroxy-3,5-dimethylphenyl)-2H-1,2-benzothiazine-1,1 -dioxide
[0152] To a 3-necked, round-bottomed flask was added 3,5-
dimethoxytoluene (6.088 g, 40 mmol) and cyclohexane (28 mL) under nitrogen.
Dimethyl carbonate (30.3 g, 336 mmol) was added and the reaction mixture was
heated at 60°C. Excess chlorosulfonic acid was added over a period of 15 min.
The liberated HCI gas was removed by inserting a tube into solid sodium
hydroxide. On completion of the addition, the reaction mixture was heated to
70-72°C for 1 h and then cooled to room temperature. The solid was filtered off
and washed with dimethyl carbonate/cyclohexane (1:1, 20 mL). The solid was
dried in vacuo to obtain pure material (6.13 g, 66%). To a mixture of the sulfonic
acid (product from above, 4.65 g, 20 mmol) and triethyl amine (2.03 g, 2.79 mL) in
acetone (40 mL) was added 2,4,6-trichloro-1,3,5-triazine (cyanuric chloride,
3.69 g; 20 mmol). The reaction mixture was heated under reflux for 20 h before
being cooled to room temperature. The solution was passed through a Celite pad
and evaporated in vacuo to leave a solid, which was filtered off and washed with
hexane. The mixture of product and salt of cyanuric hydroxide and triethyl amine
(7.58 g) was used for the next step without further purification.
[0153] To a 3-necked, round-bottomed flask, equipped with a condenser
(acetone-dry ice cooling), was added the mixture from the step above (7.58 g) and
acetone (100 mL). The reaction mixture was cooled to -78°C and ammonia gas

was bubbled through the solution for 0.5 h. The reaction mixture was kept
standing overnight, allowing slow evaporation of ammonia gas, followed by the
evaporation of solvent. Water was added and the product was extracted with
DCM. The solvent was dried and evaporated to leave a mixture of solid and a
dense liquid. The solid was filtered off and washed with hexane to leave pure
sulfonamide (3.23 g, 70%).
[0154] To a round-bottomed flask was added 3,5-dimethyl-4-
hydroxybenzoic acid (2.99 g, 18 mmol). Anhydrous DMF (20 mL) was added,
followed by sodium hydride (1.8 g, 45 mmol). The reaction mixture was stirred at
room temperature for 1 h. p-Methoxybenzyl chloride (6.20 g, 39.6 mmol) was
added and the mixture was stirred at room temperature overnight (-20 h). The
reaction mixture was poured into water, acidified with 1 N HCI and stirred for 1 h.
The precipitated solid was filtered off, washed with water and hexane to obtain
pure B-ring building block (6.93 g, 95%).
[0155] The B-ring building block (6.93 g, 17.1 mmol) was dissolved in a
mixture of methanol (50 mL) and tetrahydrofuran (50 mL). Potassium hydroxide
(1.25 g, 22.2 mmol) in water (20 mL) was added. The reaction mixture was
refluxed at 70°C for 24 h. The solvent was evaporated in vacuo. Water was added
and the reaction mixture was acidified with 1 N HCI (pH 4-5). The solid was
filtered off, washed with water and hexane. The yield was 4.61 g (94%). The
product (1.932 g, 6.75 mmol) and the sulfonamide from above (1.04 g, 4.5 mmol)
were taken in a 3-necked, round-bottomed flask under nitrogen. Dichloromethane
(100 mL) was added with stirring. To this stirred mixture was added N-(3-
dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (EDCI. HCI, 1.36 g,
7.09 mmol), followed by N,N-dimethylaminopyridine (2.06 g, 16.9 mmol). The

