Title of Invention

BICYCLOHETEROARYL COMPOUNDS AS P2X7 MODULATORS AND USES THEREOF

Abstract Bicycloheteroaryl compounds are disclosed that have formula (I). The compounds may be prepared as pharmaceutical compositions, and may be used for the prevention and treatment of a variety of conditions in mammals including humans, including by way of non-limiting example, pain, inflammation, traumatic injury, and others.
Full Text FORM 2
THE PATENT ACT 1970 (39 of 1970)
The Patents Rules, 2003 COMPLETE SPECIFICATION (See Section 10, and rule 13)
TITLE OF INVENTION
BICYCLOHETEROARYL COMPOUNDS AS P2X7 MODULATORS AND USES THEREOF



APPLICANT(S)
a) Name
b) Nationality
c) Address

RENOVIS, INC. AMERICAN Company 2 CORPORATE DRIVE, SOUTH SAN FRANCISCO, CA 94080 U.S.A.

3. PREAMBLE TO THE DESCRIPTION
The following specification particularly describes the invention and the manner in which it is to be performed : -



2008
16SEP2008

16 SEP 2008
FIELD OF THE INVENTION
This invention relates to novel compounds of the class bicycloheteroaryls that are capable of modulating P2X7 receptor activity, and to pharmaceutical compositions containing such compounds. This invention also relates to methods for preventing and/or treating conditions that are causally related to aberrant P2X7 activity, such as inflammation-related conditions in mammals, comprising (but not limited to) rheumatoid arthritis, osteoarthritis, Parkinson's disease, uveitis, asthma, cttdiovascutaff conditions including myocardial infarction, the treatment and prophylaxis of pain syndromes (acute and chronic or neuropathic), traumatic brain injury, acute spinal cord injury, neurodegenerative disorders, inflammatory bowel disease and autoimmune disorders, using the compounds arid pharmaceutical compositions of the invention.
BACKGROUND OF THE INVENTION
Cell surface receptors for ATP can be divided into metabotropic (P2Y/P2U) and ionotropic (P2X) classes. The metabotropic class belongs to the superfamily of G protein-coupled receptors, with seven transmembrane segments. The ionotropic class members (P2X i - P2X 6) are ligand-gated ion channels, currently thought to be multisubunit proteins with two transmembrane domains per subunit (Buell et al, Europ. J. Neurosci. 8:2221 (1996)). P2Z receptors have been distinguished from other P2 receptors in three primary ways (Buismafl et al, Proc. Natl. Acad. Sci. USA 85:7988 (1988); Cockcroft et al, Nature 279:541 (1979); Steinberg et al, J. Biol. Chem. 262:3118 (1987)). First, activation of P2Z receptors leads not only to an inward ionic current, but also to cell permeabilization. Second, 3'-0-(4-benzoyl)benzoyl ATP (BZATP) is the most effective agonist, and ATP itself is of rather low potency. Third, responses are strongly inhibited by extracellular magnesium ions, that has been interpreted to indicate that ATP4- is the active agonist (DiVirgilio, Immunol. Today 16:524 (1995)).
A seventh member of the P2X receptor family has been isolated from a rat cDNA library and, when expressed in human embryonic kidney (HEK293) cells, exhibits the above three properties (Surprenant et al, Science 272:735 (1996)). This receptor (rP2X7) thus corresponds to the P2Z receptor. rP2X7 is structurally related to other members of the P2X family but it has a longer cytoplasmic C-terminus domain (there is 35-40% amino acid identity in the corresponding region of homology, but the C-terminus is 239 amino acids long in the rP2X? receptor compared with 27-20 amino acids in the others). The rP2X7 receptor functions both as a channel permeable to small cations and as a cytolytic pore. Brief applications of ATP (l-2s) transiently open the channel, as is the case of other P2X receptors. Repeated or prolonged applications of agonist cause cell permeabilization reducing the extracellular magnesium concentration potentiates this effect. The unique C-tenninal domain of rP2X7 is required for cell permeabilization and the lytic actions of ATP (Suprenant et al, Science 272:735 (1996)).
The P2Z/ rP2X7 receptor has been implicated in lysis of antigen-presenting cells by cytotoxic T lymphocytes, in the mitogenic stimulation of human T lymphocytes, as well as in the formation of multinucleated giant cells (Blanchard et al, Blood 85:3173 (1995); Falzoni et al, J. Clin. Invest. 95:1207 (1995); Baricolrdi et al, Blood 87:682 (1996)). Certain functional differences exist between rodent and man (Hickman et al, Blood 84:2452 (1994)). The human macrophage P2X7 receptor (P2X7) has now been cloned and its functional properties determined (Rassendren et al, J. Biol. Chem. 272:5482
-2-

(1997). When compared with the rat P2X7 receptor, elicited cation-selective currents in the human P2X7 receptor required higher concentrations of agonists, were more potentiated by removal of extracellular magnesium ions, and revised more rapidly on agonist removal. Expression of chimeric molecules indicated that some of the differences between rat and human P2X7 receptors could be revised by exchanging the respective C-terminal domains of the receptor proteins.
It has been reported that certain compounds act as P2X7 antagonists. For example, WO99/29660 and W099/29661 disclose that certain adamantane derivatives exhibit P2X7 antagonistic activity having therapeutic efficacy in the treatment of rheumatoid arthritis and psoriasis. Similarly, W099/29686 discloses that certain heterocyclic derivatives are P2X7 receptor antagonists and are useful as immunosuppressive agents and treating rheumatoid arthritis, asthma, septic shock and atheroscelerosis. Finally, WO00/71529 discloses certain substituted phenyl compounds exhibiting immunosuppressing activity. All of the references described herein are incorporated herein by reference in their entirety.
A need therefore exists for therapeutic agents, and corresponding pharmaceutical compositions and related methods of treatment, that address the conditions causally related to aberrant P2X7 activity, and it is toward the fulfillment and satisfaction of that need, that the present invention is directed.
SUMMARY OF THE INVENTION
Bicycloaryl derivatives of formulae I-XIIId, and their pharmaceutical compositions are disclosed as therapeutic agents useful for the treatment of conditions in mammals associated with abnormal or aberrant activity of the P2X7 receptor, including inflammatory-mediated conditions such as (but not limited to) arthritis, myocardial infarction, the treatment and prophylaxis of pain syndromes (acute and chronic [neuropathic]), traumatic brain injury, acute spinal cord injury, neurodegenerative disorders, inflammatory bowel disease and immune dysfunctions such as autoimmune disorders.
It has now been found that the present bicycloheteroaryl compounds are capable of mediating the activity of the P2X7 receptor. This finding leads to novel compounds having therapeutic value. It also leads to pharmaceutical compositions having the compounds of the present invetion as active ingredients and to their use to treat, prevent or ameliorate a range of conditions in mammals such as but not limited to inflammation of various genesis or etiology, for example rheumatoid arthritis, cardiovascular disease, inflammatory bowel disease, acute, chronic, inflammatory and neuropathic pain, dental pain and headache (such as migraine, cluster headache and tension headache) and other conditions causally related to inflammation or immune dysfunction.
The compounds of the present invention are also useful for the treatment of inflammatory pain and associated hyperalgesia and allodynia. They are also useful for the treatment of neuropathic pain and associated hyperalgesis and allodynia (e.g. trigeminal or herpetic neuralgia, diabetic neuropathy, causalgia, sympathetically maintained pain and deafferentation syndromes such as brachial plexus avulsion). The compounds of the present invention are also useful as anti-inflammatory agents for the treatment of arthritis,
-3-

and as agents to treat Parkinson's Disease, uveitis, asthma, myocardial infarction, traumatic brain injury, spinal cord injury, neurodegenerative disorders, inflammatory bowel disease and autoimmune disorders, renal disorders, obesity, eating disorders, cancer, schizophrenia, epilepsy, sleeping disorders, cognition, depression, anxiety, blood pressure, lipid disorders, and atherosclerosis.
In one aspect, this invention provides bicycloheteroaryl compounds which are capable of modulating the activity of the P2X7 receptor, in vivo. In £ further aspect, the compounds of the invention are capable of antagonizing (suppressing or inhibiting) the activity of the P2X7 receptor, and thereby treating those conditions, representative ones of which are causally related to aberrant P2X7 activity.
The compounds of the present invention may show low toxicity, good absorption, good half-life, good solubility, low protein binding affinity, low drug-drug interaction, low inhibitory activity at the HERG channel, low QT prolongation and good metabolic stability.
Accordingly, in a first aspect of the invention, bicycloheteroaryl compounds are disclosed that are capable of capable of modulating the activity of the P2X7 receptor in vivo, having a formula CIV

