Title of Invention

TETRAZOLE DERIVATIVES

Abstract The present invention relates to certain tetrazole derivatives of Formula (I), and pharmaceutically acceptable salts thereof, which exhibit useful pharmacological properties, for example, as agonists for the RUP25 receptor. Also provided by the present invention are pharmaceutical compositions containing compounds of the invention, and methods of using the compounds and compositions of the invention in the treatment of metabolic-related disorders, including dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, type 2 diabetes, Syndrome-X and the like. In addition, the present invention also provides for the use of the compounds of the invention in combination with other active agents such as those belonging to the class of &agr;- glucosidase inhibitors, aldose reductase inhibitors, biguanides, HMG-CoA reductase inhibitors, squalene synthesis inhibitors, fibrates, LDL catabolism enhancers, angiotensin converting enzyme (ACE) inhibitors, insulin secretion enhancers and the like.
Full Text


FIELD OF THE INVENTION
The present invention relates to certain tetrazole derivatives, and pharmaceutically
acceptable salts thereof, which exhibit useful pharmacological properties, for example as agonists
for the nicotinic acid receptor, RUP25. Also provided by the present invention are
pharmaceutical compositions containing one or more compounds of the invention, and methods
of using the compounds and compositions of the invention in the treatment of metabolic-related
disorders, including dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, type
2 diabetes, Syndrome-X and the like. In addition, the present invention also provides for the use
of the compounds of the invention in combination with other active agents such as those
belonging to the class of α-glucosidase inhibitors, aldose reductase inhibitors, biguanides, HMG-
CoA reductase inhibitors, squalene synthesis inhibitors, fibrates, LDL catabolism enhancers,
angiotensin converting enzyme (ACE) inhibitors, insulin secretion enhancers, thiazolidinedione
and the like.
BACKGROUND OF THE INVENTION
Compounds of the invention as Antilipolytic Agents
Atherosclerosis and stroke are the numbers one and number three leading causes of
death of both men and women in the United States. Type 2 diabetes is a public health problem
that is serious, widespread and increasing. Elevated levels of low density lipoprotein (LDL)
cholesterol or low levels of high density lipoprotein (HDL) cholesterol are, independently, risk
factors for atherosclerosis and associated cardiovascular pathologies. In addition, high levels of
plasma free fatty acids are associated with insulin resistance and type 2 diabetes. One strategy
for decreasing LDL-cholesterol, increasing HDL-cholesterol, and decreasing plasma free fatty
acids is to inhibit lipolysis in adipose tissue. This approach involves regulation of hormone
sensitive lipase, which is the rate-limiting enzyme in lipolysis. Lipolytic agents increase cellular
levels of cAMP, which leads to activation of hormone sensitive lipase within adipocytes. Agents
that lower intracellular cAMP levels, by contrast, would be antilipolytic.
It is also worth noting in passing that an increase in cellular levels of cAMP down-
regulates the secretion of adiponectin from adipocytes [Delporte, ML et al. Biochem ./(2002)
July], Reduced levels of plasma adiponectin have been associated with metabolic-related
disorders, including atherosclerosis, coronary heart disease, insulin resistance and type 2 diabetes
[Matsuda, M et al. J Biol Chem (2002) July and reviewed therein]


Nicotinic acid (niacin, pyridine-3-carboxylic acid) is a water-soluble vitamin required by
the human body for health, growth and reproduction; a part of the Vitamin B complex. Nicotinic
acid is also one of the oldest used drugs for the treatment of dyslipidemia. It is a valuable drug in
that it favorably affects virtually all ofthe lipid parameters listed above [Goodman and Gilman's
Pharmacological Basis of Therapeutics, editors Harmon JG and Limbird IE, Chapter 36, Mahley
RW and Bersot TP (2001) pages 971-1002]. The benefits of nicotinic acid in the treatment or
prevention of atherosclerotic cardiovascular disease have been documented in six major clinical
trials [Guyton JR (1998) Am J Cardiol 82:18U-23U], Nicotinic acid and related derivatives,
such as, acipimox have recently been discussed [Lorenzen, A et al (2001) Molecular
Pharmacology 59:349-357], Structure and synthesis of additional analogs or derivatives of
nicotinic acid are discussed throughout the Merck Index, An Encyclopedia of Chemicals, Drugs,
and Biologicals, Tenth Edition (1983), which is incorporated herein by reference in its entirety.
Nicotinic acid inhibits the production and release of free fatty acids from adipose tissue,
likely via an inhibition of adenylyl cyclase, a decrease in intracellular cAMP levels, and a
concomitant decrease in hormone sensitive lipase activity. Agonists that down-regulate honnone
sensitive lipase activity leading to a decrease in plasma free fatty acid levels are likely to have
therapeutic value. The consequence of decreasing plasma free fatty acids is two-fold. First, it
will ultimately lower LDL-cholesterol and raise HDL-cholesterol levels, independent risk
factors, thereby reducing the risk of mortality due to cardiovascular incidence subsequent to
atheroma formation. Second, it will provide an increase in insulin sensitivity in individuals with
insulin resistance or type 2 diabetes. Unfortunately, the use of nicotinic acid as a therapeutic is
partially limited by a number of associated, adverse side-effects. These include flushing, free
fatty acid rebound, and liver toxicity.
The rational development of novel, nicotinic acid receptor agonists that have fewer side-
effects will be valuable, but to date this has been hindered by the inability to molecularly identify
the nicotinic acid receptor. Furthermore, other receptors of the same class may exist on the
surface of adipocytes and similarly decrease hormone sensitive lipase activity through a
reduction in the level of intracellular cAMP but without the elicitation of adverse effects such as
flushing, thereby representing promising novel therapeutic targets. Recent work suggests that
nicotinic acid probably acts through a specific GPCR [Lorenzen A, et al. (2001) Molecular
Pharmacology 59:349-357 and reviewed therein]. Further work has suggested that the effects of
nicotinic acid on macrophages, spleen and probably adipocytes are mediated via this specific
GPCR [Lorenzen A, et al. (2002) Biochemical Pharmacology 64:645-648 and reviewed therein].
SUMMARY OF THE INVENTION
One aspect of the present invention encompasses tetrazole derivatives as shown in
Formula (1):



wherein:
X is N H or O;
R1 is selected from the group consisting of H, halogen, hydroxy, thioxy, cyano, nitro, C|.
4 haloalkyl, amino, C1-4 alkylamino, C2.8 dialkylamino, CM alky], CM alkoxy, C2J( alkenyl, C24
alkynyl, C35 cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio, C1-4 alkylsulfinyl, C1-4 alkylsulfonyl, C1-4 haloalkylthio, C1-4 haloalkylsulfinyl and C1-4 haloalkylsulfonyl;
R2 is selected from the group consisting of H, halogen, hydroxy, thioxy, cyano, nitro, C,.
4 haloalkyl, amino, C1-4 alkylamino, C2.8 dialkylamino, CM alky], CM alkoxy, C2.4 alkenyl, C2.4
alkynyl, C3.5 cycloalkyl, C1-4 haloalkoxy, C1-4 ALKYLthio, C1-4 ALKYLsulfinyl, C1-4 alkylsulfonyl, C1-4 HALOAlkylthio, C1-4 haloalkylsulfinyl and C1-4 haloalkylsulfonyl; or R2 is absent;
— is a single bond when R2 is present, or — is a double bond when R2 is absent; and
Ring A is a 5,6 or 7-membered carbocyclic ring or a 5, 6 or 7-membered heterocyclic
ring optionally substituted with 1 to 4 substituents selected from the group consisting of halogen,
hydroxy, thioxy, cyano, nitro, C1-4 HALOAlkyl, amino, C1-4 ALKYLamino, C2.s dialkylamino, C1-4 alkyl, CM alkoxy, CM alkenyl, CM alkynyl, C3.5 cycloalkyl, C1-4 haloalkoxy, C,.4 alkylthio, C1-4 alkylsulfinyl, C1-4 alkylsulfonyl, C1-4 haloalkylthio, C1-4 haloalkylsulfinyl and C1-4 haloalkylsulfonyl; or
a pharmaceutical^ acceptable salt, solvate or hydrate thereof.
One aspect of the present invention encompasses pharmaceutical compositions
comprising at least one compound according to Formula (I), as described herein.
In some embodiments, the pharmaceutical composition further comprises one or more
agents selected from the group consisting of a-glucosidase inhibitor, aldose reductase inhibitor,
biguanide, HMG-CoA reductase inhibitor, squalene synthesis inhibitor, fibrate, LDL catabolism
enhancer, angiotensin converting enzyme inhibitor, insulin secretion enhancer and
thiazolidinedione.
One aspect of the present invention pertains to methods of treatment of a metabolic-
related disorder comprising administering to an individual in need of such treatment a
therapeutically-effective amount of a compound according to Formula (I), as described herein,
or a pharmaceutical composition thereof.

One aspect of the present invention pertains to methods of modulating a RUP25 receptor
comprising contacting the receptor with a compound according to Formula (J), as described
herein.
One aspect of the present invention pertains to methods of modulating a RUP25 receptor
for the treatment of a metabolic-related disorder in an individual in need of such modulation
comprising contacting said receptor with a therapeutically-effective amount of a compound
according to Formula (1), as described herein.
One aspect of the present invention pertains to methods of raising HDL in an individual
comprising administering to the individual a therapeutically-effective amount of a compound
according to Formula (I), as described herein.
One aspect of the present invention pertains to a compound of Formula (I), as described
herein, for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to a compound of Formula (I), as described
herein, for use in a method of treatment of a metabolic-related disorder of the human or animal
body by therapy.
One aspect of the present invention pertains to the use of compounds of Formula (I), as
described herein, for the manufacture of a medicament for use in the treatment of a metabolic-
related disorder.
In some embodiments of the present invention, the metabolic-related disorder is of the
group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance,
obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X,
heart disease and type 2 diabetes. In some embodiments the metabolic-related disorder is
dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance and type 2 diabetes. In
some embodiments the metabolic-related disorder is dyslipidemia. In some embodiments the
metabolic-related disorder is atherosclerosis. In some embodiments the metabolic-related
disorder is coronary heart disease. In some embodiments the metabolic-related disorder is insulin
resistance. In some embodiments the metabolic-related disorder is type 2 diabetes.
One aspect of the present invention encompasses a method of producing a
pharmaceutical composition comprising admixing at least one compound according to Formula
(I), as described herein, and a pharmaceutically acceptable carrier or excipient.
These and other aspects of the invention disclosed herein will be set forth in greater
detail as the patent disclosure proceeds.
DETAILED DESCRIPTION OF THE INVENTION
The scientific literature has adopted a number of terms, for consistency and clarity, the
following definitions will be used throughout this patent document.


AGONISTS shall mean moieties that interact and activate the receptor, such as the
RUP25 receptor and initiates a physiologica' or pharmacological response characteristic of that
receptor. For example, when moieties activate the intracellular response upon binding to the
receptor, or enhance GTP binding to membranes.

The term ANTAGONISTS is intended to mean moieties that competitively bind to the
receptor at the same site as agonists (for example, the endogenous ligand), but which do not
activate the intracellular response initiated by the active form of the receptor, and can thereby
inhibit the intracellular responses by agonists or partial agonists. Antagonists do not diminish the
baseline intracellular response in the absence of an agonist or partial agonist.
ATHEROSCLEROSIS is intended herein to encompass disorders of large and
medium-sized arteries that result in the progressive accumulation within the intima of smooth
muscle cells and lipids.
CHEMICAL GROUP, MOIETY OR RADICAL:

The term "CM acyl" denotes a C| A alkyl radical attached to a carbonyl wherein
the definition of alkyl has the same definition as described herein; some examples
include but not limited to, acetyl, propionyl, n-butanoyl, wo-butanoyl, sec-butanoyl, t-
butanoyl (i.e., pivaloyl), pentanoyl and the like.
The term "CM acyloxy" denotes an acyl radical attached to an oxygen atom
wherein acyl has the same definition has described herein; some examples include but
not limited to acetyloxy, propionyloxy, butanoyloxy, wo-butanoyloxy, sec-butanoyloxy,
t-butanoyloxy and the like.
The term "CM alkenyl" denotes a radical containing 2 to 4 carbons wherein at
least one carbon-carbon double bond is present, some embodiments are 2 to 3 carbons,
and some embodiments have 2 carbons. Both E and Z isomers are embraced by the term
"alkenyl." Furthermore, the term "alkenyl" includes di-enes. Accordingly, if more than
one double bond is present, then the bonds may be all £orZor a mixtures ofEandZ.
Examples of an alkenyl include vinyl, propenyl, allyl, isopropenyl, 2-methyl-propenyl I -
methyl-propenyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-l,3-dienyl, and the like.
The term "CM alkoxy" denotes an alkyl radical, as defined herein, attached
directly to an oxygen atom. Examples include methoxy, ethoxy, n-propoxy, iso-
propoxy, n-butoxy, t-butoxy, iso-butoxy, sec-butoxy and the like.
The term "C1-4 alkyl" denotes a straight or branched carbon radical containing
the number of carbons as indicated, for examples^ in some embodiments, alkyl is a "C1-4 alkyl" and the group contains 1 to 4 carbons. In some embodiments alkyl contains 1 to
13 carbons, some embodiments contain 1 to 2 carbons, some embodiments contain 1
carbon. Examples of an alkyl include, but not limited to, methyl, ethyl, n-propyl, iso-
propyl, n-butyl, iso-butyl, t-butyl, sec-butyl, and the like.
The term "C1-4 alkylsulfinyl" denotes a C1-4 ALKYL radical attached to a sulfoxide
radical of the formula: -S(O)- wherein the alkyl radical has the same definition as
described herein. Examples include, but not limited to, methylsulfinyl, ethylsulfinyl, n-
propylsulfinyl, iso-propylsulfinyl, n-butylsulfinyl, sec-butylsulfinyl, iso-butylsulfinyl, t-
butyl, and the like.
The term "C1-4 ALKYLsulfonyl" denotes a C1-4 alkyl radical attached to a sulfone
radical of the formula: -S(0)2- wherein the alkyl radical has the same definition as
described herein. Examples include, but not limited to, methylsulfonyl, ethylsulfonyl, n-
propylsulfonyl, iso-propylsulfonyl, n-butylsulfonyl, sec-butylsulfonyl, iso-butylsulfonyl.
t-butylsulfonyl, and the like,
The term "C1-4 alkylthio" denotes a C1-4 alkyl radical attached to a sulfide group
of the formula: -S- wherein the alkyl radical has the same definition as described herein.
Examples include, but not limited to, methylsulfanyl (i.e., CH3S-), ethylsulfanyl, n-


propylsulfanyl, iso-propylsulfanyl, n-butylsulfanyl, sec-butylsulfanyl, iso-butylsulfanyl,
t-butyl, and the like.
The term "C2Jt alkynyl" denotes a radical containing 2 to 4 carbons and at least
one carbon-carbon triple bond, some embodiments are 2 to 3 carbons, and some
embodiments have 2 carbons. Examples of an alkynyl include, but not limited to,
ethynyl, prop-1-ynyl, 3-prop-2-ynyl, but-1-ynyl, 1 -methyl-prop-2-ynyl, buta-l,3-diynyl,
and the like. The term "alkynyl" includes di-ynes.
The term "amino" denotes the group -NH2.
The term "C1-4 alkylamino" denotes one alkyl radical attached to an amino
radical wherein the alkyl radical has the same meaning as described herein. Some
examples include, but not limited to, methylamino, ethylamino, n-propylamino, iso-
propylamino, n-butylamino, sec-butylamino, iso-butylamino, t-butylamino, and the like.
Some embodiments are "C|.2 alkylamino."
The term "aryl" denotes an aromatic ring radical containing 6 to 10 ring
carbons. Examples include phenyl and naphthyl.
The term "carbo-CM-alkoxy" denotes a C1-4 alkyl ester of a carboxylic acid,
wherein the alkyl group is as defined herein. Examples include, but not limited to,
carbomethoxy, carboethoxy, carbopropoxy, carboisopropoxy, carbobutoxy, carbo-sec-
butoxy, carbo-iso-butoxy, carbo-t-butoxy, and the like.
The term "carboxamide" refers to the group -CONH2.
The term "carboxy" or "carboxyl" denotes the group -C02H; also referred to
as a carboxylic acid group.
The lerm "cyano" denotes the group -CN.
The term "Cj.s cycloalkyl" denotes a saturated ring radical containing 3 to 6
carbons; some embodiments contain 3 to 5 carbons; some embodiments contain 3 to 4
carbons. Examples include, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
The term "C2.8 dialkylamino" denotes an amino substituted with two of the
same or different alkyl radicals wherein alkyl radical has the same definition as
described herein. A C2.8 dialkylamino may be represented by the following groups:

Examples of C2.8 dialkylamino include, but not limited to, dimethylamino,
methylethylamino, diethylamino, methylpropylamino, methylisopropylamino, and the
like.
The term "C1-4 haloalkoxy" denotes a haloalkyl, as defined herein, which is
directly attached to an oxygen atom. Examples include, but not limited to,

difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, pentafluoroethoxy and the
like.
The term "C1-4 HALOAlkyi" denotes an alkyl group wherein the alkyl is
substituted with halogen ranging from one to fully substituted, wherein a fully
substituted haloalkyi can be represented by the formula ChL2h»i wherein L is a halogen
and "h" represents the number of carbon atoms; when more than one halogen is present
then the halogens may be the same or different and selected from the group consisting of
F, CI, Br and I; it is understood that the terms "alkyl" and "halogen" have the same
definition as found herein. In some embodiments, haloalkyi is a "C1-4 haloalkyi" and
the group contains 1 to 4 carbons, some embodiments contain 1 to 3 carbons, some
embodiments contain I to 2 carbons, some embodiments contain 1 carbon. When the
haloalkyi is fully substituted with halogen atoms, this group is referred herein as a
perhaloalkyl, one example, is an alkyl fully substituted with fluorine atoms and is
referred to herein as a "perfluoroalkyl." In some embodiments, examples of a haloalkyi
include, but not limited to, difluoromethyl, fluoromethyl, 2,2,2-trifluoro-ethyl, 2,2-
difluoro-ethyl, 2-fluoro-ethyl, 1,2,2-trifluoro-ethyl, 1,2-difluoro-ethyl, 1,1-difluoro-ethyl,
1,1,2-trifluoro-ethyl, 3,3,3-trifluoro-propyl, 2,2-difluoro-propyl, 3,3-difluoro-propyl, 3-
fluoro-propyl, 2,3,3-trifluoro-propyl, 2,3-Difluoro-propyl, 2,2,3,3,3-pentafluoro-propyl,
2,2,3,3-tetrafluoro-propyl, 2,2,3-trifluoro-propyl, 1,2,3,3-tetrafluoro-propyl, 1,2,3-
trifluoro-propyl, 3,3-difluoro-propyl, 1,2,2,3-tetrafluoro-propyl, 4,4-difluoro-butyl, 3,3-
difluoro-butyl, 4,4,4-trifluoro-butyl, 3,3-difluoro-butyl, and the like. In some
embodiments, examples of a perfluoroalkyl include, but not limited to, trifluoromethyl,
pentafluoroethyl, heptafluoropropyl, 1,2,2,2-tetrafluoro-l-trifluoromethyl-ethyl, and the
like.
The term "C1-4 haloalkylsulfinyl" denotes a haloalkyi radical attached to a
sulfoxide group of the formula: -S(O)- wherein the haloalkyi radical has the same
definition as described herein.
The term "C1-4 HALOAlkylsulfonyl" denotes a haloalkyi radical attached to a
sulfone group of the formula: -S(0)2- wherein haloalkyi has the same definition as
described herein.
The term "C1-4 haloalkylthio" denotes a haloalkyi radical directly attached to a
sulfur atom wherein the haloalkyi has the same meaning as described herein.
The term "halogen" or "halo" denotes to a fluoro, chloro, bromo or iodo group.
The term "hydroxyI" denotes the group -OH.
The term "nitro" denotes the group -N02.
The term "thioxy" denotes the group -SH.
The acronym DMF denotes dimethylformamide.


The acronym DMSO denotes dimethylsulfoxide.
The acronym THF denotes tetrahydrofuran.
The acronym DCM denotes dichloromethane.
The acronym Hex denotes hexanes.
The acronym TBDMS denotes tert-butyldimethylsilyl.
The acronym PTSA denotes para-tolucncsulfonic acid.
The acronym LDA denotes lithium diisopropylamide.
The acronym LHMDS denotes lithium hexamethyldisilazane.
The acronym TFA denotes trifluoroacetic acid.
The acronym EDC denotes l-ethyl-3-(3-dimethylaminopropyl)carbodiimide
hydrochloride.
The acronym dppf denotes 1,l'-bis(diphenylphosphino)ferrocene.
The term CODON shall mean a grouping of three nucleotides (or equivalents to
nucleotides) which generally comprise a nucleoside (adenosine (A), guanosine (G), cytidine (C),
uridine (U) and thymidine (T)) coupled to a phosphate group and which, when translated,
encodes an amino acid.
The term COMPOSITION shall mean a material comprising at least two compounds or
two components; for example, and without limitation, a Pharmaceutical Composition is a
Composition comprising a compound of the present invention and a pharmaceutically acceptable
carrier.
The term COMPOUND EFFICACY shall mean a measurement of the ability of a
compound to inhibit or stimulate receptor functionality, as opposed to receptor binding affinity.
The term CONSTITUTIVELY ACTIVATED RECEPTOR shall mean a receptor
subject to constitutive receptor activation.
The term CONSTITUTIVE RECEPTOR ACTIVATION shall mean stabilization of
a receptor in the active state by means other than binding of the receptor with its endogenous
ligand or a chemical equivalent thereof.
The terms CONTACT or CONTACTING shall mean bringing the indicated moieties
together, whether in an in vitro system or an in vivo system. Thus, "contacting" a RUP25
receptor with a compound of the invention includes the administration of a compound of the
present invention to an individual, for example a human, having a RUP25 receptor, as well as,
for example, introducing a compound of the invention into a sample containing a cellular or more
purified preparation containing a RUP25 receptor.
CORONARY HEART DISEASE is intended herein to encompass disorders
comprising a narrowing of the small blood vessels that supply blood and oxygen to the heart.
Coronary heart disease usually results from the build up of fatty material and plaque. As the


coronary arteries narrow, the flow of blood to the heart can slow or stop. Coronary heart disease
can cause chest pain (stable angina), shortness of breath, heart attack, or other symptoms.
DECREASE is used to refer to a reduction in a measurable quantity and is used
synonymously with the terms "reduce", "diminish", "lower", and "lessen".
DIABETES as used herein is intended to encompass the usual diagnosis of DIABETES
made from any of the methods including, but not limited to, the following list: symptoms of
diabetes (e.g., polyuria, polydipsia, polyphagia) plus casual plasma glucose levels of greater than
or equal to 200 mg/dl, wherein casual plasma glucose is defined any time of the day regardless of
the timing of meal or drink consumption; 8 hour fasting plasma glucose levels of less than or
equal to 126 mg/dl; and plasma glucose levels of greater than or equal to 200 mg/dl 2 hours
following oral administration of 75 g anhydrous glucose dissolved in water.
The phrase DISORDERS OF LIPID METABOLISM is intended herein to include,
but not be limited to, dyslipidemia.
The term DYSLIPIDEMIA is intended herein to encompass disorders comprising any
one of elevated level of plasma free fatty acids, elevated level of plasma cholesterol, elevated
level of LDL-cholesterol, reduced level of HDL-cholesterol, and elevated level of plasma
triglycerides.
The phrase IN NEED OF TREATMEINT, as used herein, refers to a judgment made by
a caregiver (e.g. physician, nurse, nurse practitioner, etc. in the case of humans; veterinarian in
the case of animals, including non-human mammals) that an individual or animal requires or will
benefit from treatment. This judgment is made based on a variety of factors that are in the realm
of a caregiver's expertise, that includes the knowledge that the individual is ill, or will be ill, as
the result of a disease, condition or disorder that is treatable by the compounds of the invention.
Further, the phrase "'in need of treatment" also refers to the "prophylaxis" of an individual which
is the judgment made by the caregiver that the individual will become ill. In this context, the
compounds of the invention are used in a protective or preventive manner. Accordingly, "in
need of treatment" refers to the judgment of the caregiver that the individual is already ill or will
become ill and the compounds of the present invention can be used to alleviate, inhibit,
ameliorate or prevent the disease, condition or disorder.
The term INDIVIDUAL as used herein refers to any animal, including mammals, for
example, mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates,
and in one embodiment, humans.
The terms INHIBIT or INHIBITING, in relationship to the term "response" shall mean
that a response is decreased or prevented in the presence of a compound as opposed to in the
absence of the compound.
INSULIN RESISTANCE as used herein is intended to encompass the usual diagnosis
of insulin resistance made by any of a number of methods, including but not restricted to: the


intravenous glucose tolerance test or measurement of the fasting insulin level. It is well known
that there is an excellent correlation between the height of the fasting insulin level and the degree
of insulin resistance. Therefore, one could use elevated fasting insulin levels as a surrogate
marker for insulin resistance for the purpose of identifying which normal glucose tolerance
(NGT) individuals have insulin resistance. A diagnosis of insulin resistance can also be made
using the euglycemic glucose clamp test.
The term INVERSE AGONISTS shall mean moieties that bind the endogenous form of
the receptor or to the constitutively activated form of the receptor, and which inhibit the baseline
intracellular response initiated by the active form of the receptor below the normal base level of
activity which is observed in the absence of agonists or partial agonists, or decrease GTP binding
to membranes. In some embodiments, the baseline intracellular response is inhibited in the
presence of the inverse agonist by at least 30%, in other embodiments, by at least 50%, and in
still other embodiments, by at least 75%, as compared with the baseline response in the absence
of the inverse agonist.
The term LICAND shall mean an endogenous, naturally occurring molecule specific for
an endogenous, naturally occurring receptor.
The phrase METABOLIC-RELATED DISORDERS is intended herein to include,
but not be limited to, dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance,
obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X,
heart disease and type 2 diabetes.
As used herein, the terms MODULATE or MODULATING shall mean to refer to an
increase or decrease in the amount, quality, response or effect of a particular activity, function or
molecule.
The term PHARMACEUTICAL COMPOSITION shall mean a composition for
preventing, treating or controlling a disease state or condition comprising at least one active
compound, for example, a compound of the present invention including pharmaceutical^
acceptable salts, pharmaceutical^ acceptable solvates and/or hydrates thereof, and at least one
pharmaceutical^ acceptable carrier.
The term PHARMACEUTICALLY ACCEPTABLE CARRIER or EXdPlENT
shall mean any substantially inert substance substance used as a diluent or vehicle for a
compound of the present invention.
The phrase THERAPEUTICALLY-EFFECT1VE AMOUNT as used herein refers to
the amount of active compound or pharmaceutical agent that elicits the biological or medicinal
response in a tissue, system, animal, individual or human that is being sought by a researcher,
veterinarian, medical doctor or other clinician, which includes one or more of the following:


(1) Preventing the disease; for example, preventing a disease, condition or disorder in an
individual that may be predisposed to the disease, condition or disorder but does not yet
experience or display the pathology or symptomatology of the disease,
(2) Inhibiting the disease; for example, inhibiting a disease, condition or disorder in an
individual that is experiencing or displaying the pathology or symptomatology of the disease,
condition or disorder (i.e., arresting further development of the pathology and/or
symptomatology), and
(3) Ameliorating the disease; for example, ameliorating a disease, condition or disorder
in an individual that is experiencing or displaying the pathology or symptomatology of the
disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
COMPOUNDS OF THE INVENTION
One aspect of the present invention encompasses tetrazole derivatives as shown in
Formula (I):

wherein:
X isNHorO;
R, is selected from the group consisting of H, halogen, hydroxy, thioxy, cyano,
nitro, C1-4 haloalkyl, amino, C1-4 alkylamino, C2.8 dialkylamino, C1-4 ALKYL, CM alkoxy,
C2.4 alkenyl, C2J) alkynyl, C3.5 cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio, C1-4 alkylsulfinyl, C1-4 alkylsulfonyl, C1-4 haloalkylthio, C1-4 haloalkylsulfinyl and C1-4 HALOAlkylsulfonyl;
R2 is selected from the group consisting of H, halogen, hydroxy, thioxy, cyano,
nitro, C1-4 haloalkyl, amino, C1-4 alkylamino, C2.8 dialkylamino, C1-4 alkyl, CM alkoxy,
C1A alkenyl, CM alkynyl, C3.5 cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio, C1-4 alkylsulfinyl, C1-4 alkylsulfonyl, C1-4 HALOAlkylthio, C1-4 HALOAlkylsulfinyl and C1-4 HALOAlkylsulfonyl; or R2 is absent;
— is a single bond when R2 is present, or — is a double bond when R2 is
absent; and
Ring A is a 5, 6 or 7-membered carbocyclic ring or a 5, 6 or 7-membered
heterocyclic ring optionally substituted with 1 to 4 substituents selected from the group
consisting of halogen, hydroxy, thioxy, cyano, nitro, C1-4 haloalkyl, amino, CM

alkylamino, C;.» dialkylamino, C1-4 alkyl, CM alkoxy, C2.4 alkenyl, C2.4 alkynyl, C^
cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio, C1-4 alkylsulfinyl, Ct 4 alkylsulfonyl, C1-4 haloalkylthio, C1-4 haloalkylsulflnyl and C1-4 haloalkylsulfonyl; or
a pharmaceutically acceptable salt, solvate or hydrate thereof.
Compounds of the present invention may exist in various tautomeric forms. For
example, it is well appreciated to those of skill in the art that tetrazoles can exist in at least two
tautomeric forms and although Formula (I) represents one form it is understood that all
tautomeric forms are embraced by the present invention; by way of illustration, two possible
tautomers for the tetrazole in Formula (1) are shown below:

Another example includes embodiments wherein X is Nl I. It is well appreciated to
those of skill in the art that pyrazole heterocycles can exist in at least two tautomeric forms and
although Formula (I) represents one form it is understood that all tautomeric forms are embraced
by the present invention; by way of illustration, two possible tautomers for the pyrazole wherein
X is NH in Formula (I) are shown below:

Further, it is understood that when X is NH then tautomers can exist for both Ring B and also the
tetrazole ring in combination. It is understood that all tautomers that can exist for the compounds
disclosed herein are within the scope of the invention.
The present invention also encompasses diastereomers as well as optical isomers, e.g.
mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and
diastereomers, which arise as a consequence of structural asymmetry in certain compounds of the
present invention In some embodiments, compounds of the present invention are R. In some
embodiments, compounds of the present are S. In some embodiments, compounds of the present
invention are racemic mixtures.
It is appreciated that certain features of the invention, which are, for clarity, described in
the context of separate embodiments, may also be provided in combination in a single

embodiment. Conversely, various features of the invention which are, for brevity, described in
the context of a single embodiment, may also be provided separately or in any suitable
subcombination.
As used herein, "substituted" indicates that at least one hydrogen atom of the chemical
group is replaced by a non-hydrogen substituents or group. When a chemical group herein is
"substituted" it may have up to the full valance of substitution; for example, a methyl group can
be substituted by 1,2, or 3 substituents, a methylene group can be substituted by 1 or 2
substituents, a phenyl group can be substituted by 1, 2, 3, 4, or 5 substituents, and the like.
One embodiment of the present invention pertains to compounds of Formula (f) wherein
X is NH. This embodiment can be represented by Formula (la) as illustrated below:

wherein each variable in Formula (la) has the same meaning as described herein, supra and
infra.
One embodiment of the present invention pertains to compound of Formula (I) wherein
X is NH, R| is H or hydroxy; R2 is H or absent; — is a single bond when R? is H, or — is a
double bond when R2 is absent; and Ring A is a 5-membered carbocyclic ring or a 5-membered
heterocyclic ring optionally substituted with I to 4 substituents selected from the group
consisting of halogen, C1-4 alkyl, CM alkoxy and C3.5 cycloalkyl; or a pharmaceutical^
acceptable salt, solvate or hydrate thereof.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
X is O. This embodiment can be represented by Formula (Ic) as illustrated below:

wherein each variable in Formula (lc) has the same meaning as described herein, supra and infra.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ri is selected from the group consisting of H, halogen, hydroxy, C1-4 haloalkyl, C1-4 alkyl, CM
alkoxy, C24 alkenyl, C2^, alkynyl, C1-4 ALKYLthio, and C1-4 haloalkoxy. In some embodiments, R|

is selected from the group consisting of H, halogen, C1-4 haloalkyl, and C1-4 alkyl. In some
embodiments, R| is F. In some embodiments, Ri is H.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
R2 is selected from the group consisting of H, halogen, hydroxy, C1-4 haloalkyl, C1-4 alkyl, CM
alkoxy, CM alkenyl, C2J) alkynyl, C1-4 alkylthio, and C1-4 haloalkoxy. In some embodiments, R2
is selected from the group consisting of H, halogen, C1-4 HALOAlkyl, and C1-4 ALKYL. In some
embodiments, R2 is F. In some embodiments, R2 is H.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
R, and R2 are both H.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ring A is a 5, 6 or 7-membered carbocyclic ring. The term "5, 6 or 7-membered carbocyclic
ring" denotes a ring containing 5, 6 or 7 ring carbons wherein two ring carbons are shared by
rings A and B. Ring A can be saturated (i.e. no ring double bonds), unsaturated (i.e., containing
ring double bonds) or a combination thereof. In some embodiments, — is a single bond and R2
is present. This embodiment can be represented by Formula (le) as illustrated below:

wherein each variable in Formula (le) has the same meaning as described herein, supra and infra.
One embodiment of the present invention pertains to compounds having Formula (If):

wherein:
R, is H or hydroxy; and Ring A is optionally substituted with 1 or 2 subslituents selected
from the group consisting of halogen, C1-4 ALKYL, CM alkoxy and C3.5 cycloalkyl; or a
pharmaceutical^ acceptable salt, solvate or hydrate thereof.
In some embodiments, Ring A is a 5-membered carbocyclic ring. In one embodiment,
Ring A is a 5-membered carbocyclic ring and can be represented by Formula (Ig) as illustrated
below:


wherein each variable in Formula (Ig) has the same meaning as described herein, supra and
infra. In some embodiments, R| is CM alkoxy. In some embodiments, R| is C1-4 ALKYL. In some
embodiments, R, and R2 are both H.
One embodiment of the present invention pertains to compounds having Formula (lh):

wherein:
Ring A is optionally substituted with 1 or 2 substituents selected from the group
consisting of halogen, C1-4 alkyl, CM alkoxy and C3.5 cycloalkyl; or a pharmaceutically
acceptable salt, solvate or hydrate thereof.
One embodiment of the present invention pertains to compounds having Formula (lh):

wherein:
Ring A is substituted with 1 or 2 substituents selected from the group consisting of
halogen, ^-propyl, n-butyl, CM alkoxy and C3.5 cycloalkyl; or a pharmaceutical^ acceptable salt,
solvate or hydrate thereof.
One embodiment of the present invention pertains to compounds having Formula (lh):

wherein:

Ring A is unsubstituted or is substituted with ethyl; or a pharmaceutically acceptable
salt, solvate or hydrate thereof..
In one embodiment, Ring A is a 5-membered carbocyclic ring and is further unsaturated
(i.e., a ring double bond). This embodiment can be represented by Formula (li) as illustrated
below:

wherein each variable in Formula (li) has the same meaning as described herein, supra and infra.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ring A is a 6-mernbered carbocyclic ring. This embodiment can be represented by Formula (Ik)
as illustrated below:

wherein each variable in Formula (Ik) has the same meaning as described herein, supra and
infra.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ring A is a 7-membered carbocyclic ring. This embodiment can be represented by Formula
(Im) as illustrated below:

wherein each variable in Formula (Im) has the same meaning as described herein, supra and
infra.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ring A is a 5, 6 or 7-membered carbocyclic ring, as described herein supra. In some

embodiments, — is a double bond and R2 is absent. This embodiment can be represented by
Formula (lo) as illustrated below:

wherein each variable in Formula (lo) has the same meaning as described herein, supra and
infra. In some embodiments, Ring A is a 5-membered carbocyclic ring. This embodiment can
be represented by Formula (Iq) as illustrated below:

wherein each variable in Formula (Iq) has the same meaning as described herein, supra and
infra. In some embodiments, Ring A is a 6-mcmbered carbocyclic ring. In some embodiments,
Ring A is a 6-membered carbocyclic ring provided that Ring A is not aromatic. In some
embodiments, Ring A is a 7-membered carbocyclic ring.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ring A is a 5, 6 or 7-membered heterocyclic ring. The term "5, 6 or 7-membered heterocyclic
ring" denotes a 5, 6 or 7-membered carbocyclic ring, as described herein supra, wherein 1, 2 or 3
ring carbons not shared by Rings A and B are independently replaced with -0-, -S-, -S(O)-, or -
S(0)2-. For clarity, as described herein supra, Ring A can be saturated (i.e. no ring double
bonds), unsaturated (i.e., containing ring double bonds) or a combination thereof. In some
embodiments, — is a single bond and R2 is present. In some embodiments, Ring A is a 5-
membered heterocyclic ring. In some embodiments, one ring carbon of the 5-membered
heterocyclic ring is replaced with a ring oxygen atom; these embodiments can be represented by
the following Formulae (Is) and (It):


wherein each variable in Formulae (Is) and (It) have the same meaning as described herein,
supra and infra. In some embodiments, compounds of the present invention are of Formula (Is)
wherein X is NH. In some embodiments, compounds of the present invention are of Formula
(Is) wherein X is O (an oxygen atom). In some embodiments, compounds of the present
invention are of Formula (It) wherein X is NH. In some embodiments, compounds of the
present invention are of Formula (It) wherein X is O (an oxygen atom). In some embodiments,
compounds of the present invention are of Formula (Is) wherein R| is C1-4 alkyl and R2 is H. In
some embodiments, compounds of the present invention are of Formula (Is) wherein both R, and
R2 are H. In some embodiments, compounds of the present invention are of Formula (It)
wherein R, is C1-4 alkyl and R2 is H. In some embodiments, compounds of the present invention
are of Formula (It) wherein both R, and R2 are H. In some embodiments, one ring carbon of the
5-membered heterocyclic ring is replaced with a ring sulfur atom; these embodiments can be
represented by the following Formulae (lu) and (Iv):

wherein each variable in Formulae (lu) and (Iv) have the same meaning as described herein,
supra and infra. In some embodiments, the ring sulfur in Formulae (lu) and (Iv) is further
oxidized to a sulfoxide (i.e., -S(O)-). In some embodiments, the ring sulfur in Formulae (lu) and
(Iv) is further oxidized to a sulfone (i.e., -S(0)2-). In some embodiments, compounds of the
present invention are of Formula (lu) wherein X is NH. In some embodiments, compounds of
the present invention are of Formula (lu) wherein X is O (an oxygen atom). In some
embodiments, compounds of the present invention are of Formula (Iv) wherein X is NH. In
some embodiments, compounds of the present invention are of Formula (Iv) wherein X is 0 (an
oxygen atom). In some embodiments, Ring A is a 6-membered heterocyclic ring. In some
embodiments, one ring atom of the 6-membered heterocyclic ring is replaced by a ring oxygen
atom; these embodiments can be represented by the following Formulae (Ix), (ly), and (Iz):


wherein each variable in Formulae (Ix), (ly), and (Iz) have the same meaning as described
herein, supra and infra. In some embodiments, Ring A is a 7-membered heterocyclic ring.
One embodiment of the present invention pertains to compounds of Formula (I) wherein
Ring A is a 5, 6 or 7-membered heterocyclic ring. In some embodiments, ^^ is a double bond
and R? is absent. In some embodiments, Ring A is a 5-membered heterocyclic ring. This
embodiment can be represented by Formula (Ha) as illustrated below:

wherein each variable in Formula (Ha) has the same meaning as described herein, supra and
infra, and Z is -0-, -S-, -S(O)-, or -S(0)r.
In some embodiments, Ring A is a 6-membered heterocyclic ring. In some
embodiments, the 6-membered heterocyclic ring is a dihydro-pyranyl ring (i.e., one ring carbon
is replaced by an oxygen atom); these embodiments can be represented by the following
Formulae (lie) and (lid):

wherein each variable in Formulae (lie) and (lid) have the same meaning as described herein,
supra and infra. In some embodiments, Ring A is a 7-membered heterocyclic ring.
One embodiment of the present invention pertains to compounds of Formula (I), and
subgenera disclosed herein, wherein Ring A is optionally substituted with substituents selected
from the group consisting of halogen, hydroxy, C1-4 haloalkyl, C1-4 alkyl, CM alkoxy, C?^
alkenyl, C2^ alkynyl, C1-4 ALKYLthio, and C1-4 HALOAlkoxy. In some embodiments, Ring A is
optionally substituted with substituents selected from the group consisting of halogen, C1-4 haloalkyl, and C1-4 alkyl. In some embodiments, Ring A is optionally substituted with F. In
some embodiments, Ring A is optionally substituted with 1 to 4 substituents. In some
embodiments, Ring A is optionally substituted with 1 to 3 substituents. In some embodiments,
Ring A is optionally substituted with 1 to 2 substituents. In some embodiments, Ring A is
optionally substituted with I substituent. In some embodiments, Ring A is not substituted.

CHEMISTRY OF THE PRESENT INVENTION
Synthesis of Compounds of Formula (0
In one embodiment of the present invention is a synthetic process for the preparation of
novel tetrazoles of Formula (I). The compounds of the present invention can be readily prepared
according to this novel process utilizing a variety of starting materials that are commercially
available or readily prepared by synthetic regimes which would be familiar to one skilled in the
art. In the illustrated syntheses outlined below, unless stated otherwise, the labeled substituents
have the same identifications as set out in the definitions of the compound described above for
Formula (I).
One method that can be used to prepare compounds of the invention wherein X is NH
(i.e., Ring B is a pyrazole) utilizes intermediates derived from the cyclic ketone of Formula (A)
as illustrated in Reaction Scheme I below:

Compounds of Formula (la) can be prepared by reacting a cyclic ketone of Formula (A) with
dialkyloxalate of formula (C(O)OR|0)2, wherein R,0 is a C|.8 alkyl, in the presence of a base and a
polar solvent such as, but not limited to, Ci.g alkanol, methanol, ethanol, butanol, pentanol,
hexanol. 2-methoxyethanol, isopropanol, THF, DMF and the like to give ketoester of Formula
(B). Suitable bases include alkali metal alkoxides, for example, sodium methoxide, sodium
ethoxide, potassium ethoxide, potassium /-butoxide, and the like; alkali metal amides (i.e., alkali
metal-NRu wherein RM is C,.8 alkyl or silyl-Ci.8-alkyl), for example, lithium diisopropylamide,
lithium hexamethyldisilazane, sodium hexamethyldisilazane, potassium hexamethyldisilazane
and like bases. Ketoester (B) is reacted with hydrazine, either protected or unprotected hydrazine
can be used, under suitable conditions to give pyrazole ester of Formula (C). Optionally, the
pyrazole can be protected, for example, with a benzyl group and the like. The ester is converted
to amide of Formula (D) using methods known to one of skill in the art, for example, treating

with ammonia in a polar solvent at temperature from room temperature to the boiling point of the
solvent. Amide (D) is reacted with a dehydrating reagent, such as phosphorous oxychloride,
phosphorous pentoxide, thionyl chloride, and the like, either neat or in the presence of a
nonprotic solvent, such as acetonitrile, DMF, and the like, to give nitrile (E). Nitrile (E) is
reacted with an azide (i.e., N3) or azide equivalent, such as, sodium azide, potassium azide,
trimethylsilyl azide (i.e., (CH3)SiN3), and the like to give tetrazole of Formula (la). In some
instances it can be beneficial to include the presence of a Lewis acid, for example, A1C13, ZnBr2,
and the like, in a suitable solvent, such as, DMF and the like.
One method that can be used to prepare compounds of Formula (I) wherein X is a ring
oxygen (i.e., Ring B is a isoxazole) utilizes intermediates derived from the alkynyl alcohol of
Formula (J) as illustrated in Reaction Scheme II below;

Compounds of Formula (Ic) can be prepared by protecting an alkynyl alcohol of Formula (J)
with a suitable protecting group, for example, THP, TBDMS, and the like to give alkynyl (K).
Alkynyl (K) is converted to an alkynyl ester of Formula (L, wherein Rl5 is C|.a alkyl) by
treatment with a strong base followed by reacting with a C,.8 alkyl chloroformate. A suitable
strong base is an alkyl lithium, for example but not limited to, «-butyl lithium, /-butyl lithium and
the like Intermediate (L) is subsequently deprotected using methods known to those of skill in
the art, for example, the THP group can typically be removed via treatment with an acid (e.g.
PTSA) and TBDMS group can typically be removed via treatment with a tetra-alkylammonium
fluoride. The resulting alcohol is oxidized to aldehyde (M) using any variety of methods, for
example, Dess-Martin periodinane(i.e., l,l,l-triacetoxy-l,l-dihydro-l,2-benziodoxol-3(lH)-
one), Swern Oxidation, Corey oxidation with NCS or any other suitable method as described by
Hudlicky, M. in Oxidations in Organic Chemistry, ACS Monograph 186 (1990). Aldehyde (1M)
is treated with hydroxylamine in the presence of a base, followed by NCS and Base to give

isoxazole alkylester (N). lsoxazole (N) can be converted to compounds of Formula (Ic) in a
substantially similar manner as described above in Reaction Scheme I, (i.e., -CXVCi.g-alkyl ->
-CONH2 -» -C=N -> -tetrazole).
One method that can be used to prepare certain compounds of Formula (I) utilizes
intermediate (AJ) as illustrated in Reaction Schemes III and IV below:

Compounds of the structure (AJ) (wherein R is C,.4 alkyl, C2-4 alkenyl, and C2.4
alkynyl) can be prepared by treating the unsaturated pyrazole (AA) with benzyl bromide in a
suitable solvent like THF in the presence of NaOH as the base to give the N-benzyl pyrazole
(AB). The pyrazole (AB) can be saponified using methods known to one of skill in the art,
for example, treating with aqueous sodium hydroxide in a solvent mixture such as
THF/MeOH. The acid (AC) is coupled with yV-hydroxy succinimide using a coupling reagent
such as EDC. The ester (AD) is converted to the amide (AE) by treatment with concentrated
NH4OH solution in a solvent such as 1,4-dioxane. The amide (AE) can be reacted with a
dehydrating reagent such as cyanuric chloride, trifluro acetic anhydride, thionyl chloride and
like, in the presence of a non protic solvent such as DMF to give the nitrile (AF). The nitrile
(AF) is treated with an excess of borane-THF solution in a solvent like THF at low

temperature, followed by oxidation with hydrogen peroxide in the presence of sodium
hydroxide to give a 1:1 mixture of alcohols shown as (AC) and (AG").
Utilizing either alcohol (AC) or alcohol (AG") a variety of ethers can be prepared.
A representative synthesis is shown in Reaction Scheme IV using alcohol (AC)- It is
understood that a similar synthetic scheme can be utilized starting with alcohol (AG").

Compounds of the structure (AH) can be prepared by treating the alcohol
intermediate (AC) with an excess of alkyl halide in the presence of a base such as sodium
hydride in an aprotic solvent such as DMF. The nitrile (AH) is reacted with an azide such as
sodium azide, in the presence of a Lewis acid such as zinc bromide, to give the tetrazole of
the structure (Al). Final compounds can be prepared by removal of the benzyl protecting
group under oxidative conditions in a solvent like DMSO using a base such as potassium t-
butoxide and oxygen gas.
One method that can be used to prepare certain compounds of Formula (I) utilizes
intermediate (AS) as illustrated in Reaction Scheme V below:



Compounds of the structure (AV) can be prepared from 3-ethoxy-cyclopentenone by
treatment with dialkyloxalate such as di-tert-butyl oxalate or diethyloxalate in the presence of
a non-nucleophilic base such as LDA or LHMDS in a solvent such as THF to give the keto-
ester (AM). The keto ester (AIM) is reacted with benzyl hydrazine under reflux in a polar
solvent, such as ethanol or methanol containing glacial acetic acid to give the pyrazole (AN).
Alternatively, the keto-ester (AM) can be reacted with hydrazine, followed by alkylation of
the pyrazole with benzyl bromide using cesium carbonate as the base in a non-protic solvent
such as DMF. The pyrazole ester (AN) can be converted to the nitrile (AR) using a similar
sequence of steps described for (AC). The ketone (AR) is converted to the vinyl triflate
(AS) using Commins' reagent in the presence of LDA in a solvent such as THF.
Utilizing compound (AS), a variety of substituents (wherein R'is (^.4 alkyl, C2.4
alkenyl, and C2.4 alkynyl) can be introduced at C-5 as shown in Reaction Scheme VI.