reaction mixture was stirred at room temperature for 24 h before being washed
with 1 N HCI, 2.5% NaOH and saturated sodium bicarbonate solutions. The
organic layers were dried and evaporated in vacuo to leave a residue, which was
purified by silica gel (100 g) column chromatography, employing 20-50% ethyl
acetate in hexane and 5% methanol in dichloromethane as eluents. Fractions
30-66 were combined to obtain pure materials (1.35 g, 60%). The compound from
the step above (0.105 g, 0.21 mmol) was dissolved in tetrahydrofuran under
nitrogen and cooled to -78°C. n-Butyllithium was added and the reaction mixture
was allowed to warm to room temperature slowly and stirred overnight (-14 h).
TLC showed incomplete conversion. The reaction mixture was quenched with
saturated ammonium chloride solution and extracted with ethyl acetate. The
solvent was evaporated in vacuo to leave a residue that was purified by silica gel
(15 g) column chromatography, employing 20-50% ethyl acetate in hexane as
eluents. The product was not pure enough, so another column was used,
employing 0.5% methanol in hexane as eluent, and finally preparative TLC was
employed to purify the material. The compound from the step above (0.277 g) was
dissolved in trifluoroacetic acid (10 mL) under nitrogen and the reaction mixture
was refluxed (bath temperature 80°C) for 4 d. The solvent was evaporated in
vacuo and the residue was dissolved in 0.25 N NaOH (20 mL), and acidified with
acetic acid. The solid had precipitated out at this point. The solid was filtered off
and washed with water, hexane and dried. From one batch, 0.005 g of pure
material was isolated. From another batch, 0.060 g compound was isolated, which
was not pure enough. This compound was further purified by preparative HPLC
to give pure 6,8-dimethoxy-3-(4-hydroxy-3,5-dimethylphenyl)-2H-1,2-
benzothiazine-1,1-dioxide (0.010 g). Selected data: MP 246.6-247.4°C.


3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxy-7-
(morpholinomethyl)isoquinolin-1(2H)-one
[0156] Methyl acetoacetate (69.67 g, 0.6 mol) in dry THF (350 mL) was
cooled to -5°C and sodium hydride in mineral oil (24.5 g, 60%) was added at -5 to
0°C over 30 min. Diketene (50.4 g) in dry THF (80 mL) was added drop-wise at
5°C over 20 min. The resulting solution was allowed to stir for 1.0 h at -5°C, after
which it was allowed to warm to room temperature and stir overnight. Acetic acid
(35 mL) was added and the THF solvent was removed. Water (200 mL) and ethyl
acetate (300 mL) were added to the residue and the pH was adjusted to 5.0 by
addition of HCI solution. The organic layer was separated and washed with brine
and dried over sodium sulfate. After column purification and recrystalization,
compound A (26.6 g, 24.3%) was obtained.
[0157] Sodium hydride in mineral oil (11.2 g, 0.279 mol, 60%) was added
to compound A (24.8 g, 0.136 mol) in DMF (150 mL). The reaction was cooled to
-30°C and methyl iodide (21.3 mL, 0.341 mol) was added and the reaction was
kept at room temperature overnight. Sodium iodide was filtered off and DMF was
removed. The residue was mixed with water (100 mL) and extracted with ethyl
acetate. The organic layer was washed with brine and dried over sodium sulfate.
The crude mixture was purified by column chromatography to yield compound B
(11.40 g, 39.9%). To a solution of compound B (11.4 g, 0.054 mole) in dry CCl4