I wherein
A is CR2aR2b or CO; B and Y are independently selected from CR2a and CR28R2b W, W and Z are independently selected from CR4 and N, provided that all three of W and Z are not N at the same time;
L1 is a bond, SO, S02 or substituted or unsubstituted C1C5 alkylene; n is 0,1, 2, 3 or 4;
R1 is selected from a substituted or unsubstituted 3-13 membered cycloalkyl, heterocycloalkyl, aryl and heteroaryl ring;
each of R2a, R2b, R2' and R2" is independently selected from hydrogen, halo, and substituted or unsubstituted C1-C6 alkyl; or any of R2' and R2" join together to form a cycloalkyl or cycloheteroalkyl ring of 3-7 atoms;
R3 is selected from hydrogen, a hydrogen bond donor group, substituted or unsubstituted cycloalkyl, substituted or unsubstituted alkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted or unsubstituted bicycloheteroaryl;
R4 is independently selected from H, alkyl, substituted alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy, alkoxycarbonyl, substituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, arylalkyloxy, substituted arylalkyloxy, amino, aryl, substituted aryl, arylalkyl, substituted or
-4-

unsubstituted sulfoxide, substituted or unsubstituted sulfore, substituted or unsubstituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted arylsulfonyl, sulfuric acid, sulfuric acid ester, substituted or unsubstituted dihydroxyphosphoryl, substituted or unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroalkyl, hydroxy, nitro, and thio; and the dotted bond is a single or a double bond; or a pharmaceutically acceptable salt, solvate or prodrug th£re°f; and stereoisomers, isotopic variants and tautomers thereof.
In a further embodiment, with respect to compounds of forrnulae I, n is 0-4.
In a further embodiment, with respect to compounds of forfnula I, L is a bond, or a CrC5 alkylene group unsubstituted or substituted by one or mpre substituents selected from alkyl, hydroxy, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, halogen, carbamoyl, oxo, aryl, and Cr6 alkoxy.
In a further embodiment, with respect to compounds of formula I, L1 is CrC5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, oxo, aryl, hydroxyl, and hydroxyalkyl
In a further embodiment, with respect to compounds of fofmula I, L is a Q-C5 alkylene group substituted with two alkyl groups and wherein any two alkyl groups on the same carbon atom can join together to form a cycloalkyl or cyclofreteroalkyl ring of 3-7 atoms.
In a further embodiment, with respect to compounds of formula I, L is a bond, a C1-C5 alkylene group; and R3 is a hydrogen bond donor group. In one embodiment, R is -OH. In another embodiment, R3 is NH2. In yet another embodiment R is -NH-.
In a further embodiment, with respect to compounds of formula I, L is a bond, a CrC5 alkylene group substituted with oxo; and R3 is a hydrogen bond donor group. In one embodiment, R3 is -OH. In another embodiment, R3 is NH2- In yet another embodiment R3 is -NH-.
In a further embodiment, with respect to compounds of formula I, L is a bond, a CrC5 alkylene group; and R3 is a heterocycloalkyl group containg -NH-.
In a further embodiment, with respect to compounds of forrflula I, A is CR R .
In a further embodiment, with respect to compounds of formula I, A is CH2.
In one particular embodiment, with respect to compounds of formula I, A is CO.
In a further embodiment, with respect to compounds of formula I, B and Y are independently selected from CR2a and CR2aR2b.
-5-

In a further embodiment, with respect to compounds of formula I B and Y are independently selected from CR2aR2b and the dotted bond is a single bond.
In a further embodiment, with respect to compounds of formula I, B and Y may all represent CH2 and the dotted bond is a single bond.
In a further embodiment, with respect to compounds of formula I, B and Y are independently selected from CR2a and the dotted bond is a double bond.
In a further embodiment, with respect to compounds of formula I, B and Y may all represent CH and the dotted bond is a double bond.
In a further embodiment, with respect to compounds of formula I, n is 0,1 or 2. In one particular embodiment, n is 1.
In another embodiment, with respect to compounds of formula I, each of R2 and R2 of
the group is H or Me. In one particular embodiment, each of R2 and R2 is H.
In a further embodiment, with respect to compounds of formula I, one of R" and R2 of
the group may be selected from Me, Et, halo and CI, and the other is H.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted aryl. In one particular embodiment, R1 is substituted phenyl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted naphthyl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted naphthyl.
In a further embodiment, with respect to compounds of formula I, R! is substituted or unsubstituted heteroaryl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted pyridyl, substituted or unsubstituted quinoline, substituted or unsubstituted benzodioxole, substituted or unsubstituted benzodioxane, substituted or unsubstituted benzofuran, substituted or unsubstituted benzothiophene, and substituted or unsubstituted benzodioxepine.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted adamantyl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted cyclopropyl, cyclopentyl, cyclohexyl or cycloheptyl.
In a further embodiment, with respect to compounds of formula I, each of W and W is N.
-6-

In a further embodiment, with respect to compounds of formula I, each of W, Z and W is CR4. In one particular embodiment, each of W, Z and W is CH.
In a further embodiment, with respect to compounds of formula I, each of W and Z is CR4, W is CR5 and R5 is selected from H, alkyl or halo. In one embodiment.R5 is halo or alkyl. In a particular embodiment, R5 is H or halo. In a yet further particular embodiment, R5 is H, CI, F or Me.
In a further aspect, the present invention provides pharmaceutical compositions comprising a bicycloheteroaryl compound of the invention, and a pharmaceutical carrier, excipient or diluent. In this aspect of the invention, the pharmaceutical composition can comprise one or more of the compounds described herein. Moreover, the compounds of the present invention useful in the pharmaceutical compositions and treatment methods disclosed herein, are all pharmaceutically acceptable as prepared and used.
In a further aspect of the invention, this invention provides a method of treating a mammal susceptible to or afflicted with a condition from among those listed herein, and particularly, such condition as may be associated with e.g. inflammation, such as rheumatoid arthritis, osteoarthritis, uveitis, asthma, myocardial infarction, traumatic brain injury; septic shock, atherosclerosis, chronic pulmonary obstructive disease (COPD), acute spinal cord injury, inflammatory bowel disease and immune dysfunction, including autoimmune disorders, which method comprises administering an effective amount of one or more of the pharmaceutical compositions just described.
In yet another method of treatment aspect, this invention provides a method of treating a mammal susceptible to or afflicted with a condition that is causally related to aberrant P2X7 receptor activity, and that for example, gives rise to pain responses or that relates to imbalances in the maintenance of basal activity of sensory nerves. The amine compounds of the invention have use as analgesics for the treatment of pain of various geneses or etiology, for example acute, inflammatory pain (such as pain associated with osteoarthritis and rheumatoid arthritis); various neuropathic pain syndromes (such as post-herpetic neuralgia, trigeminal neuralgia, reflex sympathetic dystrophy, diabetic neuropathy, Guillian Barre syndrome, fibromyalgia, phantom limb pain, post-masectomy pain, peripheral neuropathy, HIV neuropathy, and chemotherapy-induced and other iatrogenic neuropathies); visceral pain, (such as that associated with gastroesophageal reflex disease, irritable bowel syndrome, inflammatory bowel disease, pancreatitis, and various gynecological and urological disorders), dental pain and headache (such as migraine, cluster headache and tension headache).
In additional method of treatment aspects, this invention provides methods of treating a mammal susceptible to or afflicted with conditions that are causally related to abnormal activity of the P2X7 receptor, such as neurodegenerative diseases and disorders including, for example, Parkinson's disease, multiple sclerosis; diseases and disorders which are mediated by or result in neuroinflammation such as, for example traumatic brain injury and encephalitis; centrally-mediated neuropsychiatric diseases and disorders such as, for example depression mania, bipolar disease, anxiety, schizophrenia, eating disorders, sleep disorders and cognition disorders; epilepsy and seizure disorders; prostate, bladder and bowel dysfunction such as, for example urinary incontinence, urinary hesitancy, rectal
-7-

hypersensitivity, fecal incontinence, benign prostatic hypertrophy and inflammatory bowel disease; respiratory and airway disease and disorders such as, for example, allergic rhinitis, asthma and reactive airway disease and chronic obstructive pulmonary disease; diseases and disorders which are mediated by or result in inflammation such as, for example rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus such as, for example psoriasis; obesity; lipid disorders; cancer; blood pressure; spinal cord injury; and cardiovascular and renal disorders method comprises administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions just described.
In additional aspects, this invention provides methods for synthesizing the compounds of the invention, with representative synthetic protocols and pathways disclosed later on herein.
Accordingly, it is a principal object of this invention to provide a novel series of compounds, which can modify the activity of the P2X7 receptor and thus avert or treat any maladies that may be causally related thereto.
It is further an object of this invention to provide a series of compounds that can treat or alleviate maladies or symptoms of same, such as pain and inflammation, that may be causally related to the activation of the P2X7 receptor.
A still further object of this invention is to provide pharmaceutical compositions that are effective in the treatment or prevention of a variety of disease states, including the diseases associated with the central nervous system, cardiovascular conditions, chronic pulmonary obstructive disease COPD), inflammatory bowel disease, rheumatoid arthritis, osteoarthritis, and other diseases where an inflammatory component is present.
Other objects and advantages will become apparent to those skilled in the art from a consideration of the ensuing detailed description.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
When describing the compounds, pharmaceutical compositions containing such compounds and methods of using such compounds and compositions, the following terms have the following meanings unless otherwise indicated. It should also be understood that any of the moieties defined forth below may be substituted with a variety of substituents, and that the respective definitions are intended to include such substituted moieties within their scope. By way of non-limiting example, such substituents may include e.g. halo (such as fluoro, chloro, bromo), -CN, -CF3, -OH, -OCF3, C2-C6 alkenyl, C3-C6 alkynyl, CrC6 alkoxy, aryl and di-C1-C6 alkylamino. It should be further understood that the terms "groups" and "radicals" can be considered interchangeable when used herein.
"Acyl" refers to a radical -C(0)R20, where R20 is hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl as defined
-8-

herein. Representative examples include, but are not limited to, formyl, acetyl, cylcohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl, b^nzylcarbonyl and the like.
"Acylamino" refers to a radical -NR2lC(0)R22, where R21 is hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl heteroarylalkyl and R22 is hydrogen, alkyl, alkoxy, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl or heteroarylalkyl, as defined herein. Representative examples include, but are not limited to, formylamino, acetylamino, cyclohexyIcarbc>nylarnino, cyclohexylmethyl-carbonylamino, benzoylamino, benzylcarbonylamino and the like.
"Acyloxy" refers to the group -OC(0)R23 where R23 is hydrogen, alkyl, aryl or cycloalkyl.
"Substituted alkenyl" includes those groups recited in tfre definition of "substituted" herein, and particularly refers to an alkenyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 tc> 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, fhioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryl-S(0)2-.
"Alkoxy" refers to the group -OR24 where R24 is alkyl. Particular alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropo^y, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
"Substituted alkoxy" includes those groups recited in the definition of "substituted" herein, and particularly refers to an alkoxy group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, heteroaryl, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2-andaryl-S(0)2-.
"Alkoxycarbonylamino" refers to the group -NR25C(0)OR26, where R25 is hydrogen, alkyl, aryl or cycloalkyl, and R26 is alkyl or cycloalkyl.
"Alkyl" refers to monovalent saturated alkane radical groups particularly having up to about 11 carbon atoms, more particularly as a lower alkyl, from 1 to 8 carbon atoms and still more particularly, from 1 to 6 carbon atoms. The hydrocarbon chain may be either straight-chained or branched. This term is exemplified by groups such as methyl, ethyl, n-propyl, isopropyl, K-butyl, iso-buty\, tert-butyl, -hexyl, -octyl, tert-octyl and the like. The term "lower alkyl" refers to alkyl groups having 1 to 6 carbon atoms. The term "alkyl" also includes "cycloalkyls" as defined below.
-9-