The triflate (AS) can be reacted with a suitable stannane reagent in the presence of a
base such as lithium chloride and a catalyst such as tetrakis triphenyl phosphine palladium (0)
in a suitable solvent such as THF or toluene. Alternatively, the triflate (AS) can be reacted
with a suitable alkenyl boronic acid in the presence of a base such as potassium phosphate and
a catalyst such as tetrakis triphenyl phosphine palladium (0) in a suitable solvent such as 1,4-
dioxane. The nitrile (AT) is reacted with an azide such as sodium azide, in the presence of a
Lewis acid such as zinc bromide, to give the tetrazole of the structure (AU). Final compounds
are prepared by the removal of the benzyl protecting group that can be performed under
reductive conditions using palladium black in a polar solvent such as methanol or ethanol and
acid such as formic acid or concentrated hydrochloric acid.
Alternatively, alcohol (AC) may be fluorinated using methods known to those
skilled in the art, such as DAST [(diethylamino)sulfur trifluoride], to provide a fluoro
compound which can be elaborated to its tetrazole derivative and deprotected using methods
described herein.
One method that can be used to prepare certain compounds of Formula (I) is illustrated
in Reaction Scheme VII below:



A compound of the structure (BF) can be prepared from the keto ester (AM) by
reacting with hydrazine hydrate in a polar solvent such as ethanol containing glacial acetic
acid to give the pyrazole (AW). The pyrazole (AW) can be reacted with a sufonyl chloride
such as p-toluene sulfonyl chloride in a solvent such as CH2CI2 in the presence of a base such
as pyridine to give the TV-sulfonylated derivative (AX). The pyrazole ester (AX) can be
deprotected under acidic conditions using an acid such as TFA in CH2CI2 to form (AY). The
pyrazole acid (AY) can be converted to the nitrile (BB) using a similar sequence of steps
described for (AC). The ketone (BB) can be converted to the vinyl triflate (BC) using
Commins' reagent in the presence of a base such as LDA in a solvent such as THF.
The triflate (BC) can be coupled with tetramethyltin in the presence of a base such as
lithium chloride and a catalyst such as tetrakis triphenyl phosphine palladium (0) in a suitable
solvent such as THF or toluene. The p-toluene sulfonyl group can be removed by reacting
with tetra butyl ammonium fluoride solution in a solvent such as THF to give the pyrazole
(BE). The final compound is prepared by reacting the nitrile (BE) with an azide such as
sodium azide, in the presence of a Lewis acid such as zinc bromide, to give the tetrazole (BF).
The various organic group transformations and protecting groups utilized herein can
be performed by a number of procedures other than those described above. References for
other synthetic procedures that can be utililized for the preparation of intermediates or
compounds disclosed herein can be found in, for example, Smith, M. B.; and March, J.,
Advanced Organic Chgemistry, 5lh Edition, Wiley-lnterscience (2001); Larock, R.C.,
Comprehensive Organic Transformations, A Guide to Functional Croup Preparations, 2nd

Edition, VCH Publishers, Inc. (1999), or Wuts, P. G. M.; Greene, T. W.; Protective Groups in
Organic Synthesis, 3rd Edition, John Wiley and Sons, (1999), all three incorporated herein by
reference in their entirety,
Compounds of Formula (I) may have one or more chiral centers, and therefore exist as
enantiomers or diastereomers. The invention is understood to extend to all such enantiomers,
diastereomers and mixtures thereof, including racemates. Formula (I) and the formulae
described herein, supra, are intended to represent all individual isomers and mixtures thereof,
unless stated or shown otherwise.
Racemic mixtures can be resolved into the optical pure enatiomers by known methods,
for example, by separation of diastereomeric salts thereof with an optically active acid, and
liberating the optically active amine compound by treatment with a base. Another method for
resolving racemates into the optical pure enatiomers is based upon chromatography on an
optically active matrix or chiral support. Certain racemic compounds of the present invention
can thus be resolved into their optical antipodes, e.g., by fractional crystallization of d- or 1-
(tartrates, mandelates, or camphorsulphonate) salts for example. The compounds of the present
invention may also be resolved by the formation of diastereomeric amides or ester by reaction of
the compounds of the present invention with an optically active activated carboxylic acid such as
that derived from (+) or (-) phenylalanine, (+) or (-) phenylglycine, (+) or (-) camphanic acid or
by the formation of diastereomeric carbamates by reaction of the compounds of the present
invention with an optically active chloroformate or the like subsequently hydrolyzed.
Additional methods for the resolution of optical isomers known to those skilled in the art
can be used and will be apparent to the average worker skilled in the art. Such methods include
those discussed by J. Jaques, A. Collet, and S. Wilen in "Enantiomers, Racemates, and
Resolutions", John Wiley and Sons, New York (1981).
It is understood that the chemistry described herein is representative and is not intended
to be limiting in any manner.
Representative examples of compound of Formula (1) are shown below in TABLE A.








Methods and Uses
Compounds of the present invention are useful in the inhibition of the production of
free fatty acids. Further, compounds of the present invention are useful in the inhibition of
the production of free fatty acids while resulting in substantially lower or in some instances
no measurable flushing side effects, which effects are commonly associated with the
administration of niacin. Compounds of the present invention typically do not cause
vasodilation at doses as high as about 300 mpk as measured using methods know in the art,
such as the method shown in Example 7.
In some embodiments, compounds of the present invention cause essentially no
measurable flushing in an individual compared to an essentially equally effective dose of niacin.
In other embodiments compounds ofthe present invention cause less than about 80%, 75%, 70%,
65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5%, or I % measurable
flushing in an individual compared to an essentially equally effective dose of niacin.
Compounds ofthe present invention can modulate the activity ofthe RUP25 receptor.
The term "modulate" is meant to refer to the ability to increase or decrease activity ofthe
receptor. In some embodiments, compounds ofthe invention can be used in methods of
modulating a RUP25 receptor by contacting the receptor widi any one or more ofthe compound
as described herein. In still other embodiments, compounds ofthe invention can be used in
methods of method of modulating a RUP25 receptor for the treatment of a metabolic-related
disorder in an individual in need of such modulation comprising contacting the receptor with a
therapeutically-effective amount of a compound of Formula (I). In some embodiments,
compounds ofthe invention increase activity ofthe RUP25 receptor. In further embodiments,
compounds ofthe invention are agonists ofthe RUP25 receptor. The term "agonist", as used
herein, refers to agents that can stimulate activity ofthe receptor (i.e., activate), like the RUP25
receptor In some embodiments, compounds ofthe invention are partial agonists ofthe RUP25
receptor.
Another aspect ofthe present invention pertains to methods of treatment of a metabolic-
related disorder comprising administering to an individual in need of such treatment a
therapeutically-effective amount of a compound of Formula (1).

Another aspect of the present invention pertains to methods of raising HDL in an
individual comprising administering to said individual a therapeutically-effective amount of a
compound of Formula (I).
Another aspect of the present invention pertains to compounds of Formula (1), as
described herein, for use in a method of treatment of the human or animal body by therapy.
Another aspect of the present invention pertains to compounds of Formula (I), as
described herein, for use in a method of treatment of a metabolic-related disorder of the human or
animal body by therapy.
Another aspect of the present invention pertains to compounds of Formula (I), as
described herein, for use in a method of treatment of a metabolic-related disorder of the human or
animal body by therapy wherein said metabolic-related disorder is selected from the group
consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity,
impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart
disease and type 2 diabetes.
Another aspect of the present invention pertains to compounds of Formula (I), as
described herein, for use in a method of treatment of a metabolic-related disorder of the human or
animal body by therapy wherein said metabolic-related disorder is selected from the group
consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance and type 2
diabetes.
Another aspect of the present invention pertains to compounds of Formula (I), as
described herein, for use in a method of treatment of atherosclerosis of the human or animal body
by therapy.
Another aspect of the present invention pertains to compounds of Formula (1), as
described herein, for use in a method of raising HDL of the human or animal body by therapy.
Another aspect of the present invention pertains to uses of the compounds of Formula
(I), as described herein, for the manufacture of a medicament for use in the treatment of a
metabolic-related disorder.
Another aspect of the present invention pertains to uses of the compounds of Formula
(I), as described herein, for the manufacture of a medicament for use in the treatment of a
metabolic-related disorder selected from the group consisting of dyslipidemia, atherosclerosis,
coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous
disease, hypertension, stroke, Syndrome X, heart disease and type 2 diabetes.
Another aspect of the present invention pertains to uses of the compounds of Formula
(I), as described herein, for the manufacture of a medicament for use in the treatment of
atherosclerosis.

Another aspect of the present invention pertains to uses of the compounds of Formula
(I), as described herein, for the manufacture of a medicament for use in raising HDL in an
individual.
Some embodiments of the present invention relate to methods of treatment of metabolic-
related disorders. In some embodiments the metabolic-related disorder is of the group consisting
of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired
glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease and
type 2 diabetes. In some embodiments the metabolic-related disorder is dyslipidemia,
atherosclerosis, coronary heart disease, insulin resistance and type 2 diabetes. In some
embodiments the metabolic-related disorder is dyslipidemia. In some embodiments the
metabolic-related disorder is atherosclerosis. In some embodiments the metabolic-related
disorder is coronary heart disease. In some embodiments the metabolic-related disorder is insulin
resistance. In some embodiments the metabolic-related disorder is type 2 diabetes.
In some embodiments related to methods of the present invention, the individual is a
mammal. In futher embodiments, the mammal is a human.
Another aspect of the present invention pertains to methods of producing a
pharmaceutical composition comprising admixing or combining a compound of Formula (I), as
described herein, and a pharmaceutically acceptable carrier.
Compositions of the Present Invention
Some embodiments of the present invention include pharmaceutical compositions
comprising a compound according to Formula (I) in combination with a pharmaceutically
acceptable carrier.
Some embodiments of the present invention include a method of producing a
pharmaceutical composition comprising admixing at least one compound according to any of the
compound embodiments disclosed herein and a pharmaceutically acceptable carrier.
Formulations can be prepared by any suitable method, typically by uniformly mixing the
active compound(s) with liquids or finely divided solid carriers, or both, in the required
proportions, and then, if necessary, forming the resulting mixture into a desired shape.
Conventional excipients, such as binding agents, fillers, acceptable wetting agents,
tabletting lubricants, and disintegrants can be used in tablets and capsules for oral administration.
Liquid preparations for oral administration can be in the form of solutions, emulsions, aqueous or
oily suspensions, and syrups. Alternatively, the oral preparations can be in the form of dry
powder that can be reconstituted with water or another suitable liquid vehicle before use.
Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including
edible oils), preservatives, and flavorings and colorants can be added to the liquid preparations.
Parenteral dosage forms can be prepared by dissolving the compound of the invention in a


suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate
vial or ampoule. These are just a few examples of the many appropriate methods well known in
the art for preparing dosage forms.
A compound of the present invention can be formulated into pharmaceutical
compositions using techniques well known to those in the art. Suitable pharmaceutically-
acceptable carriers, outside those mentioned herein, are known in the art; for example, see
Remington, The Science and Practice of Pharmacy, 20'h Edition, 2000, Lippincott Williams &
Wilkins, (Editors; Gennaro, A. R., et al.).
While it is possible that a compound for use in the treatment of the present invention
may, in an alternative use, be administered as a raw or pure chemical, it is preferable however to
present the compound or "active ingredient" as a pharmaceutical formulation or composition
further comprising a pharmaceutical ly acceptable carrier. Therefore, one aspect of the present
invention encompasses pharmaceutical compositions comprising a pharmaceutical ly acceptable
carrier in combination with at least one compound according to Formula (1).
The invention provides pharmaceutical formulations comprising a compound of the
invention or a pharmaceutical ly acceptable salt, hydrate or solvate thereof together with one or
more pharmaceutically acceptable carriers therefor. The carrier(s) must be "acceptable" in the
sense of being compatible with the other ingredients of the formulation and not overly deleterious
to the recipient thereof.
Pharmaceutical formulations include those suitable for oral, rectal, nasal, topical
(including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous
and intravenous) administration or in a form suitable for administration by inhalation, insufflation
or by a transdermal patch. Transdermal patches dispense a drug at a controlled rate by presenting
the drug for absorption in an efficient manner with a minimum of degradation of the drug.
Typically, transdermal patches comprise an impermeable backing layer, a single pressure
sensitive adhesive and a removable protective layer with a release liner. One of ordinary skill in
the art will understand and appreciate the techniques appropriate for manufacturing a desired
efficacious transdermal patch based upon the needs of the artisan.
The compounds of the invention, together with a conventional adjuvant, carrier, or
diluent, may thus be placed into the form of pharmaceutical formulations and unit dosages
thereof, and in such form can be employed as solids, such as tablets or filled capsules, or liquids
such as solutions, suspensions, emulsions, elixirs, gels or capsules filled with the same, all for
oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable
solutions for parenteral (including subcutaneous) use. Such pharmaceutical compositions and
unit dosage forms thereof may comprise conventional ingredients in conventional proportions,
with or without additional active compounds or principles, and such unit dosage forms may


contain any suitable effective amount of the active ingredient commensurate with the intended
daily dosage range to be employed.
For oral administration, the pharmaceutical composition can be in the form of, for
example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably
made in the form of a dosage unit containing a particular amount of the active ingredient.
Examples of such dosage units are capsules, tablets, powders, granules or a suspension, with
conventional additives such as lactose, mannitol, corn starch or potato starch; with binders such
as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators
such as corn starch, potato starch or sodium carboxymethyl-cellulose; and with lubricants such as
talc or magnesium stearate. The active ingredient may also be administered by injection as a
composition wherein, for example, saline, dextrose or water can be used as a suitable
pharmaceutically acceptable carrier.
Compounds of the present invention or a solvate or physiologically functional derivative
thereof can be used as active ingredients in pharmaceutical compositions, specifically as RUP25
receptor agonists. By the term "active ingredient" is defined in the context of a "pharmaceutical
composition" and shall mean a component of a pharmaceutical composition that provides the
primary pharmacological effect, as opposed to an "'inactive ingredient" which would generally be
recognized as providing no pharmaceutical benefit.
The dose when using the compounds of the present invention can vary within wide
limits, and as is customary and is known to the physician, it is to be tailored to the individual
conditions in each individual case. It depends, for example, on the nature and severity of the
illness to be treated, on the condition of the patient, on the compound employed or on whether an
acute or chronic disease state is treated is conducted or on whether further active compounds are
administered in addition to the compounds of the present invention. Representative doses of the
present invention include, but not limited to, about 0.001 mg to about 5000 mg, about 0.001 to
about 2500 mg, about 0.001 to about 1000 mg, 0.001 to about 500 mg, 0.001 mg to about 250
mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg, and about 0.001 mg to about 25
mg. Multiple doses can be administered during the day, especially when relatively large amounts
are deemed to be needed, for example 2, 3 or 4, doses. Depending on the individual and as
deemed appropriate from the patient's physician or care-giver it may be necessary to deviate
upward or downward from the doses described herein.
The amount of active ingredient, or an active salt or derivative thereof, required for use
in treatment will vary not only with the particular salt selected but also with the route of
administration, the nature of the condition being treated and the age and condition of the patient
and will ultimately be at the discretion of the attendant physician or clinician. In general, one
skilled in the art understands how to extrapolate in vivo data obtained in a model system to
another, for example, an animal model to a human. Typically, animal models include, but are not


limited to, the rodents diabetes models as described in Example I, infra; the mouse
atherosclerosis model as described in Example 2, infra; or the in vivo animal athosclerosis model
as described in Example 5, infra. In some circumstances, these extrapolations may merely be
based on the weight of the animal model in comparison to another, such as a mammal, preferably
a human, however, more often, these extrapolations are not simply based on weight differences,
but rather incorporate a variety of factors. Representative factors include the type, age, weight,
sex, diet and medical condition of the patient, the severity of the disease, the route of
administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic
and toxicology profiles of the particular compound employed, whether a drug delivery system is
utilized, on whether an acute or chronic disease state is being treated is conducted or on whether
further active compounds are administered in addition to the compounds of the Formula (I) and
as part of a drug combination. The dosage regimen for treating a disease condition with the
compounds and/or compositions of this invention is selected in accordance with a variety factors,
such as, those cited above. Thus, the actual dosage regimen employed may vary widely and
therefore may deviate from a preferred dosage regimen and one skilled in the art will recognize
that dosage and dosage regimen outside these typical ranges can be tested and, where
appropriate, can be used in the methods of this invention.
The desired dose may conveniently be presented in a single dose or as divided doses
administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
The sub-dose itself can be further divided, e.g., into a number of discrete loosely spaced
administrations. The daily dose can be divided, especially when relatively large amounts are
administered as deemed appropriate, into several, for example 2, 3 or 4, part administrations. If
appropriate, depending on individual behavior, it can be necessary to deviate upward or
downward from the daily dose indicated.
The compounds of the present invention can be administrated in a wide variety of oral
and parenteral dosage forms. It will be obvious to those skilled in the art that the following
dosage forms may comprise, as the active component, either a compound of the invention or a
pharmaceutically acceptable salt of a compound of the invention.
For preparing pharmaceutical compositions from the compounds of the present
invention, pharmaceutically acceptable carriers can be either solid or liquid. Solid form
preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible
granules. A solid carrier can be one or more substances which may also act as diluents,
flavouring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet
disintegrating agents, or an encapsulating material.
In powders, the carrier is a finely divided solid which is in a mixture with the finely
divided active component.


In tablets, the active component is mixed with the carrier having the necessary binding
capacity in suitable proportions and compacted to the desire shape and size
The powders and tablets may contain varying percentage amounts of the active
compound. A representative amount in a powder or tablet may contain from 0.5 to about 90
percent of the active compound; however, an artisan would know when amounts outside of this
range are necessary. Suitable carriers for powders and tablets are magnesium carbonate,
magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth,
methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
The term "preparation" is intended to include the formulation of the active compound with
encapsulating material as carrier providing a capsule in which the active component, with or
without carriers, is surrounded by a carrier, which is thus in association with it. Similarly,
cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can
be used as solid forms suitable for oral administration.
For preparing suppositories, a low melting wax, such as an admixture of fatty acid
glycerides or cocoa butter, is first melted and the active component is dispersed homogeneously
therein, as by stirring. The molten homogenous mixture is then poured into convenient sized
molds, allowed to cool, and thereby to solidify.
Formulations suitable for vaginal administration can be presented as pessaries, tampons,
creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers
as are known in the art to be appropriate.
Liquid form preparations include solutions, suspensions, and emulsions, for example,
water or water-propylene glycol solutions. For example, parenteral injection liquid preparations
can be formulated as solutions in aqueous polyethylene glycol solution. Injectable preparations,
for example, sterile injectable aqueous or oleaginous suspensions can be formulated according to
the known art using suitable dispersing or wetting agents and suspending agents. The sterile
injectable preparation may also be a sterile injectable solution or suspension in a nontoxic
parcnterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among
the acceptable vehicles and solvents that can be employed are water, Ringer's solution, and
isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as
a solvent or suspending medium. For this purpose any bland fixed oil can be employed including
synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the
preparation of injectables.
The compounds according to the present invention may thus be formulated for parenteral
administration (e.g. by injection, for example bolus injection or continuous infusion) and can be
presented in unit dose form in'ampoules, pre-filled syringes, small volume infusion or in multi-
dose containers with an added preservative. The compositions may take such forms as
suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory


agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active
ingredient can be in powder form, obtained by aseptic isolation of sterile solid or by
lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free
water, before use.
Aqueous solutions suitable for oral use can be prepared by dissolving the active
component in water and adding suitable colorants, flavours, stabilizing and thickening agents, as
desired.
Aqueous suspensions suitable for oral use can be made by dispersing the finely divided
active component in water with viscous material, such as natural or synthetic gums, resins,
methylcellulose, sodium carboxymethylcellulose, or other well known suspending agents.
Also included are solid form preparations which are intended to be converted, shortly
before use, to liquid form preparations for oral administration. Such liquid forms include
solutions, suspensions, and emulsions. These preparations may contain, in addition to the active
component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants,
thickeners, solubilizing agents, and the like.
For topical administration to the epidermis the compounds according to the invention
can be formulated as ointments, creams or lotions, or as a transdermal patch.
Ointments and creams may, for example, be formulated with an aqueous or oily base
with the addition of suitable thickening and/or gelling agents. Lotions can be formulated with an
aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing
agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
Formulations suitable for topical administration in the mouth include lozenges
comprising active agent in a flavored base, usually sucrose and acacia or tragacanth; pastilles
comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and
acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Solutions or suspensions are applied directly to the nasal cavity by conventional means,
for example with a dropper, pipette or spray. The formulations can be provided in single or
multi-dose form. In the latter case of a dropper or pipette, this can be achieved by the patient
administering an appropriate, predetermined volume of the solution or suspension. In the case of
a spray, this can be achieved for example by means of a metering atomizing spray pump.
Administration to the respiratory tract may also be achieved by means of an aerosol
formulation in which the active ingredient is provided in a pressurized pack with a suitable
propellant. If the compounds of the Formula (I) or pharmaceutical compositions comprising
them are administered as aerosols, for example as nasal aerosols or by inhalation, this can be
carried out, for example, using a spray, a nebulizer, a pump nebulizer, an inhalation apparatus, a
metered inhaler or a dry powder inhaler. Pharmaceutical forms for administration of the
compounds of the Formula (1) as an aerosol can be prepared by processes well-known to the


person skilled in the art. For their preparation, for example, solutions or dispersions of the
compounds of the Formula (1) in water, water/alcohol mixtures or suitable saline solutions can be
employed using customary additives, for example benzyl alcohol or other suitable preservatives,
absorption enhancers for increasing the bioavailability, solubilizers, dispersants and others, and,
if appropriate, customary propellants, for example include carbon dioxide, CFC's, such as,
dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane; and the like. The
aerosol may conveniently also contain a surfactant such as lecithin. The dose of drug can be
controlled by provision of a metered valve.
In formulations intended for administration to the respiratory tract, including intranasal
formulations, the compound will generally have a small particle size for example of the order of
10 microns or less. Such a particle size can be obtained by means known in the art, for example
by micronization. When desired, formulations adapted to give sustained release of the active
ingredient can be employed.
Alternatively the active ingredients can be provided in the form of a dry powder, for
example, a powder mix of the compound in a suitable powder base such as lactose, starch, starch
derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
Conveniently the powder carrier will form a gel in the nasal cavity. The powder composition can
be presented in unit dose form for example in capsules or cartridges of, e.g., gelatin, or blister
packs from which the powder can be administered by means of an inhaler.
The pharmaceutical preparations are preferably in unit dosage forms. In such form, the
preparation is subdivided into unit doses containing appropriate quantities of the active
component. The unit dosage form can be a packaged preparation, the package containing
discrete quantities ofpreparation, such as packeted tablets, capsules, and powders in vials or
ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can
be the appropriate number of any of these in packaged form.
Tablets or capsules for oral administration and liquids for intravenous administration are
preferred compositions.
Compounds of the present invention can be converted to 'pro-drugs." The term "pro-
drugs" refers to compounds that have been modified with specific chemical groups known in the
art and when administered into an individual these groups undergo biotransformation to give the
parent compound. Pro-drugs can thus be viewed as compounds of the invention containing one
or more specialized non-toxic protective groups used in a transient manner to alter or to eliminate
a property of the compound. In general, the "pro-drug" approach is utilized to facilitate oral
absorption. A thorough discussion is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel
Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in
Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press,
1987, both of which are hereby incorporated by reference in their entirety.