(90 mL) was added N-bromosuccinimide (10.6 g, 0.0596 mol). The mixture was
refluxed overnight and CCI4 solvent was removed. Water (100 mL) was added to
the residue. After stirring for a while the solid was filtered off and washed with
water, ethyl acetate (10 mL) and hexane (30 mL) to yield compound (13.1 g,
83.9%). Compound C (12.5 g, 0.043 mol), chloromethyl methyl ether (81.0 g) and
anhydrous zinc chloride (7.0 g, 0.051 mol) were kept at room temperature
overnight. Chloromethyl methyl ether was removed and the residue was mixed
with water and the pH was adjusted to 7.0 using sodium bicarbonate. The mixture
was extracted with ethyl acetate. The organic layer was washed with brine and
dried over sodium sulfate. Compound D (7.39 g, 50.6%) was obtained after
column chromatography. A solution of compound D (7.39 g, 0.022 mol),
morpholine (7.62 g, 0.088 mol) and anhydrous THF (20 mL) was kept at room
temperature overnight. The solvent was evaporated. Water and ethyl acetate
were added to the residue, and pH was adjusted to 9.0 with sodium bicarbonate.
The organic layer was washed with brine and dried over sodium sulfate, and
concentrated. Compound E (5.4 g, 63.8%) was obtained after column
chromatography. The hydrogenation reaction was carried out at 50 psi with
compound E (5.4 g, 0.014 mol) in THF (100 mL) and triethyl amine (3.9 mL) with
10% Pd/C (2.6 g) as a catalyst for 2 d. After the catalyst was filtered off, the
organic layer was purified by column chromatography to yield product F (3.20 g,
74.4%). Compound F (3.20 g, 0.0103 mol) was dissolved in ethanol (30 mL) and
potassium hydroxide (2.31 g, 0.041 mol) in water (20 mL) was added and the
reaction mixture was heated to 100°C overnight. The solvent was removed, pH
was adjusted to 6.0 and the water was removed. The residue was further dried
under high vacuum and the compound was extracted with ethanol to yield

compound G (2.95 g, 99%). Compound G (1.80 g, 6.1 mmol) with thionyl chloride
(3 mL, 0.0411 mol) was refluxed for 1 h before the excess thionyl chloride was
removed and the residue was dried under high vacuum. Anhydrous THF (20 mL)
was added and ammonia gas was bubbled into the reaction mixture for 2 h. THF
was removed and pH was adjusted to 8.0-9.0. The mixture was extracted with
dichloromethane and dried over sodium sulfate to give compound H (1.30 g,
72.4%).
[0158] NaH in mineral oil (1.14 g, 0.0285 mol, 60%) was added to 4-
hydroxy-3,5-dimethylbenzonitrile (4.0 g, 0.027 mol) in anhydrous DMF (20 mL)
followed by benzyl bromide (3.27 mL, 0.027 mol). The reaction was kept at room
temperature overnight. The reaction mixture was poured into water and the solid
was filtered off and washed with hexane to yield Compound I (5.7 g, 89%).
Compound I was used for the next step reaction without further purification. BuLi
(1.60 M, 10.2 mL) was added drop-wise to compound H (0.8 g, 2.72 mmol) in
anhydrous THF (25 mL) at -10°C. The reaction mixture was kept at 0°C for one h
before the cooling bath was removed. The reaction mixture was stirred for 45
minutes. Compound I (0.65 g, 2.72 mmol) in anhydrous THF (5 mL) was added
drop-wise at -10°C and the reaction was continued for a further 45 min. Water
(20 mL) was added. The mixture was extracted with ethyl acetate. The solvent
was removed and the residue was purified by column chromatography to yield
compound J (0.180 g, 12.8%). Compound J (180 mg) in methanol (80 mL) was
hydrogenated at 50 psi for 3 h, using 10% Pd/C as the catalyst. The catalyst and
solvent were removed and the residue was purified by column chromatography to
yield 3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxy-7-