"Substituted alkyl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an alkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, heteroaryl, keto, nitro, thioEilkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2-, and aryl-S(0)2-.
"Alkylene" refers to divalent saturated alkene radical groups having 1 to 11 carbon atoms and more particularly 1 to 6 carbon atoms which can be straight-chained or branched. This term is exemplified by groups such as methylene (-CH2-), ethylene (-CH2CH2-), the propylene isomers (e.g., -CH2CH2CH2- and -CH(CH3)CH2-) and the like.
"Substituted alkylene" includes those groups recited in the definition of "substituted" herein, and particularly refers to an alkylene group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 tc 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, amino-carbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryI-S(0)2-.
"Alkenyl" refers to monovalent olefinically unsaturated hydrocarbyl groups preferably having 2 to 11 carbon atoms, particularly, from 2 to 8 carbon atoms, and more particularly, from 2 to 6 carbon atoms, which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of olefmic unsaturation. Particular alkenyl groups include ethenyl (-CH=CH2), n-propenyl (-CH2CH=CH2), isopropenyl (-C(CH3)=CH2), vinyl and substituted vinyl, and the like.
"Alkenylene" refers to divalent olefinically unsaturated hydrocarbyl groups particularly having up to about 11 carbon atoms and more particularly 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of olefinic unsaturation. This term is exemplified by groups such as ethenylene (-CH=CH-), the propenylene isomers (e.g., -CH=CHCH2- and -C(CH3)=CH- and -CH=C(CH3)-) and the like.
"Alkynyl" refers to acetylenically or alkynically unsaturated hydrocarbyl groups particularly having 2 to 11 carbon atoms and more particularly 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of alkynyl unsaturation. Particular non-limiting examples of alkynyl groups include acetylenic, ethynyl (-C CH), propargyl (-CH2C CH), and the like.
"Substituted alkynyl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an alkynyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl,
-10-

aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryl-S(0)2-.
"Alkanoyl" or "acyl" as used herein refers to the group R27-C(0)-, where R27 is hydrogen or alkyl as defined above.
"Aryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like. Particularly, an aryl group comprises from 6 to 14 carbon atoms.
"Substituted Aryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an aryl group that may optionally be substituted with 1 or more substituents, for instance from 1 to 5 substituents, particularly 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkoxycarbonyl, alkyl, substituted alkyl, alkynyl, substituted alkynyl, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryl-S(0)2-.
"Fused Aryl" refers to an aryl having two of its ring carbon in common with a second aryl ring or with an aliphatic ring.
"Alkaryl" refers to an aryl group, as defined above, substituted with one or more alkyl groups, as defined above.
"Aralkyl" or "arylalkyl" refers to an alkyl group, as defined above, substituted with one or more aryl groups, as defined above.
"Aryloxy" refers to -O-aryl groups wherein "aryl" is as defined above.
"Alkylamino" refers to the group alkyl-NR28R29, wherein each of R28 and R29 are independently selected from hydrogen and alkyl.
"Arylamino" refers to the group aryl-NR30R31, wherein each of R30 and R31 are independently selected from hydrogen, aryl and heteroaryl.
"Alkoxyarnino" refers to a radical -N(H)OR32 where R32 represents an alkyl or cycloalkyl group as defined herein.
- 11 -

"Alkoxycarbonyl" refers to a radical -C(0)-alkoxy where alkoxy is as defined herein.
"Alkylarylamino" refers to a radical -NR33R34 where R33 represents an alkyl or cycloalkyl group and R34 is an aryl as defined herein.
"Alkylsulfonyl" refers to a radical -S(0)2R35 where R35 is an alkyl or cycloalkyl group as defined herein. Representative examples include, but are not limited to, methylsulfonyl, ethylsulfonyl, propylsulfonyl, butylsulfonyl and the like.
"Alkylsulfinyl" refers to a radical -S(0)R35 where R35 is an alkyl or cycloalkyl group as defined herein. Representative examples include, but are not limited to, methylsulfinyl, ethylsulfinyl, propylsulfinyl, butylsulfmyl and the like.
"Alkylthio" refers to a radical -SR35 where R35 is an alkyl or cycloalkyl group as defined herein that may be optionally substituted as defined herein. Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, and the like.
"Amino" refers to the radical -NH2.
"Substituted amino" includes those groups recited in the definition of "substituted" herein, and particularly refers to the group -N(R36)2 where each R36 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, cycloalkyl, substituted cycloalkyl, and where both R groups are joined to form an alkylene group. When both R groups are hydrogen, -N(R36)2 is an amino group.
"Ammocarbonyl" refers to the group -C(0)NR37R37 where each R37 is independently hydrogen, alkyl, aryl and cycloalkyl, or where the R37 groups are joined to form an alkylene group.
"Aminocarbonylamino" refers to the group -NR38C(0)NR38R38 where each R38 is independently hydrogen, alkyl, aryl or cycloalkyl, or where two R groups are joined to form an alkylene group.
"Aminocarbonyloxy" refers to the group -OC(0)NR39R39 where each R39 is independently hydrogen, alkyl, aryl or cycloalky, or where the R groups are joined to form an alkylene group.
"Arylalkyloxy" refers to an -O-arylalkyl radical where arylalkyl is as defined herein.
"Arylamino" means a radical -NHR40 where R40 represents an aryl group as defined herein.
"Aryloxycarbonyl" refers to a radical -C(0)-0-aryI where aryl is as defined herein.
"Arylsulfonyl" refers to a radical -S(0)2R41 where R41 is an aryl or heteroaryl group as defined herein.
-12-

"Azido" refers to the radical -N3.
"Bicycloaryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent bicycloaromatic ring system. Typical bicycloaryl groups include, but are not limited to, groups derived from indane, indene, naphthalene, tetrahydronaphthalene, and the like. Particularly, an aryl group comprises from 8 to 11 carbon atoms.
"Bicycloheteroaryl" refers to a monovalent bicycloheteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent bicycloheteroaromatic ring system. Typical bicycloheteroaryl groups include, but are not limited to, groups derived from benzofuran, benzimidazole, benzindazole, benzdioxane, chromene, chromane, cinnoline, phthalazine, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, benzothiazole, benzoxazole, naphthyridine, benzoxadiazole, pteridine, purine, benzopyran, benzpyrazine, pyridopyrimidine, quinazoline, quinoline, quinolizine, quinoxaline, benzomofphan, tetrahydroisoquinoline, tetrahydroquinoline, and the like. Preferably, the bicycloheteroaryl group is between 9-11 membered bicycloheteroaryl, with 5-10 membered heteroaryl being particularly preferred. Particular bicycloheteroaryl groups are those derived from benzothiophene, benzofuran, benzothiazole, indole, quinoline, isoquinoline, benzimidazole, benzoxazole and benzdioxane.
"Carbamoyl" refers to the radical -C(0)N(R42)2 where each R42 group is independently hydrogen, alkyl, cycloalkyl or aryl, as defined herein, which may be optionally substituted as defined herein.
"Carboxy" refers to the radical -C(0)OH.
"Carboxyamino" refers to the radical -N(H)C(0)OH.
"Cycloalkyl" refers to cyclic hydrocarbyl groups having from 3 to about 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring systems, which optionally can be substituted with from 1 to 3 alkyl groups. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, 1-methylcyclopropyl 2-methylcyclopentyl, 2-methylcyclooctyl, and the like, and multiple ring structures such as adamantanyl, and the like.
"Substituted cycloalkyl" includes those groups recited in the definition of "substituted" herein, and particularly refers to a cycloalkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryl-S(0)r.
-13-

"Cycloalkoxy" refers to the group -OR where R is cycloalkyl. Such cycloalkoxy groups include, by way of example, cyclopentoxy, cyclohexoxy and the like.
"Cycloalkenyl" refeTS to cyclic hydrocarbyl groups having from 3 to 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring systems and having at least one and particularly from 1 to 2 sites of olefmic unsaturation. Such cycloalkenyl groups include, by way of example, single ring structures such as cyclohexenyl, cyclopentenyl, cyclopropenyl, and the like.
"Substituted cycloalkenyl" includes those groups recited in the definition of "substituted" herein, and particularly refers to a cycloalkenyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryl-S(0)2-.
"Fused Cycloalkenyl" refers to a cycloalkenyl having two of its ring carbon atoms in common with a second aliphatic or aromatic ring and having its olefmic unsaturation located to impart aromaticity to the cycloalkenyl ring.
"Cyanato" refers to the radical -OCN.
"Cyano" refers to the radical -CN.
"Dialkylamino" means a radical -NR^R45 where R44 and R45 independently represent an alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroaryl, or substituted heteroaryl group as defined herein.
"Ethenyl" refers to substituted or unsubstituted -(C=C)-.
"Ethylene" refers to substituted or unsubstituted -(C-C)-.
"Ethynyl" refers to -(C C).
"Halo" or "halogen" refers to fluoro, chloro, bromo and iodo. Preferred halo groups are either fluoro or chloro.
"Hydroxy" refers to the radical -OH.
"Nitro" refers to the radical -N02.
"Substituted" refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s). Typical substituents include, but are not limited to, -X, -R46, -0 =0, -OR46, -SR46, -S', -S, -NR46R47, =NR46, -
- 14-