Combination Therapy:
While the compounds of the present invention can be administered as the sole active
pharmaceutical agent (i.e., mono-therapy), they can also be used in combination with other
pharmaceutical agents (i.e., combination-therapy), such as, for the treatment of the
diseases/conditions/disorders described herein. Therefore, another aspect of the present
invention includes methods of treatment of metabolic related diseases comprising administering
to an individual in need of such treatment a therapeutically-effective amount of a compound of
the present invention in combination with one or more additional pharmaceutical agent as
described herein.
Suitable pharmaceutical agents that can be used in combination with the compounds of
the present invention include anti-obesity agents such as apolipoprotein-B secretion/microsomal
triglyceride transfer protein (apo-B/MTP) inhibitors, MCR-4 agonists, cholescystokinin-A
(CCK-A) agonists, serotonin and norepinephrine reuptake inhibitors (for example, sibutramine),
sympathomimetic agents, pt adrenergic receptor agonists, dopamine agonists (for example,
bromocriptine), melanocyte-stimulating hormone receptor analogs, cannabinoid 1 receptor
antagonists [for example, SR141716: /V-(piperidin-l-yl)-5-(4-chlorophenyl)-l-(2,4-
dichlorophenyl)-4-methyl-l//-pyrazole-3-carboxamide], melanin concentrating hormone
antagonists, leptons (the OB protein), leptin analogues, leptin receptor agonists, galanin
antagonists, lipase inhibitors (such as tetrahydrolipstatin, i.e., Orlistat), anorectic agents (such as
a bombesin agonist), Neuropeptide-Y antagonists, thyromimetic agents, dehydroepiandrosterone
or an analogue thereof, glucocorticoid receptor agonists or antagonists, orexin receptor
antagonists, urocortin binding protein antagonists, glucagon-like peptide-1 receptor agonists,
ciliary neutrotrophic factors (such as Axokine™ available from Regeneron Pharmaceuticals, Inc.,
1 arrytown, NY and Procter & Gamble Company, Cincinnati, OH), human agouti-related
proteins (AGRP), ghrelin receptor antagonists, histamine 3 receptor antagonists or reverse
agonists, neuromedin U receptor agonists, noradrenergic anorectic agents (for example,
phentermine, mazindol and the like) and appetite suppressants (for example, bupropion).
Other anti-obesity agents, including the agents set forth infra, are well known, or will be
readily apparent in light of the instant disclosure, to one of ordinary skill in the art.
In some embodiments, the anti-obesity agents are selected from the group consisting of
orlistat, sibutramine, bromocriptine, ephedrine, leptin, and pseudoephedrine. In a further
embodiment, compounds of the present invention and combination therapies are administered in
conjunction with exercise and/or a sensible diet.
It is understood that the scope of combination-therapy of the compounds of the present
invention with other anti-obesity agents, anorectic agents, appetite suppressant and related agents


is not limited to those listed above, but includes in principle any combination with any
pharmaceutical agent or pharmaceutical composition useful for the treatment of overweight and
obese individuals.
Other suitable pharmaceutical agents, in addition to anti-obesity agents, that can be used
in combination with the compounds of the present invention include agents useful in the
treatment of concomitant disorders. Treatment of such disorders include the use of one or more
pharmaceutical agents known in the art that belong to the classes of drugs referred to, but not
limited to, the following: sulfonylureas, meglitinides, biguanides, a-glucosidase inhibitors,
peroxisome proliferators-activated receptor-y (i.e., PPAR-y) agonists, insulin, insulin analogues,
HMG-CoA reductase inhibitors, cholesterol-lowering drugs (for example, fibrates that include:
fenofibrate, bezafibrate, gemfibrozil, clofibrate and the like; bile acid sequestrants which include:
cholestyramine, colestipol and the like; and niacin), antiplatelet agents (for example, aspirin and
adenosine diphosphate receptor antagonists that include: clopidogrel, ticlopidine and the like),
angiotensin-converting enzyme inhibitors, angiotensin II receptor antagonists and adiponectin.
In accordance to one aspect of the present invention, a compound of the present can be used in
combination with a pharmaceutical agent or agents belonging to one or more of the classes of
drugs cited herein.
It is understood that the scope of combination-therapy of the compounds of the present
invention with other pharmaceutical agents is not limited to those listed herein, supra or infra,
but includes in principle any combination with any pharmaceutical agent or pharmaceutical
composition useful for the treatment of diseases, conditions or disorders that are linked to
metabolic-related disorders.
Some embodiments of the present invention include methods of treatment of a disease,
disorder or condition as described herein comprising administering to an individual in need of
such treatment a therapeutically effect amount or dose of a compound of the present invention in
combination with at least one pharmaceutical agent selected from the group consisting of:
sulfonylureas, meglitinides, biguanides, a-glucosidase inhibitors, peroxisome proliferators-
activated receptor-y (i.e., PPAR-y) agonists, insulin, insulin analogues, HMG-CoA reductase
inhibitors, cholesterol-lowering drugs (for example, fibrates that include: fenofibrate, bezafibrate,
gemfibrozil, clofibrate and the like; bile acid sequestrants which include: cholestyramine,
colestipol and the like; and niacin), antiplatelet agents (for example, aspirin and adenosine
diphosphate receptor antagonists that include: clopidogrel, ticlopidine and the like), angiotensin-
converting enzyme inhibitors, angiotensin II receptor antagonists and adiponectin. In some
embodiments, the pharmaceutical composition further comprises one or more agents selected
from the group consisting of a-glucosidase inhibitor, aldose reductase inhibitor, biguanide,


HMG-CoA reductase inhibitor, squalene synthesis inhibitor, fibrate, LDL catabolism enhancer,
angiotensin converting enzyme inhibitor, insulin secretion enhancer and thiazolidinedione.
One aspect of the present invention encompasses pharmaceutical compositions
comprising at least one compound according to Formula (1), as described herein. In some
embodiments, the pharmaceutical composition further comprises one or more agents selected
from the group consisting of, for example, a-glucosidase inhibitor, aldose reductase inhibitor,
biguanide, HMG-CoA reductase inhibitor, squalene synthesis inhibitor, fibrate, LDL catabolism
enhancer, angiotensin converting enzyme inhibitor, insulin secretion enhancer and
thiazolidinedione.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include a-glucosidase inhibitors. a-Glucosidase inhibitors belong to the class
of drugs which competitively inhibit digestive enzymes such as a-amylase, maltase, a-
dextrinase, sucrase, etc. in the pancreas and or small intestine. The reversible inhibition by a-
glucosidase inhibitors retard, diminish or otherwise reduce blood glucose levels by delaying the
digestion of starch and sugars. Some representative examples of a-glucosidase inhibitors include
acarbose, N-(l ,3-dihydroxy-2-propyl)valiolamine (generic name; voglibose), miglitol, and a-
glucosidase inhibitors known in the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include sulfonylureas. The sulfonylureas (SU) are drugs which promote
secretion of insulin from pancreatic (3 cells by transmitting signals of insulin secretion via SU
receptors in the cell membranes. Examples of the sulfonylureas include glyburide, glipizide,
glimepiride and other sulfonylureas known in the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the meglitinides. The meglitinides are benzoic acid derivatives
represent a novel class of insulin secretagogues. These agents target postprandial hyperglycemia
and show comparable efficacy to sulfonylureas in reducing HbA|C. Examples of meglitinides
include repaglinide, nateglinide and other meglitinides known in the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the biguanides. The biguanides represent a class of drugs that stimulate
anaerobic glycolysis, increase the sensitivity to insulin in the peripheral tissues, inhibit glucose
absorption from the intestine, suppress of hepatic gluconeogenesis, and inhibit fatty acid
oxidation. Examples of biguanides include phenformin, metformin, buformin, and biguanides
known in the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the a-glucosidase inhibitors. The a-glucosidase inhibitors
competitively inhibit digestive enzymes such as a-amylase, maltase, a-dextrinase, sucrase, etc.


in the pancreas and or small intestine. The reversible inhibition by a-glucosidase inhibitors
retard, diminish or otherwise reduce blood glucose levels by delaying the digestion of starch and
sugars. Examples of a-glucosidase inhibitors include acarbose, N-(l ,3-dihydroxy-2-
propyl)valiolamine (generic name; voglibose), rniglitol, and a-glucosidase inhibitors known in
the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the peroxisome proliferators-activated receptor-y (i.e., PPAR-y)
agonists. The peroxisome proliferators-activated receptor-y agonists represent a class of
compounds that activates the nuclear receptor PPAR-y and therefore regulate the transcription of
insulin-responsive genes involved in the control of glucose production, transport and utilization.
Agents in the class also facilitate the regulation of fatty acid metabolism. Examples of PPAR-y
agonists include rosiglitazone, pioglitazone, tesaglitazar, netoglitazone, GW-409544, GW-
5015 16 and PPAR-y agonists known in the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the HMG-CoA reductase inhibitors. The HMG-CoA reductase
inhibitors are agents also referred to as Statin compounds that belong to a class of drugs that
lower blood cholesterol levels by inhibiting hydroxymethylglutalyl CoA (HMG-CoA) reductase.
HMG-CoA reductase is the rate-limiting enzyme in cholesterol biosynthesis. The statins lower
serum LDL concentrations by upregulating the activity of LDL receptors and are responsible for
clearing LDL from the blood. Some representative examples the statin compounds include
rosuvastatin, pravastatin and its sodium salt, simvastatin, lovastatin, atorvastatin, fluvastatin,
cerivastatin, pitavastatin, BMS's "superstatin", and HMG-CoA reductase inhibitors known in the
art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the angiotensin converting enzyme (ACE) inhibitors. The angiotensin
converting enzyme inhibitors belong to the class of drugs that partially lower blood glucose
levels as well as lowering blood pressure by inhibiting angiotensin converting enzymes.
Examples of the angiotensin converting enzyme inhibitors include captopril, enalapril, alacepril,
delapril; ramipril, lisinopril, imidapril, benazepril, ceronapril, cilazapril, enalaprilat, fosinopril,
moveltopril, perindopril, quinapril, spirapril, temocapril, trandolapril, and angiotensin converting
enzyme inhibitors known in the art.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the
present invention include the angiotensin II receptor antagonists. Angiotensin II receptor
antagonists target the angiotensin II receptor subtype 1 (i.e., ATI) and demonstrate a beneficial
effect on hypertension. Examples of angiotensin II receptor antagonists include losartan (and the
potassium salt form), and angiotensin II receptor antagonists known in the art.


Other treatments for one or more of the diseases cited herein include the use of one or
more pharmaceutical agents known in the art that belong to the classes of drugs referred to, but
not limited to, the following: amylin agonists (for example, pramlintide), insulin secretogogues
(for example, GLP-1 agonists; exendin-4; insulinotropin (NN2211); dipeptyl peptidase inhibitors
(for example, NVP-DPP-728), acyl CoA cholesterol acetyltransferase inhibitors (for example,
Ezetimibe, eflucimibe, and like compounds), cholesterol absorption inhibitors (for example,
ezetimibe, pamaqueside and like compounds), cholesterol ester transfer protein inhibitors (for
example, CP-529414, JTT-705, CETi-1, torcetrapib and like compounds), microsomal
triglyceride transfer protein inhibitors (for example, implitapide, and like compounds),
cholesterol modulators (for example, NO-1886, and like compounds), bile acid modulators (for
example, G'l'l03-279 and like compounds) and squalene synthase inhibitors.
Squalene synthesis inhibitors belong to a class of drugs that lower blood cholesterol
levels by inhibiting synthesis of squalene. Examples of the squalene synthesis inhibitors include
(S)-ct-[Bis[2,2-dimethyl-1 -oxopropoxy)methoxy] phosphinyl]-3-phenoxybenzenebutanesulfonic
acid, mono potassium salt (BMS-188494) and squalene synthesis inhibitors known in the art.
In accordance with the present invention, the combination can be used by mixing the
respective active components either all together or independently with a pharmaceutically
acceptable carrier, excipient, binder, diluent, etc., as described herein above, and administering
the mixture or mixtures either orally or non-orally as a pharmaceutical composition. When a
compound or a mixture of compounds of Formula (I) are administered as a combination therapy
with another active compound the therapeutic agents can be formulated as separate
pharmaceutical compositions given at the same time or at different times, or the therapeutic
agents can be given as a single composition.
In accordance with the present invention, the combination of a compound of the present
invention and pharmaceutical agent can be prepared by mixing the respective active components
either all together or independently with a pharmaceutically acceptable carrier, excipient, binder,
diluent, etc., as described herein, and administering the mixture or mixtures either orally or non-
orally as a pharmaceutical composition. When a compound or a mixture of compounds of
Formula (I) are administered as a combination therapy with another active compound the
therapeutic agents can be formulated as a separate pharmaceutical compositions given at the
same time or at different times, or the therapeutic agents can be given as a single composition.
Labeled Compounds and Assay Methods
Another object of the present invention relates to radio-labeled compounds of Formula
(1) that are useful not only in radio-imaging but also in assays, both in vitro and in vivo, for
localizing and quantitating RUP25 in tissue samples, including human, and for identifying


RUP25 ligands by inhibition binding of a radio-labeled compound. It is a further object of this
invention to include novel RUP25 assays of which comprise such radio-labeled compounds.
The present invention embraces isotopically-labeled compounds of Formula (I) and any
subgenera herein, such as but not limited to, Formulae (la) to (iz); and (lla) to (lid). An
"isotopically" or "radio-labeled" compounds are those which are identical to compounds
disclosed herein, but for the fact that one or more atoms are replaced or substituted by an atom
having an atomic mass or mass number different from the atomic mass or mass number typically
found in nature (i.e., naturally occurring). Suitable radionuclides that can be incorporated in
compounds of the present invention include but are not limited to 2H (also written as D for
deuterium), 3H (also written as T for tritium), "C, nC, l4C, l3N, 15N, l50, l70,180, l8F, ,5S, 36CI,
82Br, 75Br, 76Br, 77Br, l23l, 124l, 125I and ml. The radionuclide that is incorporated in the instant
radio-labeled compounds will depend on the specific application of that radio-labeled compound.
For example, for in vitro RUP25 labeling and competition assays, compounds that incorporate
3H, !4C, 82Br, l25I, l3'l, 35S or will generally be most useful. For radio-imaging applications "C,
"F, l25l, '23l, 124l, "'1,75Br, 76Br or 77Br will generally be most useful.
It is understood that a "radio-labeled " or "labeled compound" is a compound of
Formula (I) that has incorporated at least one radionuclide; in some embodiments the
radionuclide is selected from the group consisting of 3H, l4C, l23l , 35S and 82Br.
Certain isotopically-labeled compounds of the present invention are useful in compound
and/or substrate tissue distribution assays. In some embodiments the radionuclide 3H and/or l4C
isotopes are useful in these studies. Further, substitution with heavier isotopes such as deuterium
(i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability
(e.g., increased in vivo half-life or reduced dosage requirements) and hence can be preferred in
some circumstances. Isotopically labeled compounds of the present invention can generally be
prepared by following procedures analogous to those disclosed in the Schemes supra and
Examples infra, by substituting an isotopically labeled reagent for a non-isotopically labeled
reagent. Other synthetic methods that are useful are discussed infra. Moreover, it should be
understood that all of the atoms represented in the compounds of the invention can be either the
most commonly occurring isotope of such atoms or the more scarce radio-isotope or nonradio-
active isotope.
Synthetic methods for incorporating radio-isotopes into organic compounds are
applicable to compounds of the invention and are well known in the art. These synthetic
methods, for example, incorporating activity levels of tritium into target molecules, and are as
follows:
A. Catalytic Reduction with Tritium Gas - This procedure normally yields high specific
activity products and requires halogenated or unsaturated precursors.


B. Reduction with Sodium Borohydride [ H] - This procedure is rather inexpensive and
requires precursors containing reducible functional groups such as aldehydes, ketones, lactones,
esters, and the like.
C. Reduction with Lithium Aluminum Hydride [3H ] - This procedure offers products at
almost theoretical specific activities. It also requires precursors containing reducible functional
groups such as aldehydes, ketones, lactones, esters, and the like.
D. Tritium Gas Exposure Labeling - This procedure involves exposing precursors
containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
E. N-Methylation using Methyl Iodide [JH] - This procedure is usually employed to
prepare O-methyl orN-methyl (3H) products by treating appropriate precursors with high
specific activity methyl iodide (3H). This method in general allows for higher specific activity,
such as for example, about 70-90 Ci/mmol.
Synthetic methods for incorporating activity levels of l23I into target molecules include:
A. Sandmeyer and like reactions - This procedure transforms an aryl or heteroaryl
amine into a diazonium salt, such as a tetrafluoroborate salt, and subsequently to 125I labeled
compound using Na12Sl. A represented procedure was reported by Zhu, D.-G. and co-workers in
J. Org. Chem. 2002, 67, 943-948.
B. Ortho '"lodination of phenols - This procedure allows for the incorporation of l23l at
the ortho position of a phenol as reported by Collier, T. L. and co-workers in J. Labeled Compd
Radiopharm. 1999, 42, S264-S266.
C. Aryl and heteroaryl bromide exchange with '251 - This method is generally a two
step process. The first step is the conversion of the aryl or heteroaryl bromide to the
corresponding tri-alkyltin intermediate using for example, a Pd catalyzed reaction (i.e. Pd(Ph:,P)4]
or through an aryl or heteroaryl lithium, in the presence of a tri-alky Itinhalide or hexaalkylditin
[e.g., (CH-,)3SnSn(CHi)v|. A represented procedure was reported by Bas, M.-D. and co-workers
in./ Labeled Compd Radiopharm 2001, 44, S280-S282.
A radio-labeled RUP25 compound of Formula (I) can be used in a screening assay to
identify/evaluate compounds. In general terms, a newly synthesized or identified compound (i.e.,
test compound) can be evaluated for its ability to reduce binding of the "radio-labeled compound
of Formula (1)" to the RUP25 receptor. Accordingly, the ability of a test compound to compete
with the "radio-labeled compound of Formula (I)" for the binding to the RUP25 receptor directly
correlates to its binding affinity.
The labeled compounds of the present invention bind to the RUP25 receptor. In one
embodiment the labeled compound has an IC50 less than about 500 u,M, in another embodiment
the labeled compound has an ICJO less than about 100 u.M, in yet another embodiment the labeled
compound has an IC50 less than about 10 u,M, in yet another embodiment the labeled compound


has an IC50 less than about 1 u.M, and in still yet another embodiment the labeled inhibitor has an
IC50 less than about 0.1 U.M.
Other uses of the disclosed receptors and methods will become apparent to those in the art based
upon, inter alia, a review of this disclosure.
As will be recognized, the steps of the methods of the present invention need not be
performed any particular number of times or in any particular sequence. Additional objects,
advantages, and novel features of this invention will become apparent to those skilled in the art
upon examination of the following examples thereof, which are intended to be illustrative and not
intended to be limiting.
EXAMPLES
The following Examples are provided for illustrative purposes and not as a means of
limitation. One of ordinary skill in the art would be able to design equivalent assays and methods
based on the disclosure herein, all of which form part of the present invention.
Example 1
RODENT DIABETES MODELS
Rodent models of type 2 diabetes associated with obesity and insulin resistance have
been developed. Genetic models such as db/db and ob/ob [see Diabetes (1982)31:1-6] in mice
and fa/fa in zucker rats have been developed for understanding the pathophysiology of disease
and for testing candidate therapeutic compounds [Diabetes (1983) 32:830-838; Annu Rep
Sankyo Res Lab (1994) 46:1-57]. The homozygous animals, C57 BL/KsJ-db/db mice developed
by Jackson Laboratory are obese, hyperglycemic, hyperinsuhnemic and insulin resistant [J Clin
Invest (1990) 85:962-967], whereas heterozygotes are lean and normoglycemic. In the db/db
model, mice progressively develop insulinopenia with age, a feature commonly observed in late
stages of human type 2 diabetes when sugar levels are insufficiently controlled. Since this model
resembles that of human type 2 diabetes, the compounds of the present invention are tested for
activities including, but not limited to, lowering of plasma glucose and triglycerides. Zucker
(fa/fa) rats are severely obese, hyperinsuhnemic, and insulin resistant {Coleman, Diabetes (1982)
31:1; E Shafrir in Diabetes Mellitus, H Rifkinand D Porte, Jr, Eds [Elsevier Science Publishing
Co, New York, ed. 4, (1990), pp. 299-340]}, and the fa/fa mutation may be the rat equivalent of
the murine db mutation [Friedman et al, Cell (1992) 69:217-220; Truett et al, Proc Natl Acad Sci
USA (1991) 88:7806]. Tubby (tub/tub) mice are characterized by obesity, moderate insulin
resistance and hyperinsulineinia without significant hyperglycemia [Coleman et al, Heredity
(1990)81:424].


The present invention encompasses the use of compounds of the invention for reducing
the insulin resistance and hyperglycemia in any or all of the above rodent diabetes models, in
humans with type 2 diabetes or other preferred metabolic-related disorders or disorders of lipid
metabolism described previously, or in models based on other mammals. Plasma glucose and
insulin levels will be tested, as well as other factors including, but not limited to, plasma free
fatty acids and triglycerides.
In Vivo Assay for Anti-Hvperglvcemic Activity of Compounds of the Invention
Genetically altered obese diabetic mice (db/db) (male, 7-9 weeks old) are housed (7-9
mice/cage) under standard laboratory conditions at 22°C and 50% relative humidity, and
maintained on a diet of Purina rodent chow and water ad libitum. Prior to treatment, blood is
collected from the tail vein of each animal and blood glucose concentrations are determined
using One Touch Basic Glucose Monitor System (Lifescan). Mice that have plasma glucose
levels between 250 to 500 mg/dl are used. Each treatment group consists of seven mice that are
distributed so that the mean glucose levels are equivalent in each group at the start of the study,
db/db mice are dosed by micro-osmotic pumps, inserted using isoflurane anesthesia, to provide
compounds of the invention, saline, or an irrelevant compound to the mice subcutaneous ly (s.c).
Blood is sampled from the tail vein at intervals thereafter and analyzed for blood glucose
concentrations. Significant differences between groups (comparing compounds of the invention
to saline-treated) are evaluated using Student t-test.
Example 2
MOUSE ATHEROSCLEROSIS MODEL
Adiponectin-deficient mice generated through knocking out the adiponectin gene have
been shown to be predisposed to atherosclerosis and to be insulin resistant. The mice are also a
suitable model for ischemic heart disease [Matsuda, M et al. J Biol Chem (2002) July, and
references cited therein, the disclosures of which are incorporated herein by reference in their
entirety].
Adiponectin knockout mice are housed (7-9 mice/cage) under standard laboratory
conditions at 22°C and 50% relative humidity. The mice are dosed by micro-osmotic pumps,
inserted using isoflurane anesthesia, to provide compounds of the invention, saline, or an
irrelevant compound to the mice subcutaneously (s.c). Neointimal thickening and ischemic
heart disease are determined for different groups of mice sacrificed at different time intervals.
Significant differences between groups (comparing compounds of the invention to saline-treated)
are evaluated using Student t-test.
Example 3
In Vitro Biological Activity


A modified Flash Plate™ Adinylyl Cyclase kit (New England Nuclear; Cat. No.
SMP004A) was utilized for direct identification of candidate compounds as agonists to
hRUP25 in accordance with the following protocol. The term hRUP25 includes the human
sequences found in GenBank Accession No. NM177551 for the nucleotide and GenBank
Accession No. NP 808219 for the polypeptide, and naturally-occurring allelic variants,
mammalian orthologs, and recombinant mutants thereof.
CHO cells stably transfected with an expression vector encoding hRUP25 and cultured
under condition permissive for cell surface expression of the encoded hRUP25 receptor were
harvested from flasks via non-enzymatic means. The cells were washed in PBS and
resuspended in the manufacturer's Assay Buffer. Live cells were counted using a
hemacytometer and Trypan blue exclusion, and the cell concentration was adjusted to 2xl06
cells/ml. cAMP standards and Detection Buffer (comprising 2 u,Ci of tracer [125l]-cAMP (100
ul) to 11 ml Detection Buffer) were prepared and maintained in accordance with the
manufacturer's instructions. Candidate compounds identified as per above (if frozen, thawed
at room temperature) were added to their respective wells (preferably wells of a 96-well plate)
at increasing concentrations (3ul/well; 12uM final assay concentration). To these wells,
100,000 cells in 50u.l of Assay Buffer were added and the mixture was then incubated for 30
minutes at room temperature, with gentle shaking. Following the incubation, lOOul of
Detection Buffer was added to each well, followed by incubation for 2-24 hours. Plates were
counted in a Wallac MicroBeta™ plate reader using "Prot. #31" (as per manufacturer
instructions).
Certain compounds of the invention have an ECjo in the cAMP Whole Cell method of
about 25 U.M or less.
Example 4: In vitro Biological Activity
35S-GTPyS bindinu assay:
Membranes prepared from Chinese Hamster Ovary (CHO)-K I cells stably expressing
the niacin receptor or vector control (7 u.g/assay) were diluted in assay buffer (100 mM HEPES,
100 mM NaCI and 10 mM MgCU, pH 7.4) in Wallac Scintistrip plates and pre-incubated with
test compounds diluted in assay buffer containing40 u,M GDP (final [GDP] was 10 uM) for- 10
minutes before addition of 35S-GTPvS to 0.3 nM. To avoid potential compound precipitation, all
compounds were first prepared in 100% DMSO and then diluted with assay buffer resulting in a
final concentration of 3% DMSO in the assay. Binding was allowed to proceed for one hour
before centrifuging the plates at 4000 rpm for 15 minutes at room temperature and subsequent
counting in a TopCount scintillation counter. Non-linear regression analysis of the binding
curves was performed in GraphPad Prism.