(morpho,inomethyl)isoquinolin-1(2H)-one (28 mg, 18.8%) as a white solid.
Selected data: MS (m/z): 424.21; MP 158-161°C.
Example 38: Quantification of ApoA-l mRNA
[0159] In this example, ApoA-l mRNA in tissue culture cells was quantitated
to measure the transcriptional up-regulation of ApoA-l when treated with a
compound of the invention.
[0160] HepG2 cells (~2x1o5 per well) were placed in a 24-well plate in
~400 µL MEM, supplemented with 0.5% (v/v) FBS, 24 h before addition of the
compound of interest. At time of harvesting, the spent media was removed from
the HepG2 cells and immediately placed on ice (for immediate use) or at -80°C
(for future use) in ApoA-l and albumin ELISAs. The cells remaining in the plate
wells were rinsed in 200 µL PBS. PBS was carefully removed to avoid removing
any loosely attached cells.
[0161] Once the PBS was removed, 85 µL cell lysis solution was added the
cells in each well and incubated for 5-10 min at room temperature, to allow for
complete cell lysis and detachment. mRNA was then prepared using the "mRNA
Catcher PLUS plate" from Invitrogen, according to the protocol supplied. After the
last wash, as much wash buffer as possible was aspirated without allowing the
wells to dry. Elution Buffer (E3, 80 µL) was then added to each well. mRNA was
then eluted by incubating the mRNA Catcher PLUS plate with Elution Buffer for
5 min at 68°C and then immediately placing the plate on ice.
[0162] The eluted mRNA isolated was then used in a one-step real-time
room temperature-PCR reaction, using components of the Ultra Sense Kit
together with Applied Biosystems primer-probe mixes. Real-time PCR data was
analyzed, using the Ct values, to determine the fold induction of each unknown

sample, relative to the control (that is, relative to the control for each independent
DMSO concentration).
[0163] An active compound is one that causes a >15% increase in ApoA-l
mRNA at a concentration less than or equal to 100 µM.



Example 39: ApoA-l mRNA and protein induction
[0164] In this example, ApoA-l mRNA and secreted protein from tissue
culture cells were quantitated. The assay can be used to determine the potency
for compounds of interest, including those of the present invention.
[0165] HepG2 cells and primary human hepatocytes (BD Gentest, lot 107)
(~2*105 per well) were placed in a 24-well plate in ~400 µL MEM, supplemented
with 0.5% (v/v) FBS, 24 h before addition of the compound of interest. The
compounds of interest were dissolved in DMSO at 0.05% (v/v). Appropriate
volumes of the stock solutions of the compounds in DMSO were then added to
appropriate volumes of MEM, supplemented with 0.5% (v/v) FBS, to achieve the
desired concentration (for example, 1 µL of a compound stock into 1 mL of MEM,
supplemented with 0.5% (v/v) FBS).
[0166] Just prior to compound addition to the cells, the growth media was
aspirated and replaced with 300 µL of fresh MEM, supplemented with 0.5% (v/v)
FBS, followed by addition of 300 µL of the compound of interest in MEM,

supplemented with 0.5% (v/v) FBS to achieve the desired fina, compound
concentration in a total volume of 600 µL The final concentration of diluent
(DMSO) was 0.05% (v/v).
[0167] Cells were incubated for the desired time. The cell media was then
harvested, as were the cells. ApoA-l mRNA was measured as described in
Example 39. Secreted ApoA-l was measured using an ApoA-l ELISA, as
described below:
ApoA-l ELISA
[0168] In this example, the ApoA-l secreted into the media from tissue
culture cells was quantitated to assess induction of endogenous ApoA-l protein
secretion from cells treated with various small molecule compounds, such as

those of the present invention.
[0169] At time of harvesting, spent media from the HepG2 cell cultures or
primary cell culture was removed and stored at -80°C in 1.5 mL microfuge tubes.
[0170] For the human ApoA-l ELISA, an ELISA plate was coated with
~100 µL/well human ApoA-l capture antibody diluted to -2 µg/mL in coating buffer
for ~1 h at room temperature. The plate was then washed three times in wash
buffer. The plate was then blocked with -200 µL/well human ApoA-l blocking
buffer for at least ~30 min at room temperature.
[0171] Samples for use in generating a standard curve were prepared
from spent media (MEM, supplemented with 0.5% (v/v) FBS) from HepG2 or
primary cells treated with DMSO for 48 h. Serial 2 fold dilutions of the media were
prepared in MEM, supplemented with 0.5% (v/v) FBS. The unknown samples,
from the cultures treated with the compounds of interest, were also diluted in