CX3, -CF3, -CN, -OCN, -SCN, -NO, -N02, =N2, -N3, -S(0)2()-, -S(0)2OH, -S^R46, -0S(02)0", -OS(0)2R46, -P(0)(0)2, -P(0)(OR46)(0")5 -OP(0)(OR46)(OR47), -C(0)R46, -C(S)R46, -C(0)OR46, -C(0)NR46R47, -C(0)0\ -C(S)OR46, -NR48C(0)NR46R47, -NR48C(S)NR46R47, -NR49C(NR48)NR46R47 and -C(NR48)NR46R47, where each X is independently a halogen; each R46, R47, R48 and R49 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted alkyl, arylalkyl, substituted alkyl, cycloalkyl, substituted alkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl, substituted heteroarylalkyl, -NR50R51, -C(O)RS0 or -S(0)2R50 or optionally R50 and R51 together with the atom to which they are both attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring; and R50 and R51 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted alkyl, arylalkyl, substituted alkyl, cycloalkyl, substituted alkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl.
Examples of representative substituted aryls include the following:





In these formulae one of R52 and R53 may be hydrogen and at least one of R52 and R53 is each independently selected from alkyl, alkenyl, alkynyl, cycloheteroalkyl, alkanoyl, alkoxy, aryloxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR54COR55, NR54SOR55, NR54S02R57, COOalkyl, COOaryl, CONR54R55, CONR54OR55, NR^R55, S02NR54R55, S-alkyl, S-alkyl, SOalkyl, S02alkyl, Saryl, SOaryl, S02aryl; or R52 and R53 may be joined to form a cyclic ring (saturated or unsaturated) from 5 to 8 atoms, optionally containing one or more heteroatoms selected from the group N, O or S. R54, R55, and R56 are independently hydrogen, alkyl, alkenyl, alkynyl, perfluoroalkyl, cycloalkyl, cycloheteroalkyl, aryl, substituted aryl, heteroaryl, substituted or hetero alkyl or the like.
"Hetero" when used to describe a compound or a group present on a compound means that one or more carbon atoms in the compound or group have been replaced by a nitrogen, oxygen, or sulfur heteroatom. Hetero may be applied to any of the hydrocarbyl groups described above such as alkyl, e.g. heteroalkyl, cycloalkyl, e.g. cycloheteroalkyl, aryl, e.g. heteroaryl, cycloalkenyl, cycloheteroalkenyl, and the like having from 1 to 5, and especially from 1 to 3 heteroatoms.
"Heteroaryl" refers to a monovalent heteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring system. Typical heteroaryl groups include, but are not limited to, groups derived from acridine, arsindole, carbazole, p-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, and the like. Preferably, the heteroaryl group is between 5-15 membered
-15-

heteroaryl, with 5-10 membered heteroaryl being particularly preferred. Particular heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole and pyrazine.










wherein each Y is selected from carbonyl, N, NR58, O, and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
As used herein, the term "cycloheteroalkyl" refers to a stable heterocyclic non-aromatic ring and fused rings containing one or more heteroatoms independently selected from N, O and S. A fused heterocyclic ring system may include carbocyclic rings and need only include one heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, piperazinyl, homopiperazinyl, piperidinyl and morpholinyl, and are shown in the following illustrative examples:





n

Q

,58
58
wherein each X is selected from CR582, NR36, 0 and S; and each Y is selected from NR O and S; and R5S is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like. These cycloheteroalkyl rings may be optionally substituted with one or more groups selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)r and aryl-S(0)2-. Substituting groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea derivatives.
-16-

Examples of representative cycloheteroalkenyls include the following:






wherein each X is selected from CR582, NR58, O and S; and each Y is selected from carbonyl, N, NR58, 0 and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
Examples of representative aryl having hetero atoms containing substitution include the following:



.58
wherein each X is selected from C-R582 NRM, O and S; and each Y is selected from carbonyl, NR58, O and S; and R58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.
"Hetero substituent" refers to a halo, O, S or N atom-containing functionality that may be present as an R4 in a R4C group present as substituents directly on A, B, W, Y or Z of the compounds of this invention or may be present as a substituent in the "substituted" aryl and aliphatic groups present in the compounds.
Examples of hetero substituents include:
-halo,
-N02, -NH2, -NHR59, -N(R59)2,
-NRCOR, -NR59SOR59, -NR59S02R59, OH, CN,
-C02H,
-R59-OH, -O-R59, -COOR59,
-CON(R59)2,-CONROR59,
-S03H,-R59-S,-S02N(R59)2,
-S(0)R59, -S(0)2R59 wherein each R59 is independently an aryl or aliphatic, optionally with substitution. Among hetero substituents containing R59 groups, preference is given to those materials having aryl and alkyl R59 groups as defined herein. Preferred hetero substituents are those listed above.
"Hydrogen bond donor" group refers to a group containg O-H, N-H functionality. Examples of "hydrogen bond donor" groups include -OH, -NH2, and -NH-R59a and
wherein R is alkyl, cycloalkyl, acyl, aryl, or heteroaryl.
"Dihydroxyphosphoryl" refers to the .radical -PO(OH)2.
-17-

"Substituted dihydroxyphosphoryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to a dihydroxyphosphoryl radical wherein one or both of the hydroxyl groups are substituted. Suitable substituents are described in detail below.
"Aminohydroxyphosphoryl" refers to the radical -PO(OH)NH2.
"Substituted aminohydroxyphosphoryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an aminohydroxyphosphoryl wherein the amino group is substituted with one or two substituents. Suitable substituents are described in detail below. In certain embodiments, the hydroxyl group can also be substituted,
"Thioalkoxy" refers to the group -SR60 where R60 is alkyl.
"Substituted thioalkoxy" includes those groups recited in the definition of "substituted" herein, and particularly refers to a thioalkoxy group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto> nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0)2- and aryl-S(0)r.
"Sulfanyl" refers to the radical HS-. "Substituted sulfanyl" refers to a radical such as RS-wherein R is any substituent described herein.
"Sulfonyl" refers to the divalent radical -S(02)-. "Substituted sulfonyl" refers to a radical such as R6,-(02)S- wherein R6i is any substituent described herein. "Aminosulfonyl" or "Sulfonamide" refers to the radical H2N(02)S-, and "substituted aminosulfonyl" or "substituted sulfonamide" refers to a radical such as R622N(02)S- wherein each R62 is independently any substituent described herein.
"Sulfone" refers to the group -S02R63. In particular embodiments, R63 is selected from H, lower alkyl, alkyl, aryl and heteroaryl.
"Thioaryloxy" refers to the group -SR64 where R64 is aryl.
"Thioketo" refers to the group =S.
"Thiol" refers to the group -SH.
One having ordinary skill in the art of organic synthesis will recognize that the maximum number of heteroatoms in a stable, chemically feasible heterocyclic ring, whether it is aromatic or non aromatic, is determined by the size of the ring, the degree of unsaturation and the valence of the heteroatoms. In general, a heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.
-18-

"Pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.
"Pharmaceutically acceptable salt" refers to a salt of a compound of the invention that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include- (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicycIo[2.2.2]-oct-2-ene-l-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine and the like. Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylamrnonium, and the like; and when the compound contains a basic functionality, salts of non toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like. The term "pharmaceutically acceptable cation" refers to a non toxic, acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkylamrnonium cations, and the like.
"Pharmaceutically acceptable vehicle" refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
"Preventing" or "prevention" refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).
"Prodrugs" refers to compounds, including derivatives of the compounds of the invention,which have cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention which are pharmaceutically active in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
"Solvate" refers to forms of the compound that are associated with a solvent, usually by a solvolysis reaction. Conventional solvents include water, ethanol, acetic acid and the like. The compounds of the invention may be prepared e.g. in crystalline form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates,
-19-

such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates.
"Subject" includes humans. The terms "human," "patient" and "subject" are used interchangeably herein.
"Therapeutically effective amount" means the amount of a compound that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically effective amount" can vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
"Treating" or "treatment" of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment "treating" or "treatment" refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, "treating" or "treatment" refers to delaying the onset of the disease or disorder.
Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but in the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. Preferred are the C1 to C8 alkyl, C2-C8 alkenyl, aryl, C7-C12 substituted aryl, and C7-C]2 arylalkyl esters of the compounds of the invention.
As used herein, the term "isotopic variant" refers to a compound that contains unnatural proportions of isotopes at one or more of the atoms that constitute such compound. For example, an "isotopic variant" of a compound can contain one or more non-radioactive isotopes, such as for example, deuterium (2H or D), carbon-13 (l3C), nitrogen-15 (15N), or the like. It will be understood that, in a compound where such isotopic substitution is made, the following atoms, where present, may vary, so that for example, any hydrogen may be 2H/D, any carbon may be l3C, or any nitrogen may be l5N, and that the presence and placement of such atoms may be determined within the skill of the art. Likewise, the invention may include the preparation of isotopic variants with radioisotopes, in the instance for example, where the resulting compounds may be used for drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation
-20-

and ready means of detection. Further, compounds may be prepared that are substituted with positron emitting isotopes, such as 11C, 18F, 150 and ,3N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
All isotopic variants of the compounds provided herein, radioactive or not, are intended to be encompassed within the scope of the invention.
It is also to be understood that compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers". Isomers that differ in the arrangement of then-atoms in space are termed "stereoisomers".
Stereoisomers that are not mirror images of one another are termed "diastereomers" and those that are non-superimposable mirror images of each other are termed "enantiomers". When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible. An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or Jevorotatory (i.e., as (+) or (-)-isomers respectively). A chiral compound can exist as either individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture".
"Tautomers" refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of n electrons and an atom (usually H). For example, enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base. Another example of tautomerism is the aci- and nitro- forms of phenylnitromethane, that are likewise formed by treatment with acid or base.
Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.
The compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)- stereoisomers or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art.
THE COMPOUNDS The present invention provides bicycloheteroaryl compounds useful for preventing and/or treating a broad range of conditions, associated with abnormalities in the activity of the P2X7 receptor, among them, rheumatoid arthritis, Parkinson's disease, uveitis, asthma, cardiovascular conditions such as myocardial infarction, the treatment and prophylaxis of pain syndromes (acute and chronic or neuropathic), traumatic brain injury, acute spinal
-21-