Membrane Preparation
Materials:
CHO-Kl cell culture medium: F-12 Kaighn's Modified Cell Culture Medium with 10% FBS, 2
ITIM L-Glutamine, I mM Sodium Pyruvate and 400 u.gml
G418
Membrane Scrape Buffer: 20 mM HEPES
IOmMEDTA,pH7.4
Membrane Wash Buffer: 20 mM HEPES
0.1 mMEDTA,pH7.4
Protease Inhibitor Cocktail: P-8340, (Sigma, St. Louis, MO)
Procedure:
o Aspirate cell culture media off the 15 cm2 plates, rinse with 5 mL cold PBS and aspirate.
o Add 5 mL Membrane Scrape Buffer and scrape cells. Transfer scrape into 50 mL
centrifuge tube. Add 50 (iL Protease Inhibitor Cocktail,
o Spin at 20,000 rpm for 17 minutes at 4°C.
o Aspirate off the supernatant and resuspend pellet in 30 mL Membrane Wash Buffer.
Add 50 uL Protease Inhibitor Cocktail.
o Spin at 20,000 rpm for 17 minutes at 4°C.
o Aspirate the supernatant off the membrane pellet. The pellet may be frozen at -80°C for
later use or it can be used immediately.
Assay
Materials:
Guanosine 5'-diphosphate sodium salt (GDP, Sigma-Aldrich Catalog #87127)
Guanosine 5'-[yJ5S] thiotriphosphate, triethylammonium salt ([35S]GTPyS, Amersham
Biosciences Catalog #SJ 1320, -lOOOCi/mmol)
96 well Scintiplates(Perkin-Elmer #1450-501)
Binding Buffer: 20 mM HEPES, pH 7.4


lOOmMNaCI
lOmMMgCL
GDP Buffer: binding buffer plus GDP, ranging from 0.4 to 40 uM, make fresh before assay
Procedure:
(total assay volume - lOOu/well)
25 u.L GDP buffer with or without compounds (final GDP 10 [iM - so use 40 u.M stock)
50 u.L membrane in binding buffer (0.4mg protein/mL)
25 uX [3SS]GTPyS in binding buffer. This is made by adding 5 u.1 [l3S]GTPyS stock into
1 OmL binding buffer (This buffer has no GDP)
o Thaw compound plates to be screened (daughter plates with 5 u.L compound @ 2mM in
l00%DMSO)
o Dilute the 2 mM compounds 1:50 with 245 u.L GDP buffer to 40 u.M in 2% DMSO.
Thaw frozen membrane pellet on ice
o Homogenize membranes briefly until in suspension using a POLYTRON PT3100
(probe PT-DA 3007/2 at setting of 7000 rpm). Determine the membrane protein
concentration by Bradford assay. Dilute membrane to a protein concentrations of 0.40
mg/ml in Binding Buffer. (Note: the final assay concentration is 20 u.g/well).
o Add 25 |.iL compounds in GDP buffer per well to Scintiplate.
o Add 50 u,L of membranes per well to Scintiplate.
o Pre-incubate for 5-10 minutes at room temperature,
o Add 25 jaL of diluted ["SJGTPyS. Incubate on shaker (Lab-Line model #1314, shake at
setting of 4) for 60 minutes at room temperature.
o Assay is stopped by spinning plates sealed with plate covers at 2500 rpm for 20 minutes
at 22° C
o Read on TopCount NXT scintillation counter - 35S protocol.
Certain compounds of the invention have an EC50 in the functional in vitro GTPyS
binding assay within the range of about 10-100 ^M. More advantageous compounds of the
invention have an EC50 value in this assay within the range of about 1-10 ^M. Still more
advantages compounds have an EC50 value in this assay of less than about 1 uM.
Example 5


In Vivo Animal Model
One utility of the compound of the present invention as a medical agent in the
prophylaxis and treatment of a high total cholesterol/HDL-cholesterol ratio and conditions
relating thereto is demonstrated by the activity of the compound in lowering the ratio of total
cholesterol to HDL-cholesterol, in elevating HDL-cholesterol, or in protection from
atherosclerosis in an in vivo pig model. Pigs are used as an animal model because they reflect
human physiology, especially lipid metabolism, more closely than most other animal models.
An illustrative in vivo pig model not intended to be limiting is presented here
Yorkshire albino pigs (body weight 25.5 ± 4 kg) are fed a saturated fatty acid rich and
cholesterol rich (SFA-CHO) diet during 50 days (1 kg chow 35 kg"' pig weight), composed of
standard chow supplemented with 2% cholesterol and 20% beef tallow [Royo T et al., European
Journal of Clinical Investigation (2000) 30:843-52; which disclosure is hereby incorporated by
reference in its entirety]. Saturated to unsaturated fatty acid ratio is modified from 0.6 in normal
pig chow to 1. 12 in the SFA-CHO diet. Animals are divided into two groups, one group {n = 8)
fed with the SFA-CHO diet and treated with placebo and one group (w = 8) fed with the SFA-
CHO diet and treated with the compound (3.0 mg kg"1). Control animals are fed a standard chow
for a period of 50 days. Blood samples are collected at baseline (2 days after the reception of the
animals), and 50 days after the initiation of the diet. Blood lipids are analyzed. The animals are
sacrificed and necropsied.
Alternatively, the foregoing analysis comprises a plurality of groups each treated with a
different dose of the compound. Preferred said doses are selected from the group consisting of:
0.1 mgkg"',0.3mgkg"', 1.0 mg kg"', 3.0 mg kg"', 10 mg kg', 30 mgkg"1 and lOOmgkg"1.
Alternatively, the foregoing analysis is carried out at a plurality of timepoints. Preferred said
timcpoints are selected from the group consisting of 10 weeks, 20 weeks, 30 weeks, 40 weeks,
and 50 weeks.
HDL-Cholesterol
Blood is collected in trisodium citrate (3.8%, 1:10). Plasma is obtained after
centrifugation (1200 g 15 min) and immediately processed. Total cholesterol, HDL-cholesterol,
and LDL-cholesterol are measured using the automatic analyzer Kodak Ektachem DT System
(Eastman Kodak Company, Rochester, NY, USA). Samples with value parameters above the
range are diluted with the solution supplied by the manufacturer and then re-analyzed. The total
cholesterol/HDL-cholesterol ratio is determined. Comparison is made of the level of HDL-
cholesterol between groups. Comparison is made of the total cholesterol/HDL-cholesterol ratio
between groups.


Elevation of HDL-cholesterol or reduction of the total cholesterol/HDL-cholesterol ratio
on administration of the compound is taken as indicative of the compound having the aforesaid
utility.
Atherosclerosis
The thoracic and abdominal aortas are removed intact, opened longitudinally along the
ventral surface, and fixed in neutral-buffered formalin after excision of samples from standard
sites in the thoracic and abdominal aorta for histological examination and lipid composition and
synthesis studies. After fixation, the whole aortas are stained with Sudan IV and pinned out flat,
and digital images are obtained with a TV camera connected to a computerized image analysis
system (Image Pro Plus; Media Cybernetics, Silver Spring, MD) to determine the percentage of
aortic surface involved with atherosclerotic lesions [Gerrity RG et al, Diabetes (2001) 50:1654-
65, Comhill IF et a\. Arteriosclerosis, Thrombosis, and Vascular Biology (1985) 5:415-26;
which disclosures are hereby incorporated by reference in their entirety]. Comparison is made
between groups of the percentage of aortic surface involved with atherosclerotic lesions.
Reduction of the percentage of aortic surface involved with atherosclerotic lesions on
administration of the compound is taken as indicative of the compound having the aforesaid
utility
Example 6
Receptor Binding Assay
In addition to the methods described herein, another means for evaluating a test
compound is by determining binding affinities to the RUP25 receptor. This type of assay
generally requires a radiolabelled ligand to the RUP25 receptor. Absent the use of known
ligands for the RUP25 receptor and radiolabels thereof, compounds of Formula (I) can be
labelled with a radioisotope and used in an assay for evaluating the affinity of a test compound to
the RUP25 receptor.
A radiolabelled RUP25 compound of Formula (I) can be used in a screening assay to
identify/evaluate compounds. In general terms, a newly synthesized or identified compound (i.e.,
test compound) can be evaluated for its ability to reduce binding of the "radiolabelled compound
of Formula (I)" to the RUP25 receptor. Accordingly, the ability to compete with the "radio-
labelled compound of Formula (I)" or Radiolabelled RUP25 Ligand for the binding to the
RUP25 receptor directly correlates to its binding affinity of the test compound to the RUP25
receptor.
ASSAY PROTOCOL FOR DETERMINING RECEPTOR BINDING FOR RUP25:
A. RUP25 RECEPTOR PREPARATION


293 cells (human kidney, ATCC), transiently transfected with 10 ug human RUP25
receptor and 60 ul Lipofectamine (per 15-cm dish), are grown in the dish for 24 hours (75%
confluency) with a media change and removed with 10 ml/dish of Hepes-EDTA buffer ( 20mM
Hepes + 10 mM EDTA, pH 7.4). The cells are centrifuged in a Beckman Coulter centrifuge for
20 minutes, 17,000 rpm (JA-25.50 rotor). Subsequently, the pellet is resuspended in 20 mM
Hepes + 1 mM EDTA, pH 7.4 and homogenized with a 50- ml Dounce homogenizer and again
centrifuged. After removing the supernatant, the pellets are stored at -80"C, until used in binding
assay. When used in the assay, membranes are thawed on ice for 20 minutes and then 10 mL of
incubation buffer (20 mM Hepes, 1 mM MgCI2, 100 mM NaCl, pH 7.4) added. The membranes
are vortexed to resuspend the crude membrane pellet and homogenized with a Brinkmann PT-
3100 Polytron homogenizer for 15 seconds at setting 6. The concentration of membrane protein
is determined using the BRL Bradford protein assay.
B. BINDING ASSAY
For total binding, a total volume of 50ul of appropriately diluted membranes (diluted in
assay buffer containing 50mM Tris HC1 (pH 7.4), lOmM MgCI2, and ImM EDTA; 5-50ug
protein) is added to 96-well polyproylene microtiter plates followed by addition of lOOul of assay
buffer and 50ul of Radiolabeled RUP25 Ligand. For nonspecific binding, 50 ul ofassay buffer
is added instead of lOOul and an additional 50ul of lOuM cold RUP25 is added before 50ul of
Radiolabeled RUP25 Ligand is added. Plates are then incubated at room temperature for 60-120
minutes. The binding reaction is terminated by filtering assay plates through a Microplate
Devices GF/C Unifilter filtration plate with a Brandell 96-well plate harvester followed by
washing with cold 50 mM Tris HC1, pH 7.4 containing 0.9% NaCl. Then, the bottom of the
filtration plate are sealed, 50ul of Optiphase Supermix is added to each well, the top of the plates
are sealed, and plates are counted in a Trilux MicroBeta scintillation counter. For compound
competition studies, instead of adding lOOul ofassay buffer, lOOul of appropriately diluted test
compound is added to appropriate wells followed by addition of 50 ul of Radiolabeled RUP25
Ligand.
C. CALCULATIONS
The test compounds are initially assayed at 1 and 0.1 u,M and then at a range of
concentrations chosen such that the middle dose would cause about 50% inhibition of a Radio-
RUP25 Ligand binding (i.e., 1CS0). Specific binding in the absence of test compound (Bo) is the
difference of total binding (BT) minus non-specific binding (NSB) and similarly specific binding
(in the presence of test compound) (B) is the difference of displacement binding (BD) minus non-
specific binding (NSB). lC5o is determined from an inhibition response curve, logit-log plot of %
B/B0 vs concentration of test compound.
K, is calculated by the Cheng and Prustoff transformation:


K, = ICso/(l +[L]/KD)
where [L] is the concentration of a Radio-RUP25 Ligand used in the assay and KD is the
dissociation constant of a Radio-RUP25 Ligand determined independently under the same
binding conditions.
D. ALTERNATIVE BINDING ASS,* Y PROCEDURE
3H-Nicotinic acid binding competition assay.
CHO-KI cells stably expressing the niacin receptor were used to make membrane for
binding analysis. Cells were grown to-80% confluence ingrowth medium (F-12 Kaighn's
modified medium (ATCC, #30-2004) containing 10%FBS (G1BCO, #10438-026), lmg/mlG418
(G1BCO, # 10131 -027) and IX Pen-Strep (Sigma P-0871), harvested by scraping, and
centrifuged at 12 000 X g, 4° Celsius, 10 minutes. Cell pellets were resuspendcd in harvest
buffer (20 mM HEPES, 10 mM EDTA, pH 7.4) and homogenized with 4X10 second bursts of a
12 mm Polytron homogenizer, setting 5. Lysate was centrifuged at 2 000 X g, 4°, 10 minutes to
remove unlysed cells and nuclei, and the resulting supernatant centrifuged at 39 000 X g, 4°, 45
minutes to pellet membranes. The resulting pellet was resuspended in wash buffer (20 mM
HEPES, 0.1 mM EDTA, pH 7.4), homogenized with 3 X 10 second bursts of a 12 mm Polytron,
setting 4, and re-centrif'uged at 39 000 X g, 4°, 45 minutes. The resulting pellet was resuspended
in wash buffer and stored in liquid nitrogen before use. The concentration of membrane proteins
in this preparation was determined using the Pierce BCA protein assay, with BSA as a standard.
Equilibrium binding of 3H-nicotinic acid was performed in 96-well polypropylene plates.
Reactions contained 140 u.1 membrane diluted in assay buffer (20 mM HEPES, pH 7.4, 1 mM
MgCI2, and 0.01% CHAPS; 15-30 j.ig membrane protein/assay), 20 u.1 test compounds diluted in
assay buffer (compound stocks were in 100% DMSO; final DMSO concentration in the assay
was 0.25%), and 40 u.1 250 nM tritiated niacin ([5, 6-3H] - nicotinic acid: American Radiolabeled
Chemicals, Inc.,20 u.M in ethanol; final ethanol concentration in each assay was 1.5%). Non-
specific binding was determined in the presence of 250 u,M unlabeled nicotinic acid. After
mixing at 3-4 hours ai room temperature, reactions were filtered through Packard Unifilter GF/C
plates using a Packard Harvester, and washed with 8 X 200 ul ice-cold binding buffer. Plates
were dried overnight and their backs sealed using PerkinElmer tape designed for GF/C plates. 40
u.1 PerkinElmer Microscint-20 scintillation fluid was added to each well, the tops sealed, and
plates analyzed in a Packard TopCount scintillation counter.
Caluclations were preformed as in C above.


Certain compounds of the invention have an EC50 in the 3H-nicotinic acid binding
competition assay within the range of about 10 to about 100 u.M. More advantageous
compounds of the invention have an EC5U value in this assay within the range of about I to about
10 |iM. Still more advantages compounds have an EC50 value in this assay of less than about 1
uM.
Example 7: Flushing via Laser Doppler
Procedure - Male C57BI6 mice (~25g) are anesthetized using I Omg/ml/kg Nembutal
sodium. When antagonists are to be administered they are co-injected with the Nembutal
anesthesia. After ten minutes the animal is placed under the laser and the ear is folded back to
expose the ventral side. The laser is positioned in the center of the ear and focused to an intensity
of 8.4-9.0 V (with is generally ~4.5cm above the ear). Data acquisition is initiated with a 15 by
15 image format, auto interval, 60 images and a 20sec time delay with a medium resolution. Test
compounds are administered following the 10th image via injection into the peritoneal space.
Images 1-10 are considered the animal's baseline and data is normalized to an average of the
baseline mean intensities.
Materials and Methods - Laser Doppler Pirimed Pimll; Niacin (Sigma); Nembutal (Abbott labs).
Example 8: Inhibition of Free Fatty-Acid Production, in vivo, in Catheterized Male
Sprague-Daly Rats
Non-esterified free-fatty acid (NEFA) assays were done on serum derived from live,
freely moving rats. Jugular vein catheters were surgically implanted into the jugular veins
and the animals were allowed to recover at least 48hr post surgery. Food was removed from
the animals approximately 16 hours prior to the assay. A draw of ~200ul blood was pulled
from the catheter and represents the baseline NEFA serum sample. Drug was administered
intra-peritoneally (IP) at various concentrations to individual rats and then ~200ul blood
draws were pulled from the catheter at the indicated time points for further NEFA analysis.
NEFA assays were performed according to the manufacturer's specifications (Wako
Chemicals, USA; NEFA C) and free fatty acid concentrations were determined via regression
analysis of a known standard curve (range of known free fatty acids). Data was analyzed
using Excel and PrismGraph.
Example 9:
The invention will now be illustrated by the following non-limiting examples in
which, unless stated otherwise:
(i) all operations were carried out at room or ambient temperature, that is, at a
temperature in the range 18-25°C;


(ii) evaporation of solvent was carried out using a rotary evaporator under
reduced pressure (4.5-30 mmHg) with a bath temperature of up to 50°C;
(iii) the course of reactions was followed by thin layer chromatography (TLC)
and/or tandem high performance liquid chromatography (HPLC) followed by mass
spectroscopy (MS), herein termed LCMS, and any reaction times are given for illustration
only;
(iv) the structure of all final compounds was assured by at least one of the
following techniques: MS or proton nuclear magnetic resonance ('H NMR) spectrometry, and
the purity was assured by at least one of the following techniques: TLC or HPLC;
(v) yields, if given, are for illustration only;
(vi) 'H NMR spectra were recorded on either a Bruker Avance-400 or a Varian
Unity or a Varian Inova instrument at 400 or 500 or 600 MHz using the indicated solvent;
when line-listed, NMR data is in the form of delta (5) values for major diagnostic protons,
given in parts per million (ppm) relative to residual solvent peaks (multiplicity and number of
hydrogens); conventional abbreviations used for signal shape are: s. singlet; d. doublet
(apparent); t. triplet (apparent); m. multiple!; br. broad;
(vii) MS data were recorded on a Waters Micromass unit or API 150EX,
interfaced with a Hewlett-Packard (Agilent 1100) or Shimadzu (LC-10AD VP) HPLC
instrument, and operating on MassLynx/OpenLynx or Analyst 1.2 software; electrospray
ionization was used with positive (ES+) or negative ion (ES-) detection; the method for
LCMS ES+ was 1-2 mL/min, 10-95% B linear gradient over 5.5 min (B - 0.05% TFA-
acetonitrile, A = 0.05% TFA-water), and the method for LCMS ES- was 1-2 mL/min, 10-95%
B linear gradient over 5.5 min (B - 0.1% formic acid - acetonitrile, A = 0.1% formic acid -
water), Waters XTerra CI 8 - 3.5 um - 50 x 3.0 mmID and diode array detection;
(viii) the purification of compounds by preparative reverse phase HPLC
(RPHPLC) was conducted on cither a Waters Symmetry Prep CI8 - 5 um - 30 x 100 mmID,
or a Waters Atlantis Prep dCI8-5 um-20 x 100 mmID; 20 mL/min, 10-100% B linear
gradient over 15 min (B = 0.05% TFA-acetonitrile, A - 0.05% TFA-water), and diode array
detection;
(ix) the automated purification of compounds by preparative reverse phase HPLC
was performed on a Gilson system using a YMC-Pack Pro CI 8 column (150 x 20 mm i.d.)
eluting at 20 mL/min with 0 - 50% acetonitrile in water (0.1% TFA);
(x) the purification of compounds by preparative thin layer chromatography
(PTLC) was conducted on 20 x 20 cm glass prep plates coated with silica gel, or centrifugal
chromatography on a chromatotron using glass rotors coated with silica gel, both
commercially available from Analtech;
(xi) column chromatography was carried out on a silica gel column using
Kieselgel 60, 0.063-0.200 mm (Merck).


(xii) microwave irradiations were carried out using the Smith Synthesizer
(Personal Chemistry).
(xiii) chemical symbols have their usual meanings; the following abbreviations
have also been used v (volume), w (weight), b.p. (boiling point), m.p. (melting point), L
(litre(s)), mL (millilitres), g (gram(s)), mg (milligrams(s)), mol (moles), mmol (millimoles),
eq or equi v (equivalent(s)), IC50 (molar concentration which results in 50% of maximum
possible inhibition), EC50 (molar concentration which produces 50% of the maximum
possible efficacy or response), uM (micromolar), nM (nanomolar).
The following examples are provided so that the invention might be more fully
understood. They should not be construed as limiting the invention in any way.
Example 9.1: 3-(2H-Tetrazol-5-yl)-l,4,5,6-tetrahydro-cyclopentapyrazole (Compound 1).

Method A: Preparation of Compound 1.
l,4,5,6-Tetrahydro-cyclopentapyrazole-3-carbonitrile (0.022 g, 0.165 mmol) and sodium
azide (0.086 g, 1.30 mmol) were taken up in DMF (3 cm3) at heated under microwave irradiation
to 175°C for 20 minutes. The solution was cooled to room temperature, filtered and the filtered
solid washed with ethyl acetate. The combined solutions was added to saturated aqueous sodium
bicarbonate (20 cm3) and washed with ethyl acetate. The aqueous layer was acidified to pH 1
with the addition of 1M aqueous hydrochloric acid and extracted into ethyl acetate. The ethyl
acetate washes were combined and solvent removed under reduced pressure, the resulting solid
purified by preparative HPLC to give 3-(2H-tetrazol-5-yl)-l,4,5,6-tetrahydro-
cyclopentapyrazole as a white solid (0.012 g, 0.068 mmol, 41%). 'll NMR 5 (CD,OD): 2.88 (t-
like, 2H, J=7.0), 2.82 (t-like, 2H, J=7.3), 2.64 (quintet-like, 2H, J=7.1); m/z(ES+): 177 [M+H]+.
The intermediate l,4,5,6-Tetrahydro-cyclopentapyrazole-3-carbonitrile was prepared
using the following procedure.
Step A: I,4,5,6-Tetrahydro-cyclopentapyrazole-3-carboxylic acid ethyl ester

Cyclopentanone (lO.Og, 118.9 mmol) was taken up in absolute ethanol (30 cm3) and
sodium ethoxide (53 cm3,21% in ethanol, 143 mmol) was added. The resulting solution was

stirred under argon for 10 minutes, then diethyl oxalate(19.1 g, 131 mmol) added. Further
ethanol (10 cm1) was added and the solution heated at 75°C for 3 hours and cooled to room
temperature. Hydrazine hydrochloride (8.15 g, 119 mmol), taken up in water (20 cm3) was added
and the solution heated to 75°C overnight. Solvent was removed under reduced pressure and the
resulting taken up in ethyl acetate (200 cm3) and washed with water (200 cm3), dried (Na2S04),
filtered and solvent removed under reduced pressure to give 1,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid ethyl ester as an off white solid (16.16 g, 90.0 mmol,
76%). SH (C^OD): 4.34 (q, 2H, J=7.1, OCH2CH3), 2.78 (t like, 2H, J=7.0), 2.72 (br s, 2H), 2.49
(brs,2H), 1.36(t,3H,J= 7.l,OCH2CH3). m/z(ES*): 181 [M+H]+.
Step B: l,4,5,6-Tetrahydro-cyclopentapyrazole-3-carboxylic acid amide.

l,4,5,6-Tetrahydro-cyclopentapyrazole-3-carboxylic acid ethyl ester (0.808g, 4.48
mmol) was taken up in methanolic ammonia (ca 7 M, 12 cm3) and stirred overnight at 95°C. The
resulting solution was chilled and the precipitated l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carboxylic acid amide collected by vacuum filtration as a white crystalline solid (0.438g, 2.90
mmol, 65%). 6H (CD3OD): 2.79 (t like, 2H, J=6.9), 2.73 (t like, 2H, J=7.3), 2.55 (br s, 2H); m/z
(ES+): 152 [M+H]'.
Step C: 1,4,5,6-Tetrahydro-cycIopentapyrazole-3-carbonitrile.