MEM, supplemented with 0.5% (v/v) FBS. The plate was washed three times in
wash buffer. The standard curve and unknown samples (100 µL/well), in
triplicate, were added to the plate and it was incubated for 1.5 h at room
temperature.
[0172] The plate was washed three times in wash buffer. Human ApoA-l
detection antibody, diluted 1:1000 in PBS, was added (100 µL/well) and the plate
was incubated for 1 h at room temperature. The plate was washed three times in
wash buffer.
[0173] Goat anti-rabbit IgG H & L chain specific peroxidase conjugate,
diluted 1:2000 in PBS, was added (100 µL/well) and the plate was incubated for
40 min at room temperature in the dark. The plate was washed six times in wash
buffer.
[0174] TMB liquid substrate was added (100 µL/well) and the plate was
incubated on a shaker underneath tin foil during development. Once a sufficient
"blue" color had been achieved, stop solution (50 µL/well, 1 M H2SO4) was added
and mixed thoroughly on the plate shaker. Air bubbles were removed and the
absorbance at 450 nm was determined, using a Molecular Devices SpectraMax
190 Plate Reader and the human ApoA-l ELISA Softmax software.



EXAMPLE 40: In vivo efficacy
[0175] To test whether the efficacy of compounds of the invention observed
in vitro extended to an in vivo model, transgenic mice carrying multiple copies of
the human ApoA-l gene (Bisaha et al. (1995) J. Biol. Chem. 34,19979-88) or wild-
type mice (C57BL/6 (Stock Number 000664) Jackson Laboratory (Bar Harbor,
ME)) were exposed to compounds of the invention. In the transgenic mice, the
exogenous human ApoA-l gene in these mice enables them to express the human
ApoA-l protein under the control of its own promoter.
[0176] Seven to eight week old male mice were housed five per cage
(10"x20"x8" with aspen chip bedding) with pelleted Rodent chow [Purina 5001]
and water available at all times. After an acclimation period of 1 week, animals
were individually identified by numbering on tail and weighed. Mice were pre-bled
via the retro-orbital plexus, and 100 µL of blood was collected in 1.5 mL

Eppendorf tube containing 5 µL of 0.5 mM EDTA and chilled on ice. Plasma was
collected after centrifuging the whole blood at 14000 rpm [TOMY high speed
micro-refrigerated centrifuge NTX-150] for 10 min at 4°C and frozen at -80°C.
Mice were grouped based on having an average body weight of 25 g.
[0177] A day following pre-bleed, mice were dosed by oral gavage or by
i.p. administration daily using a 20 gauge, 11/2" curved disposable feeding needle
(Popper & Sons); when B.I.D., mice were gavaged morning and afternoon (8 am
and 5 pm); when Q.D. mice were gavaged in morning (8 am). Compounds were
prepared each day in vehicle. One day prior to necropsy mice were weighed and
fasted overnight. On final day of dosing, mice were sacrificed post 2 h of dosing
by inhalation of CO2 and blood was obtained via cardiac puncture (0.7-1.0 mL).
Plasma was collected and frozen at -80°C. Samples were assayed for ApoA-l by
ELISA, and HDL-C by HPLC (Polaris 200 with an auto sampler Prostar 410 from
Varian on a Superose 6 10/30 column from Amersham). During necropsy, liver
and enterocytes from the duodenum and jejunum of small intestine were
collected, cleaned with cold PBS and frozen at -80°C for further analysis of
compound and mRNA levels by Q-PCR.
[0178] Experiment A 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (10, 30 and 60 mg/kg of body weight, mpk) were
BID administered to hApoA-l transgenic mice daily for seven days by oral gavage
in 1% DMSO, 2.5% Tween-80, 10% PEG-300 QS to water. Plasma was assayed
for ApoA-l (Fig. 1), and HDL cholesterol (Fig. 2).
[0179] Experiment B 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (10, 30 and 60 mg/kg of body weight) were BID
administered to wild-type C57BL/6 mice daily for three days by i.p. administration