cord injury, neurodegenerative disorders, inflammatory bowel disease and immune dysfunctions such as autoimmune disorders or conditions, in mammals.
In a first aspect of the invention, bicycloheteroaryl compounds are disclosed that are capable of capable of modulating the activity of the P2X7 receptor in vivo, having a formula (I):

wherein
A is CR2aR2b or CO; B and Y are independently selected from CR2a and CR2aR2b;
W, W and Z are independently selected from CR4 and N, provided that all three of
W and Z are not N at the same time;
Ll is a bond, SO, S02 or substituted or unsubstituted C1-C5 alkylene;
n is 0,1,2,3 or 4;
R1 is selected from a substituted or unsubstituted 3-13 membered cycloalkyl,
heterocycloalkyl, aryl and heteroaryl ring;
each of R2a, R2b, R2' and R2 is independently selected from hydrogen, halo, and
substituted or unsubstituted Q-C6 alkyl; or any of R2' and R2 join together to form a
cycloalkyl or cycloheteroalkyl ring of 3-7 atoms;
R3 is selected from hydrogen, a hydrogen bond donor group, substituted or unsubstituted
cycloalkyl, substituted or unsubstituted alkyl, substituted or unsubstituted
heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl,
substituted or unsubstituted bicycloaryl, and substituted or unsubstituted
bicycloheteroaryl;
R4 is independently selected from H, alkyl, substituted alkyl, acyl, substituted acyl,
substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino,
substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy, alkoxycarbonyl,
substituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, arylalkyloxy,
substituted arylalkyloxy, amino, aryl, substituted aryl, arylalkyl, substituted or
unsubstituted sulfoxide, substituted or unsubstituted sulfone, substituted or unsubstituted
sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted
arylsulfonyl, sulfuric acid, sulfuric acid ester, substituted or unsubstituted
dihydroxyphosphoryl, substituted or unsubstituted aminodihydroxyphosphoryl, azido,
carboxy, substituted or unsubstituted carbamoyl, cyano, substituted or unsubstituted
cycloalkyl, substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted
dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl, substituted or
unsubstituted heteroalkyl, hydroxy, nitro, and thio;
and the dotted bond is a single or a double bond;
or a pharmaceutically acceptable salt, solvate or prodrug thereof;
and stereoisomers, isotopic variants and tautomers thereof.
In a further embodiment, with respect to compounds of formulae I, n is 0-4.
-22-

In a further embodiment, with respect to compounds of formula I, L1 is a bond, or a CrC5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, hydroxy, hydroxyalkyl, aminoalkyl, alkylaminoalkyt, dialkylaminoalkyl, halogen, carbamoyl, oxo, aryl, and Cr6 alkoxy.
In a further embodiment, with respect to compounds of formula I, L1 is CrC5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, oxo, aryl, hydroxyl, and hydroxyalkyl
In a further embodiment, with respect to compounds of formula I, L1 is a C1-C5 alkylene group substituted with two alkyl groups and wherein any two alkyl groups on the same carbon atom can join together to form a cycloalkyl or cycloheteroalkyl ring of 3-7 atoms.
In a further embodiment, with respect to compounds of formula I, L is a bond, a C1-C5 alkylene group; and R3 is a hydrogen bond donor group. In one embodiment, R3 is -OH. In another embodiment, R3 is NH2. In yet another embodiment R3 is -NH-.
In a further embodiment, with respect to compounds of formula I, L is a bond, a C1-C5 alkylene group substituted with oxo; and R3 is a hydrogen bond donor group. In one embodiment, R3 is -OH. In another embodiment, R3 is NH2. In yet another embodiment R3is-NH-.
In a further embodiment, with respect to compounds of formula I, L1 is a bond, a Ci-C5 alkylene group; and R3 is a heterocycloalkyl group containg -NH-.
In a further embodiment, with respect to compounds of formula I, A is CR aR .
In a further embodiment, with respect to compounds of formula I, A is CH2.
In one particular embodiment, with respect to compounds of formula I, A is CO.
In a further embodiment, with respect to compounds of formula I, B and Y are independently selected from CR2a and CR2aR2b.
In a further embodiment, with respect to compounds of formula I B and Y are independently selected from CR2aR2b and the dotted bond is a single bond.
In a further embodiment, with respect to compounds of formula I, B and Y may all represent CH2 and the dotted bond is a single bond.
In a further embodiment, with respect to compounds of formula I, B and Y are independently selected from CR2a and the dotted bond is a double bond.
In a further embodiment, with respect to compounds of formula I, B and Y may all represent CH and the dotted bond is a double bond.
In a further embodiment, with respect to compounds of formula I, n is 0,1 or 2. In one particular embodiment, n is 1.
-23-


is H or Me. In one particular embodiment, each of R and R is H.


group may be selected from Me, Et, halo and CI, and the other is H.
In a further embodiment, with respect to compounds of formula I, one of R2 and R2 of
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted aryl. In one particular embodiment, R1 is substituted phenyl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted naphthyl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted heteroaryl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted pyridyl, substituted or unsubstituted quinoline, substituted or unsubstituted benzodioxole, substituted or unsubstituted benzodioxane, substituted or unsubstituted benzofuran, substituted or unsubstituted benzothiophene, and substituted or unsubstituted benzodioxepine.
In a farther embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted adamantyl.
In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted cyclopropyl, cyclopentyl, cyclohexyl or cycloheptyl.
In a further embodiment, with respect to compounds of formula I, each of W and W is N.
In a further embodiment, with respect to compounds of formula I, each of W, Z and W* is CR4. In one particular embodiment, each of W, Z and W is CH.
In a further embodiment, with respect to compounds of formula I, each of W and Z is CR4, W is CR5 and R5 is selected from H, alkyl or halo. In one embodiment.R5 is halo or alkyl. In a particular embodiment, R5 is H or halo. In a yet further particular embodiment, R5 is H, Cl, F or Me.
In another embodiment, with respect to compounds of formulae I, the compound is according to formula II, III or IV:


wherein
W is CR4; Z is CR4;
L\ R1 R2', R2', R3 and R4 are as described for formula I;
and R5 is selected from H, alkyl, substituted alkyl, acyl, substituted acyl, substituted or
unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or
unsubstituted alkythio, substituted or unsubstituted alkoxy, alkoxycarbonyl, substituted
alkoxycarbonyl, substituted or unsubstituted alkylarylamino, arylalkyloxy, substituted
arylalkyloxy, amino, aryl, substituted aryl, arylalkyl, substituted or unsubstituted
sulfoxide, substituted or unsubstituted sulfone, substituted or unsubstituted sulfanyl,
substituted or unsubstituted aminosulfonyl, substituted or unsubstituted arylsulfonyl,
sulfuric acid, sulfuric acid ester, substituted or unsubstituted dihydroxyphosphoryl,
substituted or unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or
unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl, substituted or
unsubstituted cycloheteroalkyl, substituted or unsubstituted dialkylamino, halo,
heteroaryloxy, substituted or unsubstituted heteroaryl, substituted or unsubstituted
heteroalkyl, hydroxy, nitro, and thio;
or a pharmaceutically acceptable salt, solvate or prodrug thereof;
and stereoisomers, isotopic variants and tautomers thereof.
In another embodiment, with respect to compounds of formulae II-IV, each of R2 and R2 isH.
In another embodiment, with respect to compounds of formulae II-IV, R2 is halo; and R2 isH.
In another embodiment, with respect to compounds of formulae II-IV, R2 is CI or F; and R2isH.
In another embodiment, with respect to compounds of formulae II-IV, R2 is Me or Et; and R2" is H.
In another embodiment, with respect to compounds of formulae II-IV, each of R2 and R2 is Me.
In a more particular embodiment, with respect to compounds of formulae II-IV, R2 is Me; and R2" is H.
In another embodiment, with respect to compounds of formulae II-IV, each of R1 is substituted or unsubstituted aryl.
In another embodiment, with respect to compounds of formulae II-IV, each of R1 is substituted or unsubstituted phenyl or naphthalene.
In another embodiment, with respect to compounds of formulae II-IV, each of Rl is substituted or unsubstituted naphthalene.
In another embodiment, with respect to compounds of formulae II-IV, each of R1 is
unsubstituted naphthalene. ■*
-25-

In another embodiment, with respect to compounds of formulae II-IV, each of Rl is substituted or unsubstituted phenyl.
In another embodiment, with respect to compounds of formulae II-IV, each of R1 is substituted or unsubstituted heteroaryl.
In another embodiment, with respect to compounds of formulae II-IV, each of R1 is substituted or unsubstituted pyridyl, substituted or unsubstituted quinoline, substituted or unsubstituted benzodioxole, substituted or unsubstituted benzodioxane, substituted or unsubstituted benzofuran, substituted or unsubstituted benzothiophene, and substituted or unsubstituted benzodioxepine.
In another embodiment, with respect to compounds of formulae II-IV, R1 is substituted or unsubstituted adamantyl.
In another embodiment, with respect to compounds of formulae II-IV, R1 is substituted or unsubstituted cyclopropyl, cyclopentyl, cyclohexyl or cycloheptyl
In another embodiment, with respect to compounds of formulae I, the compound is is according to formula V, VI or VII:

VI Vll
or
wherein
W is CR4; Z is CR4; L1, R1 Rr, Rr, R3 and R4 are as described for formula I; R5 is as described for formulae II-IV;
each R4a is selected from H, alkyl, substituted alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy, aryloxy, alkoxycarbonyl, substituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, arylalkyloxy, substituted arylalkyloxy, amino, aryl, substituted aryl, arylalkyl, substituted or unsubstituted sulfoxide, substituted or unsubstituted sulfone, substituted or unsubstituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted arylsulfonyl, sulfuric acid, sulfuric acid ester, substituted or unsubstituted dihydroxyphosphoryl, substituted or unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroalkyl, hydroxy, nitro, and thio; and m is selected from 0-5; or a pharmaceutical^ acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
-26-