1,4,5,6-Tetrahydro-cyclopentapyrazole-3-carboxylic acid amide (0.210 g, 1.39 mmol)
was added to anhydrous acetonitrile (12 cm3), heated to 80°C and sodium chloride (2.0 g, 34
mmol) added. After 15 minutes phosphorus oxychloride (0.128 g, 0.83 mmol) was added and the
solution heated to 80°C overnight, cooled, filtered, and the collected solid washed with
acetonitrile. Solvent was removed from the combined solutions under reduced pressure and the
resulting solid purified by preparative HPLC to give l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carbonitrile as a deep purple coloured solid (0.031 g, 0.23 mmol, 17%). 8H (CD3OD): 2.79 (t
like, 2H, J=7.3), 2.73 (t like, 2H, J=7.1), 2.65-2.55 (m, 2H); m/z (ES'): 134 [M+Hf.
Method B: Preparation of Compound 1.


Air was bubbled through a stirring solution of l-benzyl-3-(2H-tetrazol-5-yl)-l,4,5,6-
tetrahydro-cyclopentapyrazole(1.92 g, 7.21 mmol) and KO/-Bu (65 mLof'a IM solution in
THF) in DMSO (50 mL) for a period of 2.0 h. The reaction was acidified to pH = 2 by the
addition of HCI (3M aq). The mixture was filtered and the filtrate was concentrated in vacuo to
remove volatiles. The material was purified by reverse-phase HPLC: Phenomenex® Luna CI 8
column (10 u, 250 x 50 mm), 5% (v/v) CH3CN (containing 1% v/v TFA) in H20 (containing 1%
v/v TFA) gradient to 50% H20,60 ml/min, A. = 214 nm. The product was further purified by
loading material on a Varian BondElut®60 mL, lOg SCX cartridge. MeOH (150 mL) was
passed through the column to remove unbound impurities. The product was then eluted by
passing a solution of 2JVNH3 in MeOH (150 mL) through the column. Concentration of the
eluant yielded the ammonium salt of Compound 1 (947 mg, 5.38 mmol, 75% yield) as a white
solid. 'H NMR (ammonium salt, 400MHz, CDjOD): 6 2.88 (2H, t, J = 6.8 Hz), 2.74 (2H, t, J =
6.8 Hz), 2.52 (2H, quin, J = 6.8 Hz). HPLC/MS: Discovery® Cl 8 column (5u, 50 * 2.1 mm), 5%
v/v CH3CN (containing 1% v/v TFA) in H20 (containing 1% v/v TFA) gradient to 99% v/v
CH3CN in ILO, 0.75 mL/min, r, = 1.22 min, ESI' ■= 177.3 (M + H). Anal Calcd for C->H8N6
(neutral compound): C, 47.72; H, 4.58. Found: C, 47.27; H, 4.16. Anal Calcd for C7HMN7
(ammonium salt): C, 43.51; H, 5.74. Found: C, 42.94; H, 5.30.
The intermediate I -benzyl-3-(21 l-tetrazol-5-yl)-l ,4,5,6-tetrahydro-cyclopentapyrazole
was prepared using the following procedure.
Step A: Preparation of l-Benzyl-l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carboxylic acid amide and 2-Benzyl-2,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic
acid amide.

To a stirring solution of 1,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic acid amide
(2.57 g, 17.0 mmol) in DMF (34 mL) at 25 °C was added K2CO, (5.87 g, 42.5 mmol) followed
by benzyl bromide (4.36g g, 25.5 mmol). The reaction was stirred at ambient temperature for 16
h at which time the mixture was diluted with EtOAc (75 mL) and filtered. The filtrate was
washed with H20 (100 mL) and the aqueous phase was back-extracted with EtOAc (75 mL) and
CH2C12(75 ml,). The combined organic extracts were dried over MgS04, filtered, and

concentrated m vacuo. Purification by silica gel chromatography (50% EtOAc in hexanes
gradient to 95% EtOAc in hexanes) gave 2-benzyl-2,4,5,6-tetrahydro-cyclopentapyrazole-3-
carboxylic acid amide (739 mg, 3.07 mmol, 18% yield) isolated as a white solid followed by 1-
benzyl-!,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylicacid amide (3.24 g, 13.4 mmol, 79%
yield) isolated as a white solid.
l-Bcnzyl-l,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylicacid amide.
'11 NMR (400 MHz, CDC1,): 6 7.37-7.30 (3H, m), 7.19 (2H, m), 6.67 (1H, bs), 5.34 (1H, bs),
5.19 (2H,s), 2.82 (2H, m), 2.51 (4H, m). "C APT NMR (100 MHz, CZ3CI,): Sup: 164.8,
155.2, 139.0, 136.0, 129.5, 55.3,31.2,24.1; down: 129.0, 128.3, 127.8. HPLC/MS: Alltech®
Prevail CI 8 column (5u, 50 * 4.6 mm), 5% v/v CH,CN (containing 1% v/v TFA) in H20
(containing 1% v/v TFA) gradient to 99% v/v CH3CN in H20, 3.5 mL/min, t, = 2.13 min, ESf =
242.2 (M + H).
2-Benzyl-2,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylicacid amide.
'H NMR (400 MHz, CDClj): 5 7.34-7.21 (5H, m), 5.76 (2H, s), 5.70-5.38 (2H, bs), 2.78 (4H,
m), 2.49 (2H, m). IJC APT NMR (100 MHz, CZ)CI3): 8 up: 161.9, 160.1, 138.3, 128.3, 127.1,
55.1,29.9, 24.8, 24.7; down: 128.6, 128.0, 127.6. HPLC/MS: Alltech® Prevail C18 column (5u,
50 x 4.6 mm), 5% v/v CH3CN (containing 1% v/v TFA) in H20 (containing 1% v/v TFA)
gradient to 99% v/v CH3CN in H20, 3.5 mL/min, t, = 1.98 min, ESF = 242.1 (M + H).
Step B: Preparation of l-Benzyl-3-(2H-tetrazol-5-yl)-l,4,5,6-tetrahydro-
cyclopentapyrazole

To a solution of 1 -benzyl-1,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic acid
amide (3.02 g, 12.53 mmol) in DMF(25 mL) at rt was added thionyl chloride (1.94 g, 16.3
mmol). The reaction was stirred for 18 h at which time NaHCOs (sat. aq., 6 mL) was added to
quench excess thionyl chloride. The mixture was diluted with EtOAc (150 mL) and washed
sequentially with NaHC03 (sat. aq., 100 mL)and brine (100 mL). The aqueous washes were
back-extracted with EtOAc (2* 100 mL) and the combined organics were dried over MgS04,
filtered, and concentrated m vacuo to yield a crude yellow oil.
The concentrate was dissolved in DMF (20 mL) and placed in a heavy walled sealed
reaction vessel at which time to which ZnBr3 (4.70 g, 18.0 mmol) and NaN3 (2.73 g, 42.0 mmol)
were added sequentially. The vessel was sealed and heated to 120 °C for 18 h. The mixture was
cooled to rt and HC1 (3M aq., 2 mL) was added and stirring was continued for 5 min. The
mixture was diluted with EtOAc (150 mL) and washed with HCI (1M, aq., 100 mL). The

organics were dried over MgSC>4, filtered, and concentrated. Purification by silica gel
chromatography (50 : 50 : 0.2, hexanes : EtOAc : AcOH gradient to 100 : 0.2, EtOAc : AcOH)
gave 1 -benzyl-3-(2H-tetrazol-5-yl)-l ,4,5,6-tetrahydro-cyclopentapyrazole (2.06 g, 7.74 mmol,
62% yield) as a white solid. 'H NMR (400MHz, CD£)D): 6 7.36-7.25 (5H, m), 5.30 (2H, s),
2.84 (2H,t,.7=6.4 Hz), 2.62-2.56 (4H,m). I5C APTNMR (100 MHz, CD.OD): 8 up: 153.8,
151.9,137.6, 131.5, 128.9, 55.8, 31.9,24.8, 24.6; down: 129.9,129.1,129.0. HPLC/MS:
Discovery®CI 8 column (5u, 50 * 2.1 mm), 5% v/v CH3CN (containing 1% v/v TFA) in H20
(containing 1% v/v TFA) gradient to 99% v/v CH3CN in H20,0.75 mL/min, I, = 2.18 min, ESI+
= 267.1 (M + H).
Method C: Preparation of Compound I.

To a solution of l-benzyl-3-(2H-tetrazol-5-yl)-l,4,5,6-tetrahydro-cyclopentapyrazole
(59.4 g, 223 mmol) in 10% formic acid/MeOH (vol/vol, 900 mL) was added palladium black
(39.8g, 374 mmol). The mixture was mechanically stirred under N2 atmosphere for 24 h. The
reaction was filtered and concentrated. The product was further purified and converted to the
ammonium salt by the following by loading material (as a solution in McOH) on to a column
containing Bondesil SCX SPE resin (750 g). The column was flushed with MeOH (2.0 L) to
remove unbound impurities The product was eluted using 2jVNH3/MeOH (approx. 1.5 L).
Upon concentration the ammonium salt of the tetrazole (39.3 g, 203 mmol, 91% yield) was
obtained as a white solid.
The intermediate I -benzy l-3-(2H-tetrazol-5-yl)-1,4,5,6-tetrahydro-cyclopentapyrazole
was prepared using the following procedure.
Step A: Preparation of l,4,5,6-Tetrahydro-cyclopentapyrazole-3-carboxylic acid
ethyl ester.

To a solution of cyclopentanone (42.0 g, 0.50 mol) and diethyl oxalate (73.1 g, 0.50 mol)
in EtOH (2.5 L) at rt under N2 was added a solution of KOBu in THF (500 mL of a I M
solution, 0.50 mol) over 0.5 h via an addition funnel. The reaction was stirred for 3.5 h at which
time the flask was cooled to 0 °C. Hydrazine hydrochloride (37.6 g, 0.55 mol) in H20 (250 mL)
was added via addition funnel over 0.5 h. The reaction was warmed to rt and stirred for 16 h.

The volatiles were removed in vacuo and the resulting solid was washed with NaHC03 (sat. aq.,
500 mL) and H20 (500 mL). Further concentration in vacuo gave pure 1,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid ethyl ester (63.6 g, 0.35 mol, 71% yield) as a yellow solid.
Step B: Preparation of l,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic acid
amide.

l,4,5,6-Tetrahydro-cyclopentapyrazole-3-carboxylic acid ethyl ester (63.5 g, 0.35 mmol)
was dissolved in a solution of 7jVNH3/MeOH (1.0 L). The solution was divided into Tour equal
portions each of which was transferred to 350 mL heavy-walled sealed reaction vessel. The
vessels were heated to 95 °C and stirred for 20 h. The reactions were cooled to rt at which time a
solid precipitated. The solution was filtered and the solid was washed with NaOll (l/Vaq., 200
mL) giving pure 1,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic acid amide (42.0 g, 0.20
mol, 80% yield) as a white solid.
Step C: Preparation of l-Benzyl-l,4,5,6-tetrahydro-cydopentapyrazole-3-
carboxylic acid amide and 2-Benzyl-l,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic
acid amide.

To a solution of l,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic acid amide (41.5 g,
275 mmol) in THF (460 mL) at rt was added a solution of NaOH (5/Vaq., 110 mL, 0.54 mol).
After stirring for 5 min benzyl bromide (49.2 g, 0.29 mol) was added and the reaction was stirred
for 16 h. The volatiles were removed in vacuo and the resulting solid was washed with H20 (3 x
250 mL). Further concentration gave regioisomers of I-benzyl-1,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid amide and 2-benzyl-l ,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid amide (65.3 g, 270 mmol, 98% yield) as a 20 : 1 mixture
and was used without separation).
Step D: Preparation of l-Benzyl-3-(2H-tetrazol-5-yl)-l,4,5,6-tetrahydro-
cyclopcntapyrazole


A flask equipped with a drying tube under N2 atmosphere was charged with anhydrous
DMF (250 mL). The flask was cooled to 0 °C and thionyl chloride (36.7 g, 309 mmol) was
added via syringe over a period of 5 min. After stirring for an additional 10 min, a solution of 1-
benzyl-1,4,5,6-tetrahydro-cyclopentapyrazole-3-carboxylic acid amide (67.7 g, 281 mmol) in
DMF (310 mL) was added over 5 min using an addition funnel. The mixture was slowly
warmed to rt and stirred for 16 hr. NaHC03 (sat. aq., 100 mL) was added and the mixture was
stirred for 10 min. The volatiles were removed in vacuo and the residue was diluted with EtOAc
(700 mL) and NaHC03 (sat. aq., 700 mL). The layers were separated and the aqueous phase was
back-extracted with EtOAc (400 mL). The combined organics were washed with NaHC03 (sat.
aq., 600 mL) and brine (600 mL), dried over MgS04, filtered, and concentrated to give 63.1 g of
nitrile as a brown solid.
To a solution of the nitrile (from above) in DMF (560 mL) was added ZnBr2 (95.6 g,
425 mmol) followed by NaN3 (55.2 g, 849 mmol). The mixture was heated to 120°C and stirred
for 14 h. The reaction was cooled to rt and the DMF was removed in vacuo. HCI (2/Vaq., 800
mL) was added and the mixture was stirred for 15 min followed by filtration. The solid was
added to a biphasic mixture of EtOAc (500 mL) and HCI (5/Vaq., 300 mL) and stirred for 0.5 h.
The solution was filtered and the layers separated. The remaining solid was again treated with
EtOAc and HCI (5N aq.) as described above and this process (stir, filter, separate) was repeated
until all solid material was dissolved. The combined organic filtrates were concentrated to give
1 -benzyl-3-(2H-tetrazol-5-yl)-l ,4,5,6-tetrahydro-cyclopentapyrazole (61.0 g, 229 mmol, 81%
yield from the amide) as a light brown solid.
Example 9.2: 3-(l H-Tetrazol-5-yl)-2,6-dihydro-4H-thieno|3,4-c|pyrazole (Compound 2).

Compound 2 was prepared in a similar manner as described in Example 9.1, and was
characterized by NMR and MS;'H MVIR (400MHz, McOD). (400 MHz,CD3OD) £4.11 (do, J
= 4.0, 2.2 Hz, 2 H), 4.03 (dd, J = 3.6, 2.2 Hz, 2 H). HPLC/MS: Waters® YMC ODS-A CI 8
column (5u, 50 * 4.6 mm), 5% v/v CH3CN (containing 1% v/v TFA) in H20 (containing 1% v/v
TFA) gradient to 99% v/v CH3CN in H20, 3.5 mL/min, l, = 1.27 min, EST = 194 (M -r H).

Example 9.3: 6-Methyl-3-(l H-tetrazol-5-yl)-2,6-dihydro-4H-furo|3,4-c|pyrazole
(Compound 3).

Compound 3 was prepared in a similar manner as described in Example 9.1, a separation
by column chramoatography of the regioisomers was performed after the formation of the
pyrazole.
Compound 3 was characterized by NMR and MS; 'H INMR (400MHz, DMSO): £5.20 (m, 1H),
4.94 (dd, 7 = 34.7, 10.3 Hz, 2 H), 1.39 (d, J = 4.4 Hz, 3 H). HPLC/MS: Alltech09 Prevail C18
column (5p, 50 * 4.6 mm), 5% v/v CH3CN (containing 1% v/v TFA) in H20 (containing 1% v/v
TFA) gradient to 99% v/v CH3CN in H20, 3.5 mL/min, t, = 1.03 min, EST = 192 (M + H).
Example 9.4: 3-(lH-Tetrazol-5-yl)-l,4-dihydro-cyclopentapyrazole (Compound 4) and 3-
(lH-Tetrazol-5-yl)-l,6-dihydro-cyclopentapyrazole (Compound 5).

Compound 9.4A
A solution of Compound 9.4A, as an isomeric mixture, (50 mg, 0.38 mmol), sodium
azide (86.5 mg, 1.33 mmol) and zinc bromide (300 mg, 1.33 mmol) in DMF (2 mL) was
irradiated under microwave at 200 °C for 6 hours. After cooling to room temperature, the
reaction mixture was treated with a 2 N HC1 solution, extracted with EtOAc, washed with H20
and concentrated in vacuo. HPLC separation (CI 8 column, 5 to 99 % CH3CN in H20) afforded
40.3 mg (61 %) of the desired product as a 2:1 mixture ofolefinic isomers. LC-MS m/z 175
(M+l); 'H NMR (400 MHz, DMSO-d6) 5 6.94 (m, 0.5 H), 6.87 (m, 1 H),6.76(m, 1 H), 6.40 (m,
0.5 H), 3.35 (m, 3 H).
The isomers were separated by reverse-phase HPLC: Phenomenex® Luna C18 column
(10 u, 250 x 21.2 mm), 5% (v/v) CH3CN (containing 1% v/v TFA) in H20 (containing 1% v/v
TFA) gradient to 70% H20, 20 ml/min, X. = 280 nm.
Alternatively the isomers were separated by normal-phase HPLC: Dynamax Micorsorb
Si (prep) column (8 u, 250 * 10 mm), 80% (v/v) EtOAc (containing 2% v/v AcOH) in hexanes
(containing 2% v/v AcOH) gradient to 99% EtOAc, 7.5 ml/min, X = 280 nm.
The order of isomer eluiion is the same for both normal- and reverse-phase columns
Isomer I (High Rf isomer):

'H NMR (400MHZ, MeOD): 6 6.79 (2H, m), 3.42 (2H, m). HPLC/MS: Discovery®CI8 column
(5u, 50 x 2.1 mm), 5% v/v CH3CN (containing 1% v/v TFA) in H20 (containing 1% v/v TFA)
gradient to 99% v/v CH3CN in H20,0.75 mL/min, I, = 1.10 min, ESI+ = 174.9 (M + H).
Isomer 2 (Low Rf isomer):
'H NMR (400MHz, MeOD): 8 6.98 (1H, m), 6.44 (lH,m), 3.33 (2H, m). HPLC/MS:
Discovery®Cl8 column (5u, 50 * 2.1 mm), 5% v/v CH3CN (containing 1% v/v TFA) in H20
(containing 1% v/v TFA) gradient to 99% v/v CH3CN in H20,0.75 mL/min, t, = 1.11 min, EST
= 175.1 (M + H).
The intermediate Compound 9.4A, as an isomeric mixture, was prepared using the
following steps:
Step A: Preparation of 2,6-Dihydro-cyclopentapyrazole-3-carboxylic acid ethyl
ester and 2,4-Dihydro-cyclopentapyrazole-3-carboxylic acid ethyl ester (mixture).

Compound 9.4B was prepared from the corresponding ketone using a similar method as
described herein for the preparation ofpyrazole esters (see Example 14.2). A solution of
Compound 9.4B (2.0 g, 8.19 mmol) in phenyl ether (25 mL) was heated at reflux (250 - 260 °C)
under nitrogen for 2 hours.
After cooling down the solution to room temperature, it was loaded on a Si02 column,
flushed with DCM to push out the phenyl ether, and eluted with EtOAc/Hex (1/3) to afford 1.05
g (72%) of Compound 9.4Casa mixture of olefinic isomers. LC-MS m/z I79(M+I).
Step B: Preparation of 2,6-Dihydro-cyclopentapyrazole-3-carboxylic acid amide
and 2,4-Dihydro-cyclopentapyrazole-3-carboxylic acid amide (mixture).

Compound 9.4C, as an isomeric mixture, (1.0 g, 5.61 mmol) was dissolved in smallest
amount of dioxane ( tightly sealed container. The solution was stirred at room temperature for 24 hours and
concentrated in vacuo to afford Compound 9.4D, as an isomeric mixture, as a solid in
quantitative yield. LC-MS m/z 150(M+I).

Step C: Preparation of 2,6-Dihydro-cydopentapyrazole-3-carbonitrile and 2,4-
Dihydro-cyclopentapyrazole-3-carbonitrile (mixture).

To a suspension of Compound 9.4D, as an isomer mixture, (0.80 g, 5.36 mmol) and
potassium carbonate (0.445 g, 3.22 mmol) in acetonitrile (30 mL) was added POCI3 (0.785 mL,
8.58 mmol) at room temperature. The reaction mixture was heated at reflux for 2 hours. After
concentration in vacuo, the residue was diluted with EtOAc (150 mL), washed with H20 and
brine, dried (Na:S04), and concentrated to afford 141 mg (20 %) ofCompound 9.4A as an
isomer mixture. LC-MS m/z 132 (M+l).
Example 9.5: 3-(l H-Tetrazol-5-yl)-2,6-dihydro-4H-furo|3,4-c|pyrazole (Compound 6).

Compound 6 was prepared in a similar manner as described in Example 9.1, and was
characterized by NMR and MS; LC-MS m/z 179 (M+l); 'H NMR (400 MHz, CD3OD) 8 5.07 (t,
7-2.2 Hz, 2 H), 4.92 (t, J = 2.2 Hz, 2 H).
Example 9.6: 5-Ethyl-3-(lH-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 7).
1 1
Compound 7 was prepared in a similar manner as described in Example 9.1, and was
characterized by NMR and MS; 'H NMR (MeOD, 400 MHz): 8 3.07 (I H, dd, J= 14.8, 7.6 Hz),
2.94-2.82 (2H,m), 2.51 (1H, dd,7= 15.2, 6.8 Hz) 2.41 (IH,dd,./ = 13.6,5.6 Hz), 1.6(2H,m),
1.02 (3H, t, J = 7.2 Hz). HPLC/MS: Discovery®CI 8 column (5u, 50 * 2.1 mm), 5% v/v
CH,CN (containing 1% v/v TFA) in H30 (containing 1% v/v TFA) gradient to 99% v/v CH,CN
in H,0, 0.75 mL/min, I, = 1.42 min, ESf - 205.2 (M + H).

Example 9.7: Preparation of Intermediate l-Benzyl-5-hydroxy-l,4,5,6-tetrahydrocyclo-
pcnta|c|pyrazole-3-carbonitrile.

Step A: Preparation of l-BenzyI-l,6-dihydro-cyclopentapyrazole-3-carboxylic acid
ethyl ester and l-Benzyl-l,4-dihydro-cyclopentapyrazole-3-carboxylic acid ethyl ester
(mixture).

To a solution of the pyrazole (Compound 9.4C, see Example 9.4, Step A, 2.0 g, 11.22
mmol) in anhydrous THF (100 mL) was added benzyl bromide (5.36 mmol, 44.88 mmol)and
NaOH (1.79 g, 44.88 mmol). After stirring at room temperature for 1 hour, the reaction was
quenched with IN HC1 (100 mL). The resulting mixture was extracted with ethyl acetate,
washed with 1N HCI, saturated NaHC03 solution, brine and dried over anhydrous Na2S04. The
solution was filtered and concentrated in vacuo. This material was purified on the biotage flash
40M column (SiOi) using 30% ethyl acetate-hexanes. A colorless oil was obtained. LC-MS:
3.22min;(M+Na)=291.l.
Step B: Preparation of l-Benzyl-l,6-dihydro-cyclopentapyrazole-3-carboxylic acid
and l-Benzyl-l,4-dihydro-cyclopentapyrazole-3-carboxylic acid (mixture).