in 1% DMSO, 2.5% Tween-80, 10% PEG-300 QS to water. Plasma was assayed
for ApoA-l (Fig. 3), and HDL cholesterol (Fig. 4).
[0180] Experiment C 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-
dimethoxyquinazolin-4(3H)-one (30 mg/kg of body weight) were BID administered
to hApoA-l transgenic mice daily for seven days by oral gavage in 1% DMSO,
2.5% Tween-80, 10% PEG-300 QS to water. Plasma was assayed for ApoA-l
and tissues were assayed for mRNA (Fig. 5).
[0181] These results indicate that the compounds of the invention are
useful for increasing the transcription of ApoA-l in vivo, and elevating plasma
levels of ApoA-l and circulating levels of HDL-C in wild-type and hApoA-l
transgenic mice. These results demonstrate that compounds of the invention
activate the human ApoA-l transgene in mice, leading to an increase in circulating
ApoA-l.
[0182] All references referred to herein are incorporated by reference in
their entirety. Other embodiments of the invention will be apparent to those skilled
in the art from consideration of the specification and practice of the invention
disclosed herein. It is intended that the specification and examples be considered
as exemplary only, with a true scope and spirit of the invention being indicated by
the following claims.

WHAT IS CLAIMED IS:
1. Use of a therapeutically effective amount of a compound of Formula II in
the preparation of a medicament for increasing expression of ApoA-l in a
mammal:

wherein:
X is selected from N and CH;
R1 and F3 are each independently selected from alkoxy and hydrogen;
R2 is selected from alkoxy, alkyl, and hydrogen;
R6 and R8 are each independently selected from alkyl, alkoxy, chloride, and
hydrogen;
R5 is hydrogen;
R7 is selected from amino, hydroxyl, alkoxy, and alkyl substituted with a
heterocyclyl, or
two adjacent substituents selected from R6, R7, and R8 are connected to
form a heterocyclyl;
each W is independently selected from C and N, wherein if W is N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;

with the proviso that if R2 is selected from alkoxy or hydrogen, then at least
one of R1 and R3 is alkoxy;
with the proviso that if R7 is selected from hydroxyl or alkoxy, then at least
one of R6and R8are independently selected from alkyl, alkoxy, and chloride;
with the proviso that if R7 is amino, then X is N;
with the proviso that if for W-(R7)P, W is N and p is 0, then at least one of R6
and R8 is chloride;
and pharmaceutically acceptable salts and hydrates thereof.
2. The use according to claim 1, wherein at least one of R6 and R8 is
selected from alkyl, alkoxy, and chloride.
3. The use according to claim 1, wherein R6 and R8 are each hydrogen,
and W-(R7)pis C-(R7)1.
4. The use according to claim 1, wherein both R6 and R8 are not hydrogen.
5. The use according to claim 1, wherein X is CH;
R1 and R3 are alkoxy;
R6 and R8 are alkyl; and
R7 is hydroxyl.
6. The use according to claim 1, wherein X is N;
R1 and R3 are alkoxy;
R6 and R8 are alkyl; and

R7 is alkoxy substituted with a hydroxyl.
7. The use according to claim 1, wherein R7 is not diethylamino or an
alkoxy substituted with a carboxylate group.
8. The use according to claim 1, wherein R7 is selected from hydroxyl,
amino, and alkoxy.
9. The use according to claim 1, wherein the compound is selected from 3-
(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one, 7-(4-
hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-5(6H)-one, and 2-(4-
(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one, or a
pharmaceutically acceptable salt or hydrate thereof.
10. Use of a therapeutically effective amount of a compound in the
preparation of a medicament for increasing ApoA-l in a mammal, wherein the
compound is selected from:
3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one;
3-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-
1(2H)-one;
3-(4-hydroxy-3,5-dimethylphenyl)-7-(morpholinomethyl)isoquinolin-1(2H)-
one;
2-(4-hydroxy-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one;
3-(4-(2-hydroxy-2-methylpropoxy)-3,5-dimethylphenyl)-6,8-
dimethoxyisoquinolin-1 (2H)-one;