With respect to the compounds of the invention wherein m is 0-5 as set forth above, and at any and all locations herein, it is to be understood that when m=0, the ring is unsubstituted.
In one embodiment, with respect to compounds of formulae V-VII, each of R2 and R2 is H.
In another embodiment, with respect to compounds of formulae V-VII, R2' is halo; and R2" is H.
In another embodiment, with respect to compounds of formulae V-VII, R2' is CI or F; and R2"isH.
In another embodiment, with respect to compounds of formulae V-VII, R2' is Me or Et; and R2" is H.
In another embodiment, with respect to compounds of formulae V-VII, each of R2 and R2" is Me.
In a more particular embodiment, with respect to compounds of formulae V-VII, R2 is Me; and R2" is H.
In another embodiment, with respect to compounds of formula I or la, the compound is according to formula VIII, IX or X:

>
wherein
W is CR4; Z is CR4;
L1, R3 and R4 are as described for formula I; m, R4a, and R5 are as described for formulae V-VII; R2 is H or Me; Cy is adamantyl, cyclohexyl or cycloheptyl; and R4b is independently selected from hydrogen, CrC4 alkyl and hydroxy; or a pharmaceutical^ acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
In one embodiment R2 is H or Me. In another embodiment, R2 is Me. In one particular embodiment, R2' is H.
In another embodiment, with respect to compounds of formulae V-X, m is 0,1,2 or 3.
In another embodiment, with respect to compounds of formulae V-X, m is 1 or 2. In a particular embodiment m is 1.
In another embodiment, with respect to compounds of formulae V-X, each of R4a is independently selected from Me, Et, Ph, CI, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3,
-27-

CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH=CH-C02H, SOMe, S02Me, S03H, S03Me, and
pyridyl.
In one embodiment, with respect to compounds of formula VIII, Cy is adamantyl and R4b is H. In another embodiment, R4b is Me. In another embodiment, R4b is OH and m is 1.
In one embodiment, with respect to compounds of formula VIII, Cy is cyclohexyl and R4b is H. In another embodiment, R4b is Me and m is 2. In further embodiment, R4b is Me and m is 4. In yet another embodiment R4b is OH and m is 1. In another embodiment, m is 3 andtwoofR4bs are each methyl and one ofR4bishydroxyI.
In one embodiment, with respect to compounds of formula VIII, Cy is cycloheptyl and R4b is H. In another embodiment R4b is Me and m is 2. In further embodiment R4b is Me and m is 1. In yet another embodiment R4b is OH and m is 1,
In one embodiment, with respect to compounds of formulae I-X, L1 is a bond.
In another embodiment, with respect to compounds of formulae I-X, L1 is CrC5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, oxo, aryl, hydroxyl, and hydroxyalkyl
In another embodiment, with respect to compounds of formulae I-X, L1 is a C|-C5 alkylene group substituted with two alkyl groups and wherein any two alkyl groups on the same carbon atom can join together to form a cycloalkyl or cycloheteroalkyl ring of 3-7 atoms.
In another embodiment, with respect to compounds of formulae I-X, R3 is selected from hydroxyl, amino, alkylamino or carbamoyl.
In another embodiment, with respect to compounds of formulae I-X, Li is CH2 and Rl is substituted or unsubstituted aryl or heteroaryl.
In another embodiment, with respect to compounds of formulae I-X, Li is CH2 and R1 is phenyl or pyridyl substituted with one or more substituents independently selected from halo, hydroxyl, amino, cyano, sulfo, sulfanyl, sulfmyl, amido, carboxy, ester, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, and sulfonamide.
In another embodiment, with respect to compounds of formulae I-X, L] is CH2 and R1 is phenyl or pyridyl substituted with one or more substituents independently selected from Me, Et, Ph, CI, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH=CH-C02H, SOMe, S02Me, S03H, S03Me, and pyridyl.
In another embodiment, with respect to compounds of formulae I-X, R3 is a hydrogen bond donor group.
In another embodiment, with respect to compounds of formulae I-X, R3 is selected from hydroxyl, amino, alkylamino or carbamoyl.
-28-

In another embodiment, with respect to compounds of formulae I-X, the group -LrR3 is

In a particular embodiment, with respect to compounds of formulae I-X, the group -LrR is

In another embodiment, with respect to compounds of formulae I-X, the group -LrR is selected from

In another embodiment, with respect to compounds of formulae I-X, the group -LrR3 is selected from


In another embodiment, with respect to compounds of formula I, the compound is according to formula XIa, Xlb, XIc, Xld, Xle, Xlf, Xlg, Xlh or XIj:

wherein m and R4a are as described for formulae V-VII; and R5 is H, alkyl, cycloalkyl or halo.
In another embodiment, with respect to compounds of formula I, the compound is according to formula Xlla, Xllb, or XIc:


wherein m and R4a are as described for formulae V-VII; R5 is H, alkyl, cycloalkyl or halo; and R2d is selected from hydrogen, alkyl, hydroxyalkyl, and substituted or unsubstituted phenyl. In one particular embodiment, R2d is hydrogen, methyl, i-Pr and hydroxymethyl. In another particular embodiment, R2d is phenyl. In another particular embodiment, R2d is hydrogen. In yet another particular embodiment, R2d is methyl.
XII la , Xltlb
In another embodiment, with respect to compounds of formula I, the compound is according to formula XHIa, XHIb, XIIIc, or XHId:


XIIIc Of xilld
wherein m and R4a are as described for formulae V-VII; and R5 is H, alkyl, cycloalkyl or
halo.
In one embodiment, with respect to compounds of formulae XIa-XIIId, m is 1, 2 or 3.
In another embodiment, with respect to compounds of formulae XIa-XIIId, m is 1 or 2. In a particular embodiment m is 2.
In another embodiment, with respect to compounds of XIa-XIIId, each of R4a is independently selected from Me, Et, Ph, CI, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH=CH-C02H, SOMe, S02Me, S03H, S03Me, and
pyridyl.
In another embodiment, with respect to compounds of V-XIIId, m is 1 and R4a is CF3.
In another embodiment, with respect to compounds of V-XIIId, m is 2 and R4a is F and CF3.
In another embodiment, with respect to compounds of V-XIIId, m is 2 and R4a is F and CI.
In one embodiment, with respect to compounds of formulae I-X, each of W and Z is independently CR4.
In one embodiment, with respect to compounds of formulae I-X, each of W and Z is independently CH.
In one embodiment, with respect to compounds of formulae I-X, W is N.
-31 -

In one embodiment, with respect to compounds of formulae I-X, W is N and Z is H.
In one embodiment, with respect to compounds of formulae II-XIIId, R5 is H.
In one embodiment, with respect to compounds of formulae II-XIIId, R5 is selected from alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl and halo. In one particular embodiment, R5 is selected from Me, cyclopropyl, CI, F and CF3.
In one embodiment, with respect to compounds of formulae II-XIIId, R5 is Me.
In one embodiment, with respect to compounds of formulae II-XIIId, R5 is CF3.
In one embodiment, with respect to compounds of formulae II-XIIId, R5 is F.
In a further embodiment with respect to compounds of formulae II-XIIId, R5 is CI.
In a farther embodiment with respect to compounds of formulae II-XIIId, R5 is cyclopropyl.
In certain aspects, the present invention provides prodrugs and derivatives of the compounds according to the formulae above. Prodrugs are derivatives of the compounds of the invention, which have metabolically cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention, which are pharmaceutical^ active, in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. -Preferred are the Q to C8 alkyl, C2-C8 alkenyl, aryl, C7-Cl2 substituted aryl, and C7-C,2 arylalkyl esters of the compounds of the invention.
PHARMACEUTICAL COMPOSITIONS
When employed as pharmaceuticals, the compounds of this invention are typically administered in the form of a pharmaceutical composition. Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
Generally, the compounds of this invention are administered in a pharmaceutically effective amount. The amount of the compound actually administered will typically be
-32-

determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound -administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
The pharmaceutical compositions of this invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. Depending on the intended route of delivery, the compounds of this invention are preferably formulated as either injectable or oral compositions or as salves, as lotions or as patches all for transdermal administration.
The compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing. The term '*unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such compositions, the furansulfonic acid compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like. Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art. As before, the active compound in such compositions is typically a minor component, often being from about 0.05 to 10% by weight with the remainder being the injectable carrier and the like.
Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight. When formulated as a ointment, the active ingredients will typically be combined with either a paraffmic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in-water cream base. Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the
-33-

formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.
The compounds of this invention can also be administered by a transdermal device. Accordingly, transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
The above-described components for orally administrable, injectable or topically administrable compositions are merely representative. Other materials as well as processing techniques and the like are set forth in Part 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
The compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The following formulation examples illustrate representative pharmaceutical compositions of this invention. The present invention, however, is not limited to the following pharmaceutical compositions.
Formulation 1 - Tablets
A compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 240-270 mg tablets (80-90 mg of active amide compound per tablet) in a tablet press.
Formulation 2 - Capsules
A compound of the invention is admixed as a dry powder with a starch diluent in an approximate 1:1 weight ratio. The mixture is filled into 250 mg capsules (125 mg of active amide compound per capsule).
Formulation 3 - Liquid
A compound of the invention (125 mg), sucrose (1.75 g) and xanthan gum (4 mg) are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose (11:89, 50 mg) in water. Sodium benzoate (10 mg), flavor, and color are diluted with water and added with stirring. Sufficient water is then added to produce a total volume of 5 mL.
Formulation 4 - Tablets
A compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 450-900 mg tablets (150-300 mg of active amide compound) in a tablet press.
Formulation 5 - Injection
A compound of the invention is dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of approximately 5 mg/ml.
-34-