To a solution of the intermediate from step A (3.55 g, 13.23 mmol) in 1:1 THF/MeOH
(40 mL) was added NaOH solution (5N, 3.9 mL, 20 mmol). After 3 hours at room temperature,
the reaction was quenched by adding IN HCI (22 mL). The aqueous layer was extracted with
ethyl acetate (3X) dried over anhydrous Na2S04, filtered and concentrated in vacuo. A yellow
solid was obtained which was used in the next step without any further purification. LC-MS:
2.62 min;(M+H)=241.1.
Step C: Preparation of l-Benzyl-l,6-dihydro-cyclopentapyrazole-3-carboxylic acid
2,5-dioxo-pyrrolidin-l-yl ester and l-Benzyl-1,4-dihydro-cyclopentapyrazole-3-carboxylic
acid 2,5-dioxo-pyrrolidin-l-yl ester (mixture).


To a solution of the intermediate from step B (3.17 g, 13.23 mmol) in CH2C12(200 mL)
was added A'-hydroxy succinimide (3.04 g, 26.46 mmol) followed by EDC (5.07 g, 26.46 mmol).
After stirring the reaction mixture at room temperature for 18 hours, it was concentrated in
vacuo. The residue was diluted with ethyl acetate (200 mL), washed with saturated NaHC03
solution and brine. The organic layer was dried over anhydrous Na2S04) filtered and
concentrated in vacuo. A yellow solid was obtained. LC-MS: 2.99 min; (M+H)=338.1.
Step D: Preparation of l-Benzyl-l,6-dihydro-cyclopentapyrazole-3-carboxylic acid
amide and 1 -Benzyl-l,4-dihydro-cyclopentapyrazole-3-carboxylic acid amide (mixture).

To a solution of the intermediate from step C (4.45 g, 13.22 mmol) in 1,4-dioxane (150
mL) was added NH4OH( 14.8 N, 10.0 eq, 9.1 mL). A precipitate formed immediately. After
stirring at room temperature for 15 minutes the reaction mixture was filtered through a sintered
funnel and the precipitate washed with 1,4-dioxane. The filtrate was concentrated in vacuo to
give a yellow solid. LC-MS: 2.55 min; (M+H)=240.l.
Step E: Preparation of l-Benzyl-l,6-dihydro-cyclopcntapyrazole-3-carbonitrile
and l-Benzyl-l,4-dihydro-cyclopentapyrazole-3-carbonitrile (mixture).

To a solution of the intermediate from step D in anhydrous DMF (50 mL) was added
cyanuric chloride (2.33 g, 13.2 mmol). After stirring at room temperature for 15 minutes the
reaction was quenched by pouring into water (100 mL). The resulting mixture was extracted
with ethyl acetate, washed with saturated NaHCOj, brine, dried over anhydrous Na2S04, filtered
and concentrated in vacuo. The residue was purified on the biotage flash 40 M column (SiO?)
using 20 % ethyl acetate-hexanes. A white solid was obtained. LC-MS: 3.22 min;
(M+H)=222.2.
Step F: Preparation of l-Benzyl-5-hydroxy-I,4,5,6-tetrahydro-cyclopentapyrazo!e-
3-carbonitrile.


To a solution of the intermediate from step E (0.95 g, 4.29 mmol) in anhydrous THF (40
mL) cooled to 0 °C under a N2 atmosphere was added Borane-THF (23 mmol, 5.36 eq, 1.0 M
solution). The reaction was warmed to room temperature and stirred for 1 hour. The reaction
was then cooled to 0 °C. Water was added (3 mL) followed by NaOH (4.29 mmol, 1.43 mL, 3N)
and H20: (12.88 mmol, 1.32 mL, 30% solution in water). After heating the reaction at 50 °C for
30 minutes, it was cooled to room temperature and quenched by the addition of water. The
resulting mixture was extracted with ethyl acetate (3X). The organic layer was dried over
anhydrous Na2S04 filtered and concentrated in vacuo. The residue was purified by flash
chromatography using 30 % ethyl acetate-hexanes to give a 1:1 mixture of the C-5 and C-6
alcohols.
Less polar isomer (C-6 alcohol, Compound 17) 'H NMR (500 MHz, CDCI3): 6 7.2 (m,
5H), 5.35 (d,.7=14.9 Hz, 1H), 5.31 (d,7= 14.6 Hz, 1H), 4.99 (dd,7=3.4, 6.9 Hz, lH),2.9(m,
2H),2.6(m, lH),2.35(m, 1H). LC-MS: 2.76 min;(M+H)=240.l.
More polar isomer (C-5 alcohol) 'H NMR (500 MHz, CDC1,): 5 7.4-7.2 (m, 5H), 5.28
(d, 7=14.8 Hz, 1H), 5.25 (d, 7= 14.9 Hz, 1H), 5.01 (m, 1H), 3.13 (dd, 7=6.4, 15.8 Hz, 1H),2.89
(dd, 7=6.6, 16.2 Hz, IH), 2.68 (dd, 7=3.7, 16.0 Hz, 1H), 2.52 (dd, 7=3.4, 162Hz, 1H). LC-MS:
2.60min;(M+H)=240.1.
Example 9.8: Preparation of Intermediate Trifluoro-methancsulfonic acid l-benzyl-3-
cyano-l,6-dihydro-cyclopentapyrazol-5-yl ester and Trifluoro-methanesulfonic acid I-
benzyl-3-cyano-l,4-dihydro-cyclopentapyrazol-5-yl ester as a regio-isomeric mixture.

Step A: Preparation of (4-Ethoxy-2-oxo-cyclopent-3-enyl)-oxo-acetic acid tert-
butyl ester.


To a solution, of 3-ethoxy cyclopentenone (2.12 3, 16.82 mmol) in anhydrous THF (40
mL) cooled to -78 °C under a nitrogen atmosphere was added lithium diisopropyl amide (12 mL,
24 mmol, 2.0 M in THF). After 15 minutes, a solution of dwert-butyl dioxalate (3.73 g, 18.5
mmol) in THF (15 mL) was added. The reaction mixture was stirred at -78 °C for 15 minutes
and then warmed to -20 "C and stirred for an additional 15 minutes. The reaction was quenched
with IN HCI (40 mL) and extracted with ethyl acetate (3X). The organic layer was washed with
brine, dried over anhydrous Na2S04) filtered and concentrated in vacuo. The residue was
purified by flash chromatography (Si02) using 35% ethyl acetate-hexanes to give the desired
product (2.53 g) as an off-white solid.
Step B: Preparation of l-8enzyl-5-oxo-l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carboxylic acid tert-butyl ester.

To a solution of the intermediate from step A (2.15 g, 8.45 mmol) in cthanol (100 mL)
was added benzyl hydrazine hydrochloride (1.8 g, 9.22 mmol) and HO Ac (10 mL). The reaction
mixture was stirred at room temperature for 16 hours and then refluxed at 70 °C for 30 minutes.
The reaction was cooled to room temperature and concentrated in vacuo. The residue was
dissolved in ethyl acetate and washed with water, saturated NaHC03, and brine. The organic
layer was dried over anhydrous Na2S04 filtered and concentrated in vacuo. The residue was
purified by flash chromatography (Si02) using 30 % ethyl acetate-hexanes to give the desired
product (1.64 g) a brown oil.
Step C: Preparation of l-Benzyl-5-oxo-l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carboxylic acid.

To a solution of the intermediate from step, B (1.64 g, 5.25 mmol) in dichloromethane
(20 mL) was added trifluroacetic acid (20 mL) and the resulting solution stirred at room
temperature for 4 hours. The reaction mixture was concentrated in vacuo and azeotroped with
toluene (3X). This material was carried on to the next step without any further purification.
Step D: Preparation of l-Benzyl-5-oxo-l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carboxylic acid 2,5-dioxo-pyrrolidin-l-yl ester.


To a solution of the intermediate from step C (1.34 g, 5.25 mmol) in CH2C12(50 mL) was
addedN-hydroxy succinimide(l.21 g, 10.5 mmol) followed by EDC (2.01 g,10.5 mmol). After
stirring at room temperature for 18 hours, the reaction mixture was concentrated in vacuo. The
residue was diluted with ethyl acetate (200 mL), washed with saturated NaHC03, solution and
brine. The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo.
A yellow solid was obtained.
Step E: Preparation of 1-Benzyl-5-oxo-l,4,5>6-tetrahydro-cyclopentapyrazole-3-
carhoxylic acid amide.

To a solution of the intermediate from step D (2.0 g, 5.25 mmol) in 1,4-dioxane (50 mL)
was added NH4OH (14.8 N, 10.0 eq, 3.53 mL). A precipitate formed immediately. After stirring
at room temperature for 15 minutes the reaction mixture was filtered through a fritted funnel and
the precipitate washed with 1,4-dioxane. The filtrate was concentrated in vacuo to give a solid.
Step F: Preparation of 1-Benzyl-5-oxo-l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carbonitrile.

To a solution of the intermediate from step E (5.25 mmol) in DMF(60 mL) was added
cyanuric chloride (3.12g, 17 mmol) in three portions. After 30 minutes at room temperature, the
reaction was quenched with water and extracted with ethyl acetate (2X). The organic layer was
washed with water, brine and dried over anhydrous Na2S04, filtered, and concentrated in vacuo.
The residue was purified by flash chromatography (Si02) using 30% ethyl acetate-hexanes to
give the desired product (0.95 g) as a yellow solid.
Step G: Preparation of'Trifluoro-methanesulfonic acid l-benzyl-3-cyano-l,6-
dihydro-cyclopentapyrazol-5-yl ester and Trifluoro-methanesulfonic acid l-benzyl-3-
cyano-l,4-dihydro-cyclopcntapyrazol-5-yl ester (mixture).


To a solution of the intermediate from step F (447 mg, 1.87 mmol) in anhydrous THF (14 mL) at
-78 °C was added a solution of freshly prepared lithium diisopropyl amide(1.89 mmol) in THF (6
mL). After stirring the reaction at -78 °C for 30 minutes 2[/V,/V-Bis(trifluromethyl-
sufonyl)amine]-5-ch!oropyridine (1.4 g, 3.6 mmol) was added. The reaction was warmed to -20
"C and stirred for 3 hours. The reaction was quenched with saturated NH4CI solution, and the
resulting mixture was extracted with ethyl acetate, washed with 1N HCI solution, saturated
NaHC03 solution and dried over anhydrous Na2SO in vacuo. The residue was purified on the chromatotron using a2000-micron rotor (Si02) and
5% ethyl acetate-hexanes as eluant to afford 393 mg of the desired product as a 2:1 mixture of
double bond regio-isomers. 'H NMR (500 MHz, CDCI,): (Major isomer) 5 7.45-7.3 (m, 5H),
6.06 (bt, 1H), 5,41 (s,2H), 3.56(bd,2H). 'H NMR (500 MHz, CDC13): (Minor isomer) 6 7.45-
7.3 (m, 5H), 6.63 (bt, 1H), 5.39 (s, 2H), 3.18 (bd, 2H). LC-MS: (M+H)=370.25.
Example 9.9: 5-Propoxy-3-(1 H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 12).

Step A: Preparation of l-Bcnzyl-5-propoxy-l,4,5,6-tetrahydro-cyclopentapyrazole-
3-carbonitrile.

To a solution of I -benzyl-5-hydroxy-1,4,5,6-tetrahydrocyclo-penta[c]pyrazole-3-
carbonitrile (see Example 9.7, 30 mg, 0,125 mmol) in anhydrous DMF (2 ml.) was added
sodium hydride (6 mg, 0.15 mmol, 60% dispersion in oil). After stirring for 3 minutes propyl
bromide was added (14 u.L, 0.15 mmol) and the resulting mixture stirred for an hour. At the end
of this time sodium hydride (6 mg, 0.15 mmol, 60% dispersion in oil) and propyl bromide were

added. After 30 minutes the reaction was quenched by adding saturated NH4CI (3 mL). The
resulting mixture was extracted with ethyl acetate, washed with brine, dried over anhydrous
Na2S04, filtered and concentrated in vacuo. The residue was purified by PTLC (Si02) using
15% ethyl acetate-hexanes to give the desired product.
Step B: Preparation of l-Benzyl-5-propoxy-3-(2H-tetrazol-5-yl)-1,4,5,6-tetrahydro-
cyclopentapyrazole.

To a solution of the intermediate from step A (25 mg, 0.089 mmo!) in 2-propanol (1 mL)
was added water (2 mL), sodium azide (14 mg, 0.222 mmol) and zinc bromide (10 mg, 0.04
mmol). After heating the reaction mixture at 90 °C for 18 hours, it was cooled to room
temperature and HCI (3 mL, 3N) was added. The reaction mixture was extracted with ethyl
acetate, washed with brine, dried over anhydrous Na2S04 filtered and concentrated in vacuo.
The residue was purified by PTLC (Si02) using 100% ethyl acetate to give the desired product.
Step C. 5-Propoxy-3-(l H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopcntapyrazolc
(Compound 12).
To a solution of the intermediate from step B (26 mg, 0.08 mmol) in DMSO (0.6 mL)
was added potassiurrw-butoxide (0.6 mL, 0.6 mmol, 1.0 M in THF). Oxygen gas was bubbled
through the reaction mixture for 15 minutes. The reaction was quenched with HCI (3 mL, 3N).
The resulting mixture was extracted with ethyl acetate (5X) dried over anhydrous Na2SO«i filtered
and concentrated in vacuo. The residue was purified by reverse phase HPLC to afford the title
compound. 'H NMR (CDCI,) 8 4.80 (m, IH), 3.51 (m,2H), 3.24 (dd, 7=6.8, 15.5 Hz, 1H), 3.18
(dd, 7=6.9, 16.0 Hz, 1H), 2.85 (dd, 7=4.1, 15.6 Hz, IH), 2.79 (dd, 7=4.4, 16.1 Hz, IH), 1.62 (m,
2H), 0.96 (t, 7=7.6 Hz, 3H). LC-MS: 2.15 min; (M+H)=235.
Example 9.10: 5-lsobutoxy-3-(l H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 15).


The title compound was prepared from l-benzyl-5-hydroxy-l,4,5,6-tetrahydrocyclo-
penta[cJpyrazole-3-carbonitrile (see Example 9.7) using a similar procedure described for the
synthesis of Example 9.8. 'H NMR(CDC13) 5 4.77 (m, IH), 3.33 (m,2H), 3.23 (dd, 7=6.9, 15.5
Hz, IH), 3.17 (dd, .7=6.9, 16.0 Hz, 1H), 2.85 (dd, 7=4.1, 15.6 Hz, 1H), 2.79 (dd,7=4.2, 15.9 Hz,
IH), 1.85 (m, 1II), 0.94 (d, 7=6.7 Hz, 3H). LC-MS: 2.42 min; (M+H)=249.
Example 9.11: 5-Butoxy-3-(lH-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 16).

The title compound was prepared from l-benzyl-5-hydroxy-l,4,5,6-tetrahydrocyclo-
penta[c]pyrazole-3-carbonitrile (see Example 9.7) using a similar procedure described for the
synthesis of Example 9.8. 'H NMR (CDCI3) 54.78 (m, 1H), 3.56 (m, 2H), 3.24 (dd, 7=6.8, 15.6
Hz, IH), 3.17 (dd, 7=6.9, 15.9 Hz, 1H), 2.84 (dd,7=4.1, 15.6 Hz, IH), 2.77 (dd, 7=4.6, 16.0Hz,
1H), 1.58 (m,2H), 1.42 (m, 2H), 0.95 (1,7=7.3 Hz, 3H). LC-MS: 2.50 min; (M+H)=249.
Example 9.12: 5-Fluoro-3-(l H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopcntapyrazolc
(Compound 14).

Step A: Preparation of l-Bcnzyl-5-fluoro-l,4,5,6-tetrahydro-cyclopentapyrazole-3-
carbonitrile.

To a solution of l-benzyl-5-hydroxy-l,4,5,6-tetrahydrocyclo-penta[c]pyrazole-3-
carbonitrile (sec Example 9.7, 30 mg, 0.125 mmol) in anhydrous dichloromethane (0.9 mL) was
added DAST (33 ^L, 0.25 mmol) under a nitrogen atmosphere. After stirring at room
temperature for 15 minutes, the reaction mixture was diluted with ethyl acetate, washed with

saturated NaHC03 solution and brine. The organic layer was dried over anhydrous Na2S04
filtered and concentrated in vacuo. The residue was purified by PTLC (Si02) using 30% ethyl
acetate-hexanes to give the desired compound (16 mg).
Step B: Preparation of l-Benzyl-5-fluoro-3-(lH-tetrazol-5-yl)-1,4,5,6-tetrahydro-
cyclopentapyrazole.

The compound was prepared from the cyano intermediate from Step A using a similar
procedure described in Example 9.8, Step B.
StepC. 5-Fluoro-3-(lH-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 14).
To a solution of the intermediate from step B (13 mg, 0.04 mmol) in MeOH (1 mL) was
added formic acid (0.1 mL) followed by palladium black (10 mg). After stirring the reaction
mixture under nitrogen atmosphere for 96 hours, it was filtered and concentrated in vacuo. The
residue was purified by reverse phase HPLC (Gilson) to give the title compound (4.9 mg). 'H
NMR (CDjOD, 500 MHz) 8 5.8 (d, J= 51.9 Hz, 1H), 3.31-3.17 (m, 2H), 3.14-2.92 (m, 2H).
LC-MS: 0.99min;(M+H)=195.17.
Example 9.13: 5-Propyl-3-(l H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 11).

Step A: Preparation of 5-Allyl-l-benzyl-1,6-dihydro-cyclopentapyrazole-3-
carbonitrile and 5-Allyl-l-benzyl-l,4-dihydro-cyclopentapyrazole-3-carbonitrile (mixture).

To a solution of the trifluoromethansulfonic ester intermediate described in Example 9.8
(114 mg, 0.307 mmol) in anhydrous THF (2 mL) was added tri-n-butyl allyl tin (112 mg, 0.338

mmol), lithium chloride (39 mg, 0.923 mmol) and tetrakis triphenyl phosphine palladium (0)
(7.1 mg, 0.006 mmol). After refuxing the reaction mixture for 6 hours, it was cooled to room
temperature and filtered. The residue was concentrated in vacuo and purified on the
chromatotron using a 2000-micron rotor (Si02) and 20% ethyl acetate-hexanes as the eluant to
give the desired product (33 mg).
Step B: Preparation of 5-AHyl-l-benzyl-3-(lH-tetrazol-5-yl)-l,6-dihydro-
cyclopentapyrazole and 5-Allyl-l-bcnzyI-3-(lH-tetrazol-5-yl)-l,4-dihydro-
cyclopentapyrazole (mixture).

The compound was prepared from the intermediate obtained in step A above using a
similar procedure described in Example 9.8, step B.
Step C. 5-Propyl-3-(l H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cycIopentapyrazole.
To a solution of the intermediate from step, B (18 mg, 0.059 mmol) in methanol was
added a few drops of concentrated HC1 until the reaction was homogeneous. Pd/C (1.8 mg) was
added and the resulting mixture was stirred under a hydrogen atmosphere (balloon) for 24 hours.
The reaction mixture was filtered, concentrated in vacuo and purified by reverse phase HPLC to
give the title compound. 'H NMR (CD,OD, 500 MHz) 5 3.06(m,2H),2.97 (dd, .7=7.5, 15.1 Hz,
1H), 2.5 (m, 2H), 1.6 (m, 2H), 1.4 (m, 2H), 0.98 (t, .7=7.3 Hz, 3H). LC-MS: 2.60 min;
(M+H)=219.36.
Example 9.14: 5-Cyclopentyl-3-(lH-tetrazol-5-yl)-2,4,5,6-tetrahydro-cycIopentapyrazole
(Compound 13).

Step A: Preparation of l-Bcnzyl-5-cyclopent-l-enyl-l,6-dihydro-
eyclopentapyrazole-3-carbonitrileand l-Benzyl-5-cyclopent-l-enyl-l,4-dihydro-
cyclopentapyrazole-3-carbonitrile (mixture).


To a solution of the trifluoromethansulfonic ester intermediate described in Example 9.8
(185 mg, 0.501 mmol) in 1,4-dioxane was added cyclopenten-1-yl-boronic acid (62 mg, 0.551
mmol), potassium phosphate (160 mg, .751 mmol) and tetrakis triphenyl phosphine palladium
(0). The reaction mixture was heated at 85 °C. After the reaction was complete, it was diluted
with ethyl acetate, washed with IN NaOH, brine and dried over anhydrous Na2S04. The solution
was filtered and concentrated in vacuo. The residue was purified on the chromatotron using a
2000-micron rotor (Si02) and 20 % ethyl acetate-hexanes as the eluant.
Step B: Preparation of l-Benzyl-5-cyc!opent-l-cnyl-3-(l H-tetrazol-5-yl)-l,6-
dihydro-cyclopentapyrazole and l-Benzyl-5-cyclopcnt-l-enyl-3-(lH-tetrazol-5-yl)-l,4-
dihydro-cyclopentapyrazole (mixture).

The compound was prepared from the intermediate obtained in step A above using a
similar procedure described in Example 9.8, step B.
Step C. 5-Cyclopentyl-3-(lH-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 13).
To a solution of the intermediate from step B (16 mg, 0.048 mmol) in methanol (2 mL)
was added formic acid (200 ^L). Palladium black (8.2 mg, 0.078 mmol) was added, and the
resulting mixture was purged with nitrogen and stirred for 24 hours. Another portion of
palladium black was added (8.2 mg, 0.078 mmol). After stirring for 48 hours, the reaction was
filtered, concentrated in vacuo, and purified by reverse phase HPLC to give the title compound.
'H NMR (CD,OD, 500 MHz) 8 3.1 -2.9 (m, 2H), 2.6 (m, 2H), 2.03 (m, 2H), 1.87 (m, 2H), 1.63
(m, 2H), 1.59 (m,2H), 1.28 (m,2H). LC-MS: 2.99 min; (M+H)=245.46.
Example 9.15: 5-Butyl-3-(lH-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopcntapyrazole
(Compound 8).


Step A: Preparation ofl-Benzyl-5-butyl-1,6-dihydro-cyclopentapyrazole-3-
carbonitrile and l-Benzyl-5-buryl-l,4-dihydro-cyclopentapyrazole-3-carbonitrile (mixture).

To a solution of the trifluoromethansulfonic ester intermediate described in Example 9.8
(180 mg, 0.486 mmol) in toluene (3 mL) was added n-butyl boronic acid (99 mg, 0.973 mmol),
K2COj (201 mg, 1.46 mmol), PdClj(dppf)2 (12 mg, 0.0146 mmol) and Ag20 (225 mg, 0.973
mmol). After refuxing the reaction mixture for 6 hours, it was cooled to room temperature and
filtered. The residue was concentrated in vacuo and purified on the chromatotron using a 2000-
micron rotor (Si02) and 5% ethyl acetate-20% ethyl acetate-hexanes as the eluant to give the
desired product (52 mg).
Step B: Preparation of ]-Benzyl-5-butyl-3-(lH-tetrazol-5-yl)-l,6-dihydro-
cyclopentapyrazole and l-Bcnzyl-5-butyl-3-(lH-tetrazol-5-yl)-l,4-dihydro-
cyclopentapyrazole (mixture).

The compound was prepared from the intermediate obtained in step A above using a
similar procedure described in Example 9.8 step B.
Step C. 5-Butyl-3-(l H-tetrazol-5-yl)-2,4,5,6-tetrahydro-cyclopentapyrazole
(Compound 8).
The compound was prepared from the intermediate obtained in step B above using a
similar procedure described in Example 5 step C. 'H NMR (CD3OD, 500 MHz) 5 3.1 (m, 2H),
2.9 (m, 1 H), 2.5 (m, 2H), 1.6 (m, 2H), 1.4 (m, 4H), 0.9 (t, 7=7.0 Hz, 3H). LC-MS: 2.86 min,
(M+H)=233.34.

Example9.16: 5-Methyl-3-(lH-tetrazol-5-yl)-2,6-dihydro-cyclopentapyrazole(Compound
9) and S-Methyl-3-(lH-tetrazol-5-yl)-2,4-dihydro-cyclopentapyrazole (Compound 10).

Step A: Preparation of 5-Ethoxy-l,4-dihydro-cyclopentapyrazole-3-carboxylic acid
tert-butyl ester.