7-(4-hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-5(6H)-
one;
2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquina2olin-
4(3H)-one;
3-(3,5-dimethyl-4-(2-(4-methylpiperazin-1-yl)ethoxy)phenyl)-6,8-
dimethoxyisoquinolin-1 (2H)-one;
2-(4-hydroxy-3-methoxyphenyl)-5,7-dimethoxyquinazolin-4(3H)-one;
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-5,7-dimethoxyquinazolin-4(3H)-
one;
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-6,7-dimethoxyquinazolin-4(3H)-
one; '
2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxyquinazolin-4(3H)-one;
2-(4-((4-ethylpiperazin-1-yl)methyl)phenyl)-5,7-dimethoxyquinazolin-4(3H)-
one;
2-(4-(2-hydroxyethoxy)-3,5-dinnethylphenyl)-5,7-dimethoxypyrido[2,3-
d]pyrimidin-4(3H)-one;
2-(2-chloro-6-methylpyridin-4-yl)-5,7-dimethoxyquinazolin-4(3H)-one;
5,7-dimethoxy-2-(4-methoxy-3,5-dimethylphenyl)quinazolin-4(3H)-one;
2-(4-amino-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one;
N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide;
2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-
one;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzenesulfonamide;

4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihyclroquina2olin-2-yl)-
2,6-dimethylphenoxy)ethyl)benzenesulfonamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)methanesulfonamide;
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl propylcarbamate;
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl methylcarbamate;
N-(2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzamide;
2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl cyclohexylcarbamate;

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzenesulfonamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-

dimethylphenoxy)ethyl)-4-methoxybenzamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)acetamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yi)-2,6-
dimethylphenoxy)ethyl)benzamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)isobutyramide;

1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-methylurea;
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-(4-methoxyphenyl)urea;
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquina2olin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-phenylurea; and
3-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-1,1-dimethylurea,
or a pharmaceutically acceptable salt or hydrate thereof.
11. The use according to claim 1, wherein the therapeutically effective
amount of the compound of Formula II is administered with a pharmaceutically
acceptable carrier in a pharmaceutically acceptable composition.
12. The use according to claim 1, further comprising treating or preventing
a cardiovascular, cholesterol or lipid related disorder.
13. A compound of Formula II:

wherein:

X is selected from N and CH;
R1 and R3 are each independently selected from alkoxy and hydrogen;
R2 is selected from alkoxy, alkyl, and hydrogen;
R6 and R8 are each independently selected from alkyl, alkoxy, chloride, and
hydrogen;
R5 is hydrogen;
R7 is selected from amino, hydroxyl, alkoxy, and alkyl substituted with a
heterocyclyl, or
two adjacent substituents selected from R6, R7, and R8 are connected to
form a heterocyclyl;
each W is independently selected from C and N, wherein if Wis N, then p
is 0 or 1, and if W is C, then p is 1;
for W-(R4)P, W is C, p is 1 and R4 is H, or W is N and p is 0;
with the proviso that if R2 is selected from alkoxy or hydrogen, then at least
one of R1 and R3 is alkoxy;
with the proviso that if R7 is selected from hydroxyl or alkoxy, then at least
one of R6 and R8 are independently selected from alkyl, alkoxy, and chloride;
with the proviso that if R7 is amino, then X is N;
with the proviso that if for W-(R7)P, W is N and p is 0, then at least one of R6
and R8 is chloride;
and pharmaceutically acceptable salts and hydrates thereof.
14. The compound according to claim 13, wherein the compound is
selected from 3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1 (2H)-
one, 7-(4-hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-5(6H)-one,

and 2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-
one, and pharmaceutically acceptable salts or hydrates thereof.
15. A compound selected from:
3-(4-hydroxy-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-1(2H)-one;
3-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-6,8-dimethoxyisoquinolin-
1(2H)-one;
3-(4-hydroxy-3,5-dimethylphenyl)-7-(morpholinomethyl)isoquinolin-1(2H)-
one;
2-(4-hydroxy-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one;
7-(4-hydroxy-3,5-dimethylphenyl)-2,4-dimethoxy-1,6-naphthyridin-5(6H)-
one;