Formulation 6 - Topical
Stearyl alcohol (250 g) and a white petrolatum (250 g) are melted at about 75°C and then a mixture of a compound of the invention (50 g) methylparaben (0.25 g), propylparaben (0.15 g), sodium lauryl sulfate (10 g), and propylene glycol (120 g) dissolved in water (about 370 g) is added and the resulting mixture is stirred until it congeals.
METHODS OF TREATMENT
The present compounds are used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to aberrant activity of the P2X7 receptor. Accordingly, the compounds and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating autoimmune, inflammatory and cardiovascular conditions in mammals including humans.
In a method of treatment aspect, this invention provides a method of treating a mammal susceptible to or afflicted with a condition associated with arthritis, uveitis, asthma, myocardial infarction, traumatic brain injury, acute spinal cord injury, inflammatory bowel disease and autoimmune disorders, which method comprises administering an effective amount of one or more of the pharmaceutical compositions just described.
In yet another method of treatment aspect, this invention provides a method of treating a mammal susceptible to or afflicted with a condition that gives rise to pain responses or that relates to imbalances in the maintenance of basal activity of sensory nerves. The present amines have use as analgesics for the treatment of pain of various geneses or etiology, for example acute, inflammatory pain (such as pain associated with osteoarthritis and rheumatoid arthritis); various neuropathic pain syndromes (such as post-herpetic neuralgia, trigeminal neuralgia, reflex sympathetic dystrophy, diabetic neuropathy, Guillian Barre syndrome, fibromyalgia, phantom limb pain, post-masectomy pain, peripheral neuropathy, HIV neuropathy, and chemotherapy-induced and other iatrogenic neuropathies); visceral pain, (such as that associated with gastroesophageal reflex disease, irritable bowel syndrome, inflammatory bowel disease, pancreatitis, and various gynecological and urological disorders), dental pain and headache (such as migraine, cluster headache and tension headache).
In additional method of treatment aspects, this invention provides methods of treating a mammal susceptible to or afflicted with neurodegenerative diseases and disorders such as, for example Parkinson's disease, multiple sclerosis; diseases and disorders which are mediated by or result in neuroinflammation such as, for example traumatic brain injury, and encephalitis; centrally-mediated neuropsychiatric diseases and disorders such as, for example depression mania, bipolar disease, anxiety, schizophrenia, eating disorders, sleep disorders and cognition disorders; epilepsy and seizure disorders; prostate, bladder and bowel dysfunction such as, for example urinary incontinence, urinary hesitancy, rectal hypersensitivity, fecal incontinence, benign prostatic hypertrophy and inflammatory bowel disease; respiratory and airway disease and disorders such as, for example, allergic rhinitis, asthma and reactive airway disease and chronic obstructive pulmonary disease; diseases and disorders which are mediated by or result in inflammation such as, for example rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus such as, for example psoriasis; obesity; lipid disorders; cancer; blood pressure; spinal cord injury; and
-35-

renal disorders method comprises administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions just described.
As a further aspect of the invention there is provided the present compounds for use as a pharmaceutical especially in the treatment or prevention of the aforementioned conditions and diseases. Also provided herein is the use of the present compounds in the manufacture of a medicament for the treatment or prevention of one of the aforementioned conditions and diseases.
Injection dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours. A preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels. The maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
For the prevention and/or treatment of long-term conditions, such as neurodegenerative and autoimmune conditions, the regimen for treatment usually stretches over many months or years so oral dosing is preferred for patient convenience and tolerance. With oral dosing, one to five and especially two to four and typically three oral doses per day are representative regimens. Using these dosing patterns, each dose provides from about 0.01 to about 20 mg/kg of the compound of the invention, with preferred doses each providing from about 0.1 to about 10 mg/kg and especially about 1 to about 5 mg/kg.
Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
When used to prevent the onset of a neurodegenerative, autoimmune or inflammatory condition, the compounds of this invention will be administered to a patient at risk for developing the condition, typically on the advice and under the supervision of a physician, at the dosage levels described above. Patients at risk for developing a particular condition generally include those that have a family history of the condition, or those who have been identified by genetic testing of screening to be particularly susceptible to developing the condition.
The compounds of this invention can be administered as the sole active agent or they can be administered in combination with other agents, including other compounds that demonstrate the same or a similar therapeutic activity, and that are determined to safe and efficacious for such combined administration.
GENERAL SYNTHETIC PROCEDURES
The bicycloheteroaryl compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
-36-

Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein.
The following representative methods are presented with details as to the preparation of representative bicycloheteroaryls that have been listed herein above. The compounds of the invention may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis.
REPRESENTATIVE METHODS
Method A (Compound 2002) 5,6,7,8-Tetrahydro-pyrido[3,4-dJpyrimidine-4-carboxyIic acid cyclohexylmethyl-amide

a. 7-Benzyl-N-(,cvclohexvlmethvl)-5,6,7,8-tetrahvdropvridor3,4-d1pyrimidine-4-carboxamide
Into a 50 ml round bottom flask was combined 7-benzyl-5,6,7,8-tetrahydro-pyrido[3,4-d]pyrimidine-4-carboxylic acid methyl ester (0.125g, 0.411mmol), C-cyclohexyl-methylamine (0.010 g, 0.88 mmol), and methanol (10ml). The mixture was heated at reflux for 24 hrs. The mixture was allowed to cool, reduced in vacuo, and purified by flash chromatography using a methylene chloride;methanol gradient (0-10%) . The combined fractions were reduced in vacuo to yield the title compound. (0.0177g, 10.9%). b. 5,6,7,8-Tetrahvdro-pvridor3.4-d1pvrimidine-4-carboxylic acid cvclohexylmethyl-amide
Into a 500 ml hydrogenation vessel was combined 7-benzyI- 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidine-4-carboxylic acid, cyclohexylmethyl-amide (80 mg, 0.22 mmol), palladium on carbon (30 mg) and methano. The vessel was purged and evacuated with hydrogen three times and allowed to agitate overnight at 15 psi. The contents were filtered over celite and concentrated to afford the title compound (4.7 mg, 7.7%.
-37-

Method B (Compound 2028) l~Oxo-l,2-dihydro-isoquinoIine-5-carboxyIic acid (adamantan-l-ylmethyl)-amide



o o
a. l-Oxo-l,2-dihydro-isoquinoline-5-carboxylic acid (adamantan-1-vlmethvlVamide
A mixture of l,2-dihydro-l-oxoisoquinoline-5-carboxylic acid (0.107 g, 0.000566 mol), 1-adamantanemethylamine (0.120 mL, 0.000679 mol), N-(3-dimethyIaminopropyl)-N'-ethylcarbodiimide hydrochloride (0.141 g, 0.000735 mol), 1-hydroxybenzotriazole (0.099 g, 0.00074 mol) and triethylamine (0.12 mL, 0.00085 mol) in N,N-dimethylformamide (2 mL, 0.02 mol) was stirred at room temperature for 18 hours. The mixture was poured onto water (100 ml) and extracted with ethyl acetate (3 X 100 ml). The combined extracts were dried over sodium sulfate and reduced in vacuo to afford the title compound.
Method D (Compound 2031) 2-(4-Fluoro-benzyI)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylic acid (1-hydroxy-cvclnhentvlmethvn-amirie





a. 2-(4-Muoropenzyl)-5-nitroisoquinonn-1(2H)-one
5-Nitro-isochrornen-l-one (1 g, 0.005 mol) and 4-fluoro-benzene methanamine, (2 g, 0.02 mol) were refluxed in methanol (20 mL, 0.5 mol) for 2 hours. The volatiles were removed via rotovapor, and the residue was purified via flash column chromatography (40 g of silica gel, 0-50% EtOAc/Hexane) gave a bright yellow solid. MS m/z(M+H) 299.1
b. 5-Amino-2-(4-iluorobenzvl)isoquinolin-1 (2H)-one
2-(4-Fluorobenzyl)-5-nitroisoquinolin-l(2H)-one (0.66 g, 0.0022 mol) and tin dichloride dihydrate (2 g, 0.009 mol) were stirred in tetrahydrofuran (20 mL, 0.2 mol) at room temperature overnight. The volatiles were removed via rotovapor, and the residue was dissolved in MeOH, filter through a pad of basic alumina and concentrated to dryness to give the title compound as a brown oil. MS m/z (M+H) 268.8.
c. 2-(4-Fluoro-benzvn-5-iodo-2H-isoqu inolin-1-one
-38-

5-Amino-2-(4-fluorobenzyI)isoquinolin-l(2H)-one (0.6- g, 0.002 mol) was added to a solution of sodium nitrite (0.5 g, 0.008 mol) in dimethyl sulfoxide (10 mL, 0.1 mol) at 35 °C. Aqueous hydrogen iodide (2 mL, 0.02 mol) in dimethyl sulfoxide (10 mL, 0.1 mol) was added. The reaction mixture was stirred at 35 °C for 1 hour. The cooled reaction mixture was neutralized with saturated aq. Na2C03 and extracted with methylene chloride (3 x 20 mL). The combined methylene chloride extracts were washed with brine and dried over magnesium sulfate. The solvent was removed in vacuo and the residue was chromatographed on 12g of silica gel column (0-50% EtOAc/Hexane) gave the desired product as a brown solid. MS m/z (M+H) 379.7.
d. 2-(4-Fluoro-benzyl')-1-oxo-l.2-dihvdro-isoquinoline-5-carboxvlic acid (1-hydroxy-cycloheptvlmethvn-amide
A 5-mL process vial was charged with 2-(4-fluoro-benzyl)-5-iodo-2H-isoqu inolin-1-one (100 mg, 0.0004 mol), l-(aminomethyl)cycloheptanol (200 mg, 0.001 mol), molybdenum hexacarbonyl (90 mg, 0.0004 mol), p alladium acetate (8 mg, 0.00004 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.001 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 50-100% EtOAC/Hexane) gave the desired product as a white solid.
'H NMR (CDC13) 5: 8.55 (d, J = 8.5 Hz, 1H), 7.76 (d, J = 7.6 Hz, 1H), 7.48 (t, J = 7.8 Hz, 1H), 7.33-7.29 (m, 2H), 7.14 (d, J = 7.7 Hz, 1H), 7.04-6.99 (m, 3H), 6.38 (br, 1H), 5.17(s, 2H), 3.49 (d, J = 5.9 Hz, 1H), 1.78-1.45 (m, 12H). MS m/z (M+H) 423.5.
Method £ (Compound 2033) 2-(2,3-Dihydro-benzo[l,4]dioxin-6-ylmethyl)-l-oxo-l,2-dihydro-isoquinoline-5-

a.2-(2,3-Dihvdro-benzo[1,4]dioxin-6-vlmethvn-5-nitro-2H-isoquinolin-l-one
5-Nitro-isochromen-l-one (1.0 g, 0.0052 mol) and C-(2,3-Dihydro-benzo[l,4]dioxin-6-y
l)-methylamine (1.0 g, 0.0060 mol) were refluxed in methanol (40 mL, 1 mol) for 2
hours. The solvent was removed and the residue was purified via flash chromatography
(40 g of silica gel, 0-30% EtOAc/Hexanes) gave a yellow solid.
MS m/z (M+H) 339.1.
b. 5-Amino-2-(2,3-dihydro-benzo[l,4]di oxin-6-ylmethyl)-2H-isoquinolin-l-o ne
-39-