To an ethanol (5 mL) solution of the ketoester (254 mg, 1.0 mmol), prepared from 3-
ethoxy cyclopentenone as in step A of Example 9.8 above, was added hydrazine hydrate (34 u,L,
I.I mmol), followed by acetic acid (0.5 mL). After refluxing the reaction mixture for 1.5 hours,
it was cooled to room temperature and concentrated in vacuo. The residue was suspended in
water and extracted with ethyl acetate. The organic layer was dried over anhydrous Na2S04,
filtered and concentrated in vacuo. The residue was purified by flash chromatography (Si02)
using 25% ethyl acetate-hexanes to give the desired product as a white solid.
Step B: Preparation of 5-Ethoxy-l-(toluene-4-sulfonyl)-!,4-dihydro-
cyclopcntapyrazole-3-carboxylic acid tert-butyl ester.

To a solution of the pyrazole intermediate from step A (275 mg, 1.1 mmol) above in
CH2CI2 (5 mL) was added pyridine (178 p.L, 2.2 mmol) andp-toluene sulfonyl chloride (230 mg,
1.21 mmol). After stirring the resulting reaction mixture at room temperature for 3 hours it was
diluted with CH2CI2, washed with IN HCI, saturated NaHC03, dried over anhydrous Na2SOo,
filtered and concentrated in vacuo. The residue was purified by flash chromatography (Si02)
using 10% ethyl acetate-hexanes.
Step C: Preparation of 5-Oxo-l-(toluene-4-sulfonyl)-l,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid.


To a solution of the intermediate from step B above, (414 mg, 1.02 mmol) in CH2C12 (2
mL) was added trifluoroacetic acid (2 mL). After stirring the reaction at room temperature for
1.5 hours it was concentrated in vacuo and azeotroped with toluene (2X). This material was used
in the next step without any further purification.
Step D: Preparation of 5-Oxo-l-(toluene-4-sulfonyl)-l,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid 2,5-dioxo-pyrrolidin-l-yl ester.

To a solution of the intermediate from step C above, (320 mg, I .Ommol) in CH2C12(20
mL) was added /V-hydroxy succinimide (230 mg, 2.Ommol) followed by EDC (384 mg, 2.0
mmol). After stirring at room temperature for 18 hours, the reaction mixture was concentrated in
vacuo. The residue was diluted with ethyl acetate (20 mL), washed with saturated NaHCOt,
solution and brine. The organic layer was dried over anhydrous Na2S04, filtered and
concentrated in vacuo. A yellow solid was obtained.
Step E: Preparation of 5-Oxo-l-(toluene-4-sulfonyl)-l,4,5,6-tetrahydro-
cyclopentapyrazole-3-carboxylic acid amide.

To a solution of the intermediate from step D above, (380 mg, 0.91 mmol) in 1,4-
dioxane (10 mL) was added NH4OH (14.8 N, 10.0 eq, 0.61 mL). A precipitate formed
immediately. After stirring at room temperature for 15 minutes the reaction mixture was filtered
through a sintered funnel and the precipitate washed with 1,4-dioxane. The filtrate was
concentrated in vacuo to give a yellow oil.
Step F: Preparation of S-Oxo-I-(toluene-4-sulfonyl)-1,4,5,6-tetrahydro-
cyclopcntapyrazole-3-carbonitrile.


To a solution of the intermediate from step E above (0.91 mmol) in anhydrous DMF (5
mL) was added cyanuric chloride (334mg, 2.0 mmol) in two portions. After stirring at room
temperature for 15 minutes, the reaction was quenched by pouring into water (10 mL). The
resulting mixture was extracted with ethyl acetate, washed with saturated NaHC03, brine, dried
over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by tlash
chromatography (Si02) using 25 % ethyl acetate-hexanes. A white solid was obtained.
Step G: Preparation of Trifluoro-methanesulfonic acid 3-cyano-l-(toluene-4-
sulfonyl)-l,6-dihydro-cyclopentapyrazol-5-yl ester and Trifluoro-methanesulfonic acid 3-
cyano-l-(toluene-4-sulfonyl)-l,4-dihydro-cyclopentapyrazol-5-yl ester (mixture).

To a solution of the intermediate from step F (100 mg, 0.33mmol) in anhydrous THF (5
mL) at -78 °C was added a solution of lithium diisopropyl amide(0.33mmol, 166 (iL, 2.0 M in
THF) in THF (6 mL). After stirring the reaction at -78 °C for 30 minutes 2[N,N-
Bis(trifluromethylsufonyI)amine]-5-chloropyridine (195 mg, 0.496 mmol) was added. The
reaction was warmed to 0 °C and stirred for 45minutes. The reaction was quenched with IN HC1
solution, and the resulting mixture was extracted with ethyl acetate, washed with saturated
NaHCOj solution and dried over anhydrous Na2S04. The solution was filtered and concentrated
in vacuo. The residue was purified by flash chromatography (Si02) using 5% ethyl acetate-
hexanes as eluant to afford 79 mg of the desired product as a 4:1 mixture of double bond regio-
isomers.
Step H: Preparation of 5-Methyl-l-(tolucne-4-sulfonyl)-l,6-dihydro-
cyclopentapyrazole-3-carbonitrileand 5-MethyI-l-(toluene-4-sulfonyl)-l,4-dihydro-
cyclopentapyrazole-3-carbonitrile (mixture).

To a solution of the intermediate from stepG above, (79 mg, 0.182 mmol) in toluene
(1.5 mL) was added lithium chloride (39 mg, 0.912 mmol), tetramethyl tin (126 uL, 0.912 mmol)
and tetrakis triphenyl phosphine palladium (0). After refluxing the reaction mixture for 45

minutes it was cooled to room temperature, diluted with ethyl acetate, washed with water. The
organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue
was purified by flash chromatography (Si02) using 30 % ethyl aceate-hexanes to give (28 mg)
the desired product.
Step 1: Preparation of 5-Methyl-l,6-dihydro-cyclopentapyrazole-3-carbonitrile
and 5-Methyl-l,4-dihydro-cyclopentapyrazole-3-carbonitrile (mixture).

To a solution of the intermediate from step H above, (28 mg, 0.093 mmol) in anhydrous
THF (3 IDL) was added tetrabutyl ammonium fluoride (93 [iL, 0.093 mmol, 1.0 M in THF).
After refluxing the reaction mixture for 30 minutes, it was cooled to room temperature and
concentrated in vacuo. The residue was dissolved in ethyl acetate, washed with saturated
NaHCOi, brine, dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue
was purified by flash chromatography (Si02) using 30% ethyl acetate-hexanes to give a white
solid.
Step J: 5-Methyl-3-(] H-tetrazol-5-yl)-2,6-dihydro-cyclopentapyrazole (Compound
9) and 5-Methyl-3-(1H-tetrazol-5-yl)-2,4-dihydro-cyclopentapyrazole (Compound 10).
To a solution of the intermediate from step I above (9.0 mg, 0.062 mmol) in 2-propanol
(1 mL) was added water (0.5 mL), sodium azide(12 mg, 0.186 mmol) and zinc bromide (6.5 mg,
0.031 mmol). After heating the reaction mixture at 90 °C for 18 hours, it was cooled to room
temperature and HCI (1.5 mL, 3N) was added. The reaction mixture was extracted with ethyl
acetate, washed with brine, dried over anhydrous Na2S04 filtered and concentrated in vacuo to
give the desired product as a 2:1 ratio of double bond regio-isomers. Isomer (a): 'HNMR
(CD30D, 500 MHz) 6 6.43 (bs, IH), 3.1 (s, 2H), 2,2 (s, 3H). Isomer (b): 'H NMR (CD3OD, 500
MHz) 8 6.58 (bs, 1H), 3.24 (s, 2H), 2.15 (s, 3H). LC-MS: 1.86 min, (M+H)= 189.1
Throughout this application, various publications, patents and published patent
applications are cited. The disclosures of these publications, patents and published patent
applications referenced in this application are hereby incorporated by reference in their entirety
into the present disclosure. Modifications and extension of the disclosed inventions that are
within the purview of the skilled artisan are encompassed within the above disclosure and the
claims that follow.
Although a variety of expression vectors are available to those in the art, for purposes of
utilization for both the endogenous and non-endogenous human GPCRs, it is most preferred that
the vector utilized be pCMV. This vector was deposited with the American Type Culture

Collection (ATCC) on October 13, 1998 (10801 University Blvd., Manassas, VA 20110-2209
USA) under the provisions of the Budapest Treaty for the International Recognition of the
Deposit of Microorganisms for the Purpose of Patent Procedure. The DNA was tested by the
ATCC and determined to be viable. The ATCC has assigned the following deposit number to
pCMV: ATCC #203351.

WE CLAIM:
1. Tetrazole derivatives comprising a compound of Formula(I):

wherein:
X is NH or O;
R1 is selected from the group consisting of H, halogen, hydroxy,
thioxy, cyano, nitro. C1-4 haloalkyl, amino, C1-4 alkylamino, C2-8
dialkylamino, C1-4 alkyl, C1-4 alkoxy, C2-4 alkenyl, C2-4 alkynyl, C3-5
cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio, C1-4 alkylsulfinyl. C1-4
alkylsulfonyl. C1-4 haloalkylthio, C1-4 haloalkylsulfinyl and C1-4
haloalkylsulfonyl;
R2 is selected from the group consisting of H, halogen, hydroxyl,
thioxy. cyano, nitro, C1-4 haloalkyl, amino, C1-4 alkylamino. C2-8
dialkylamino, C1-4 alkyl, C1-4 alkoxy, C2-4 alkenyl, C2-4 alkynyl, C3-5
cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio, C1-4 alkylsulfinyl, C1-4
alkylsulfonyl, C1-4 haloalkylthio, C1-4 haloalkylsulfinyl and C1-4
haloalkylsulfonyl; or R2 is absent;
is a single bond when R2 is present, or is a double bond when R2 is
absent; and
Ring A is a 5-membered carbocyclic ring or a 5-membered
heterocyclic ring; wherein 5-membered carbocyclic ring denotes a ring
containing 5 ring carbons wherein two ring carbons are shared by Rings A
and B; wherein 1, 2, or 3 ring carbons not shared by Rings A and B are
independently replaced with -O-, -S-, -S(O)-, or -S(O)2-; and wherein Ring
A is optionally substituted with 1 to 4 substituents selected from the group
consisting of halogen, hydroxy, thioxy, cyano, nitro, C1-4 haloalkyl. amino,
C1-4 alkylamino, C2-8 cycloalkyl, C1-4 haloalkoxy, C1-4 alkylthio. C1-4
alkylsulfinyl, C1-4 alkylsulfonyl, C1-4 haloalkylthio, C1-4 haloalkylsulfinyl
and C1-4 haloalkylsulfonyl; or
a pharmaceutically acceptable salt, solvate or hydrate thereof.

2. A compound as claimed in claim 1, or a pharmaceutically acceptable salt, solvate
or hydrate thereof, wherein Ring A is a 5-membered carbocyclic ring.
3. A compound as claimed in claim 1, or a pharmaceutically acceptable salt, solvate
or hydrate thereof, wherein Ring A is a 5-membered heterocyclic ring.
4. A compound as claimed in claim 1, or a pharmaceutically acceptable salt, solvate
or hydrate thereof, wherein Ring A is a 5-membered heterocyclic ring, wherein 5-
membered heterocyclic ring denotes a 5-membered carbocyclic ring wherein 1 ring
carbon not shared by Rings A and B is independently replaced with -O-, -S-, -
S(O)-,or-S(O)2-;
5. The compound as claimed in any one of claims 1 to 4, wherein:
X is NH
R1 is H or hydroxy;
R2 is H or absent;
— is a single bond when R2 is H, or is a double bond when R2 is absent;
and
Ring A is a 5-membered carbocyclic ring or a 5-membered heterocyclic
ring optionally substituted with 1 to 4 substituents selected from the group consisting
of halogen, C1-4 alkyl, C1-4alkoxy and C3-5 cycloalkyl; or
a pharmaceutically acceptable salt, solvate or hydrate thereof.
6. The compound as claimed in claim 1 or 2 having Formula (IF):

wherein:
R1 is H or hydroxy; and
Ring A is optionally substituted with 1 or 2 substituents selected from the
group consisting of halogen, C1-4 alkyl, C1-4 alkoxy and C3-5 cycloalkyl; or
a pharmaceutically acceptable salt, solvate or hydrate thereof

7. The compound as claimed in claim 1 or 2 having Formula (Ih):

wherein:
Ring A is optionally substituted with 1 or 2 substituents selected from the
group consisting of halogen, C1-4 alkyl, C 1-4alkoxy and C3-5 cycloalkyl; or
8. The compound as claimed in claim 1 or 2 having Formula (Ih):

wherein:
Ring A is unsubstituted or is substituted with ethyl; or a pharmaceutically
acceptable salt, solvate or hydrate thereof.
9. The compound as claimed in claim 1 or 2 having Formula (Ih):

wherein:
Ring A is optionally substituted with 1 or 2 substituents selected from the
group consisting of halogen, n-propyl, n-butyl, C1-4 alkoxy and C3-5 cycloalkyl; or
a pharmaceutically acceptable salt, solvate or hydrate thereof
10. The compound as claimed in claim 1 that is 3-(1H-Tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.

11. The compound as claimed in claim 1 that is 3-(1H-Tetrazol-5-yl)-2,6-dihydro-4H-
thieno[3,4-c] pyrazole or a pharmaceutically acceptable salt, solvate or hydrate
thereof.
12. The compound as claimed in claim 1 that is 6-Methyl-3(1H-tetrazol-5-yl)-2,6-
dihydro-4H-furo[3,4-c] pyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
13. The compound as claimed in claim 1 that is 3-(1H-Tetrazol-5-yl)-2,4-dihydro-
cyclopentapyrazol or a pharmaceutically acceptable salt, solvate or hydrate thereof.
14. The compound as claimed in claim 1 that is 3-(1H-Tetrazol-5-yl)-2,6-dihydro-
cyclopentapyrazol or a pharmaceutically acceptable salt, solvate or hydrate thereof.
15. The compound as claimed in claim 1 that is 3-(1H-Tetrazol-5-yl)-2,6-dihydro-4H-
furo[3,4-c] pyrazole or a pharmaceutically acceptable salt, solvate or hydrate
thereof.
16. The compound according to claim 1 that is 5-Ethyl-3-(1H-tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
17. The compound as claimed in claim 1 that is 5-Butyl-3-(1H-tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
18. The compound as claimed in claim 1 that is 5-Methyl-3-(1H-tetrazol-5-yl)-2,6-
dihydro -cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
19. The compound as claimed in claim 1 that is 5-Methyl-3-(1H-tetrazol-5-yl)-2,4-
dihydro -cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
20. The compound as claimed in claim 1 that is 5-Propyl-3-(1H-tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.

21. The compound as claimed in claim 1 that is 5-Propoxy-3-(1H-tetrazol-5-yl)-
2,4,5,6-tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt,
solvate or hydrate thereof.
22. The compound as claimed in claim 1 that is 5-Cyclopentyl-3-(1H-tetrazol-5-yl)-
2,4,5,6-tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt,
solvate or hydrate thereof.
23. The compound as claimed in claim 1 that is 5-Fluoro-3-(1H-tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
24. The compound as claimed in claim 1 that is 5-Isobutoxy-3-(1H-tetrazol-5-yl)-
2.4.5,6-tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt,
solvate or hydrate thereof.
25. The compound as claimed in claim 1 that is 5-Butoxy-3-(1H-tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.
26. The compound as claimed in claim 1 that is 3-(1H-Tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole-6-ol or a pharmaceutically acceptable salt, solvate
or hydrate thereof.
27. The compound as claimed in claim 1 that is 5-Mefhoxy-3-(1H-tetrazol-5-yl)-
2,4,5,6-tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt,
solvate or hydrate thereof.
28. The compound as claimed in claim 1 that is 5,5-Difluoro-3-(1H-tetrazol-5-yl)-
2,4,5,6-tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt,
solvate or hydrate thereof.
29. The compound as claimed in claim 1 that is 5-Efhoxy-3-(1H-tetrazol-5-yl)-2,4,5,6-
tetrahydro-cyclopentapyrazole or a pharmaceutically acceptable salt, solvate or
hydrate thereof.

30 A pharmaceutical composition comprising a compound as claimed in any one of
claims 1 to 29, or a pharmaceutically acceptable salt, solvate or hydrate thereof, in
combination with a pharmaceutically acceptable carrier.
31 A method of producing a pharmaceutical composition comprising admixing a
compound as claimed in any one of claims 1 to 29, or a pharmaceutically
acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable
carrier.
32 A compound as claimed in any one of claims 1 to 29, or a pharmaceutically
acceptable salt, solvate or hydrate thereof, for use in treating the human or animal
body by therapy.
33 A compound as claimed in any one of claims 1 to 29, or a pharmaceutically
acceptable salt, solvate or hydrate thereof, for use in treating a metabolic-related
disorder of the human or animal body by therapy.
34 A compound as claimed in any one if claims 1 to 29, or a pharmaceutically
acceptable salt, solvate or hydrate thereof, for use in treating a metabolic-related
disorder of the human or animal body by therapy wherein said metabolic-related
disorder is selected from the group consisting of dyslipidemia, atherosclerosis,
coronary heart disease, insulin resistance and type 2 diabetes.
35 A compound as claimed in any one of claims 1 to 29, or a pharmaceutically
acceptable salt, solvate or hydrate thereof, for use in treating atherosclerosis of the
human or animal body by therapy.
36 A compound as claimed in any of claims 1 to 29, or a pharmaceutically acceptable
salt, solvate or hydrate thereof, for use in raising HDL of the human or animal
body by therapy.

ABSTRACT

TETRAZOLE DERIVATIVES


The present invention relates to certain tetrazole derivatives of Formula (I), and
pharmaceutically acceptable salts thereof, which exhibit useful pharmacological
properties, for example, as agonists for the RUP25 receptor. Also provided by the present
invention are pharmaceutical compositions containing compounds of the invention, and
methods of using the compounds and compositions of the invention in the treatment of
metabolic-related disorders, including dyslipidemia, atherosclerosis, coronary heart
disease, insulin resistance, type 2 diabetes, Syndrome-X and the like. In addition, the
present invention also provides for the use of the compounds of the invention in
combination with other active agents such as those belonging to the class of &agr;-
glucosidase inhibitors, aldose reductase inhibitors, biguanides, HMG-CoA reductase
inhibitors, squalene synthesis inhibitors, fibrates, LDL catabolism enhancers, angiotensin
converting enzyme (ACE) inhibitors, insulin secretion enhancers and the like.

Documents:

01249-kolnp-2006-abstract.pdf

01249-kolnp-2006-assignment.pdf

01249-kolnp-2006-claims.pdf

01249-kolnp-2006-correspondence other.pdf

01249-kolnp-2006-correspondence others-1.1.pdf

01249-kolnp-2006-description (complete).pdf

01249-kolnp-2006-form-1.pdf

01249-kolnp-2006-form-3-1.1.pdf

01249-kolnp-2006-form-3.pdf

01249-kolnp-2006-form-5.pdf

01249-kolnp-2006-international publication.pdf

01249-kolnp-2006-international search authority report.pdf

01249-kolnp-2006-pct form.pdf

01249-kolnp-2006-priority document.pdf

1249-KOLNP-2006-(14-09-2009)FORM-13.pdf

1249-KOLNP-2006-(15-02-2010)-FORM 13.pdf

1249-KOLNP-2006-(30-11-2011)-CORRESPONDENCE.pdf

1249-KOLNP-2006-ABSTRACT 1.1.pdf

1249-kolnp-2006-abstract.pdf

1249-kolnp-2006-amanded claims.pdf

1249-kolnp-2006-assignment.pdf

1249-KOLNP-2006-CLAIMS 1.1.pdf

1249-kolnp-2006-claims.pdf

1249-kolnp-2006-correspondence 1.2.pdf

1249-kolnp-2006-correspondence-1.1.pdf

1249-kolnp-2006-correspondence-1.2.pdf

1249-KOLNP-2006-CORRESPONDENCE-1.3.pdf

1249-kolnp-2006-correspondence.pdf

1249-KOLNP-2006-DESCRIPTION (COMPLETE) 1.1.pdf

1249-kolnp-2006-description (complete).pdf

1249-kolnp-2006-examination report.pdf

1249-KOLNP-2006-EXAMINATION REPORT1.1.pdf

1249-kolnp-2006-form 1-1.1.pdf

1249-KOLNP-2006-FORM 1-1.2.pdf

1249-kolnp-2006-form 1-1.3.pdf

1249-kolnp-2006-form 1.pdf

1249-kolnp-2006-form 13-1.1.pdf

1249-kolnp-2006-form 13.pdf

1249-kolnp-2006-form 18.pdf

1249-KOLNP-2006-FORM 2-1.1.pdf

1249-kolnp-2006-form 2-1.2.pdf

1249-kolnp-2006-form 2.pdf

1249-KOLNP-2006-FORM 3.1.pdf

1249-kolnp-2006-form 3.pdf

1249-kolnp-2006-form 5-1.1.pdf

1249-KOLNP-2006-FORM 5-1.2.pdf

1249-kolnp-2006-form 5-1.3.pdf

1249-kolnp-2006-form 5.pdf

1249-KOLNP-2006-GPA-1.2.pdf

1249-kolnp-2006-gpa.pdf

1249-KOLNP-2006-GPA1.1.pdf

1249-KOLNP-2006-GRANTED-ABSTRACT.pdf

1249-KOLNP-2006-GRANTED-CLAIMS.pdf

1249-KOLNP-2006-GRANTED-DESCRIPTION (COMPLETE).pdf

1249-KOLNP-2006-GRANTED-FORM 1.pdf

1249-KOLNP-2006-GRANTED-FORM 2.pdf

1249-KOLNP-2006-GRANTED-SPECIFICATION.pdf

1249-kolnp-2006-others 1.1.pdf

1249-KOLNP-2006-OTHERS.pdf

1249-KOLNP-2006-OTHERS1.2.pdf

1249-KOLNP-2006-PA-1.1.pdf

1249-kolnp-2006-pa.pdf

1249-KOLNP-2006-REPLY TO EXAMINATION REPORT 1.1.pdf

1249-kolnp-2006-reply to examination report.pdf

1249-KOLNP-2006-REPLY TO EXAMINATION REPORT1.1.pdf

1249-kolnp-2006-specification.pdf

1249-KOLNP-2006-TRANSLATED COPY OF PRIORITY DOCUMENT.pdf

abstract-01249-kolnp-2006.jpg


Patent Number 256946
Indian Patent Application Number 1249/KOLNP/2006
PG Journal Number 33/2013
Publication Date 16-Aug-2013
Grant Date 16-Aug-2013
Date of Filing 12-May-2006
Name of Patentee MERCK SHARP & DOHME CORP.
Applicant Address 126 EAST LINCOLN AVENUE, RAHWAY, NEW JERSEY 07065-0907, UNITED STATES OF AMERICA
Inventors:
# Inventor's Name Inventor's Address
1 SEMPLE, GRAEME 15920 CAMINO CODORNIZ, SAN DIEGO, CALIFORNIA 92127
2 SKINNER, PHILIP, J. 1222 OLIVER AVENUE, NO. 2, SAN DIEGO, CALIFORNIA 92109
3 COLLETTI, STEVEN, L. 3 BOROSKO PLACE, PRINCETON JUNCTION, NEW JERSEY 08550
4 GHARBAOUI, TAWFIK 2340 LOMICA PLACE, ESCONDIDO, CALIFORNIA 92029
5 IMBRIGLIO, JASON, E 280 RIVER ROAD, APARTMENT 69B, PISCATAWAY, NEW JERSEY 08854
6 JUNG, JAE-KYU 16478 CAMELAS WALK, SAN DIEGO, CALIFORNIA 92127
7 LIANG, RUI 4 SCHINDER COURT, EAST BRUNSWICK, NEW JERSEY 08816
8 RAGHAVAN, SUBHAREKHA 900 PERRY LANE, TEANECK, NEW JERSEY 07666
9 SCHMIDT, DARBY 25 DAWN DRIVE, CLARK, NEW JERSEY 07066
10 TATA, JAMES, R. 25 FAULKNER DRIVE, WESTFIELD, NEW JERSEY 07099
11 SCHRADER, THOMAS 8526 VILLA LA JOLLA DRIVE, NO.147, LA JOLLA, CALIFORNIA 92037
PCT International Classification Number C07D 403/04
PCT International Application Number PCT/US2004/035927
PCT International Filing date 2004-10-29
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/516,238 2003-10-31 U.S.A.