2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-
4(3H)-one;
3-(3,5-dimethyl-4-(2-(4-methylpiperazin-1-yl)ethoxy)phenyl)-6,8-
dimethoxyisoquinolin-1 (2H)-one;
2-(4-hydroxy-3-methoxyphenyl)-5,7-dimethoxyquinazolin-4(3H)-one;
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-5,7-dimethoxyquinazolin-4(3H)-
one;
2-(4-(bis(2-hydroxyethyl)amino)phenyl)-6,7-dimethoxyquinazolin-4(3H)-
one;
2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-6,7-dimethoxyquinazolin-4(3H)-one;
2-(4-((4-ethylpiperazin-1-yl)methyl)phenyl)-5,7-dimethoxyquinazolin-4(3H)-
one;

2-(4-(2-hydroxyethoxy)-3,5-dimethylphenyl)-5,7-dimethoxypyriclo[2,3-
d]pyrimidin-4(3H)-one;
2-(2-chloro-6-methylpyridin-4-yl)-5,7-dimethoxyquinazolin-4(3H)-one;
5,7-dimethoxy-2-(4-methoxy-3,5-dimethylphenyl)quinazolin-4(3H)-one;
2-(4-amino-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one;
N1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-N2-methylphthalamide;
2-(4-(2-aminoethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-
one;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzenesulfonamide;
4-chloro-N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-
2,6-dimethylphenoxy)ethyl)benzenesulfonamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)methanesulfonamide;
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl propylcarbamate;
2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-dimethyl-
phenoxy)ethyl methylcarbamate;
N-(2-(4-(5,7-Dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzamide;
2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl cyclohexylcarbamate;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzenesulfonamide;

N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methylbenzenesulfonamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-4-methoxybenzamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)acetamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)benzamide;
N-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)isobutyramide;
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-methylurea;
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-3-(4-methoxyphenyl)urea;
1-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethyIphenoxy)ethyl)-3-phenylurea; and
3-(2-(4-(5,7-dimethoxy-4-oxo-3,4-dihydroquinazolin-2-yl)-2,6-
dimethylphenoxy)ethyl)-1,1 -dimethylurea, and
pharmaceutically acceptable salts and hydrates thereof.
16. A pharmaceutical composition comprising a compound according to
any one of claims 13 to 15 and a pharmaceutically acceptable carrier.

17. Use of a therapeutically effective amount of a compound according to
any one of claims 13 to 15 in the preparation of a medicament for treating
cardiovascular, cholesterol or lipid related disorders.
18. (Currently Amended) Use of a therapeutically effective amount of a
compound according to any one of claims 13 to 15 in the preparation of a
medicament for increasing expression of ApoA-l in a mammal.

The present disclosure relates to compounds, which are useful for regulating the expression of apolipoproteiπ A-I
(ApoA-I), and their use for the treatment and prevention of cardiovascular disease and related disease states, including cholesterol-
or lipid-related disorders, such as, for example, atherosclerosis.

Documents:

http://ipindiaonline.gov.in/patentsearch/GrantedSearch/viewdoc.aspx?id=RoLC9Ty2f1sqwn43o/GPkw==&loc=wDBSZCsAt7zoiVrqcFJsRw==


Patent Number 277244
Indian Patent Application Number 2877/KOLNP/2009
PG Journal Number 48/2016
Publication Date 18-Nov-2016
Grant Date 16-Nov-2016
Date of Filing 10-Aug-2009
Name of Patentee RESVERLOGIX CORP.
Applicant Address 300, 4820 RICHARD ROAD SW, CALGARY ALBERTA T3E 6L1, CANADA
Inventors:
# Inventor's Name Inventor's Address
1 HANSEN, HENRIK 4903 CARNEY ROAD NW, CALGARY, ALBERTA T2L 1E6
PCT International Classification Number A61P 9/00,A61P 3/06
PCT International Application Number PCT/CA2007/000146
PCT International Filing date 2007-02-01
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 NA