2-(2,3-Dihydro-benzo[l,4]dioxin-6-y lmethyl)-5-nitro-2H-isoquinolin-l-one (0.9 g, 0.002 mol), tin dichloride dihydrate (2 g, 0.009 mol) were stirred in tetrahydrofuran (10 mL, 0.1 mol) at room temperature for 20 hours. The volatiles were removed and the residue was purified via flash column chromatography (40 g of silica gel, 50% EtOAc/Hexanes) gave a red oil. MS m/z (M+H) 309.2.
c. 2-((2,3-dihydrobenzo[l ,4]dioxin-6-yl)methyl)-5-iodoisoquinolin-1 (2H)-one
5-Amino-2-(2,3-dihydro-benzo[l,4]di oxin-6-ylmethyl)-2H-isoquinolin-l-o ne (260 mg, 0.00084 mol) was added to a solutionof sodium nitrite (200 mg, 0.003 mol) in dimethyl sulfoxide (4 mL, 0.06 mol) at 35 °C. Aqueous hydrogen iodide (0.5 mL, 0.004 mol) in dimethyl sulfoxide (4 mL, 0.06 mol) was added, and the reaction mixture was stirred for 1 hour. The cooled reaction mixture was neutralized with sat. aq. NaHC03 and extracted with methylene chloride (3x 50 mL). The combined methylene chloride extractes were washed with brine, dried over magnesium sulfate. The solvent was removed in vacuo and the residue was purified via flash column chromatography (12 g of silica gel, 0-50% EtOAc/Hexane) gave a light yellow solid. MS m/z (M+H) 420.0.
d. 2-((2.3-DihvdrobenzorL41dioxin-6-vl)methyl)-N-((l-hvdroxvcvcloheptvl')methvl')-l-
oxo-1.2-dihvdroisoquinoline-5-carboxarnide
A 5 mL process vial was charged with 2-((2,3-dihydrobenzo[l,4]dioxin-6-yl)methyl)-5-iodoisoquinolin-l(2H)~one (100 mg, 0.0002 mol) l-(aminomethyl) cycloheptanol (100 mg, 0.0007 mol), molybdenum hexacarbonyl (60 mg, 0.0002 mol), palladium acetate (5 mg, 0.00002 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (100 mg, 0.0007 mol) and 1,4-dioxane (1 mL, 0.01 mol). The vessel was sealed under air and exposed to microwave heating at 110 °C for 15 min. After cooling to RT, and the mixture was concentrated and the residue was dissolved in a small amount of CH2C12 and purified via flash column chromatography (12 g of silica gel, 0-100% EtOAc/Hexane) to give the desired product as a white solid.
'H NMR (CDC13) 5: 8.43 (d, J = 8.1 Hz, 1H), 7.60 (dd, J = 1.2, 7.3 Hz, 1H), 7.35 (t, J = 7.9 Hz, 1H), 7.13 (d, J = 7.7 Hz, 1H), 6.9 (d, J = 7.7 Hz, 1H), 6.85-6.79 (m, 3H), 6.5 (br, 1H), 5.08 (s, 2H), 4.22 (s, 4H), 3.50 (d, J = 5.92 Hz, 2H), 2.63 (s, 1H), 1.77-1.50 (m, 12H). MS m/z (M+H) 463.5

a. 2-( 1 -Hvdroxy-3-methvlbutan-2-vl)-N-(( 1 -hydroxvcycloheptyDmethyD-1 -oxo-12-dihvdroisoqufnoline-5-carboxamide
-40-
Method F (Compound 2035) 2-(l-Hydroxymethyl-2-methyI-propyl)-l -oxo-1,2-dihydro-isoquinoline-S-carboxylic acid(l-hydroxy-cycIoheptyImethyl)-amide

A 5-mL process vial was charged with 2-(l-hydroxy-3-methylbutan-2-yl)-5-iodoisoquinolin-l(2H)-one (100 mg, 0.0004 mol), l-(aminomethyl)cycloheptanol (200 mg, 0.001 mol), molybdenum hexacarbonyl (90 mg, 0.0004 mol), palladium acetate (8 mg, 0.00004 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.001 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 50-100% etOAC/Hexane) to give the desired product as a white solid. 'H NMR (CDC13) 6: 8.53 (d, J = 8.4 Hz, 1H), 7.78 (dd, J = 1.2, 7.3 Hz, 1H), 7.49 (t, J = 11.4 Hz, 1H), 7.20 (d, J = 7.8 Hz, 1H), 7.07 (d, J - 7.8 Hz, 1H), 6.41 (br, 1H), 4.43 (br, 1H), 4.09-3.99 (m, 1H), 3.51 (d, J = 5.8 Hz, 1H), 2.43 (br, 1H), 1.79-1.49 (m, 14H), 1.15 (d, J - 6.6 Hz, 3H), 0.81 (d, J = 6.6 Hz, 3H). MS m/z(M+H) 401.0
Method G (Compound 2039) 2-(2,3-Dihydroxy-propyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylicacid cycloheptylmethyl-amide

a. 2-((2.2-dimethyl-1.3-dioxolan-4-vl')methyl')-5-nitroisoquinolin-1(2H)-one
5-Nitro-isochromen-l-one (8 g, 0.04 mol) and (2,2-dimethyl-l,3-dioxolan-4-yl)methanamine (5 g, 0.04 mol) were refluxed in methanol (40 mL, 1 mol) for 2 hours. The volatiles were removed via rotovapor, and the residue was purified vai flash column chromatography (330 g of sliica gel, 0-50% EtOAc/Hexane) to give a bright yellow solids. MSm/z(M+H) 305.1.
b. 5-Amino-2-((2.2-dimethvl-1.3-dioxolan-4-vl')methyl)isoquinolin-l (2H)-one
2-((2,2-DimethyI-l,3-dioxolan-4-yl)methyl)-5-nirroisoquinolin-l(2H)-one (7.3 g, 0.024 mol) was stirred with palladium 10% wt. on calcium carbonate (1 g, 0.005 mol) in methanol (100 mL, 2 mol) under hydrogen (balloon) over 1 h at room temperature. The catalyst was filtered, the filtrate was concentrated to dryness, purified via flash chromatography (120 g of silica gel, 0-10% MeOH/CH2C12) to give a white solid. MS m/z (M+H) 276.2.
c. 2-((2,2-Dimethvl-1.3-dioxolan-4-vl)methylV5-iodoisoquinolin-1 (2HVone
-41 -

5-Amino-2-((2,2-dimethyl-l,3-dioxolan-4-yl)methyl)isoquinolin-l(2H)-one (4.2 g, 0.015 mol) was added to a solution of sodium nitrite (4 g, 0.06 mol) in dimethyl sulfoxide (80 mL, 1 mol) at 35 °C. Aqueous hydrogen iodide (8 mL, 0.06 mol) in dimethyl sulfoxide (80 mL, 1 mol) was added, and the reaction mixture was stirred for 1 hour. The cooled reaction mixture was neutralized with sat. aq. NaHC03 and extracted with methylene chloride (3x 50 mL). The combined methylene chloride extracts were washed with brine, dried over magnesium sulfate. The solvent was removed in vacuo and the residue was purified via flash column chromatography (40 g of silica gel, 0-50% EtOAc/Hexane) to give a light yellow solid. MS m/z (M+H) 385.6
d. N-(cycloheptvlmethvl)-2-((2,2-dimethv]-1,3-dioxolan-4-vl)methyl)-l-oxo-1,2-
dihydroisoquinoline-5-carboxamide
A 5 mL process vial was charged with 2-((2,2-dimethyl-l,3-dioxolan-4-yl)methyl)-5-iodoisoquinolin-l(2H)-one (200 mg, 0.0005 mol) cycloheptylmethanamine (200 mg, 0.002 mol), molybdenum hexacarbonyl (100 mg, 0.0005 mol), palladium acetate (10 mg, 0.00005 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.002 mol) and 1,4-dioxane (3 mL, 0.04 mol). The vessel was sealed under air and exposed to microwave heating at 110 °C for 15 min. After cooling to RT the mixture was concentrated, dissolved in a small amount of CH2C12 and purified via flash column chromatography (12 g of silica gel, 0-50% EtOAc/Hexane) to give the desired product as a white solid. MS m/z (M+H) 413.1.
e. N-(cvcloheptvlmethvl)-2-(2,3-dihydroxvpropyl)-1-oxo-l.2-dihvdroisoquinoline-5-
carboxamide
N-(Cycloheptylmethyl)-2-((2,2-dimethyl-l,3-dioxolan-4-yl)methyl)-l-oxo-l,2-dihydroisoquinoline-5-carboxamide (110 mg, 0.00028 mol), hydrogen chloride (5 mL, 0.006 mol) as 2M solution in ether and methylene chloride (5 mL, 0.08 mol) were stirred at room temperature for 2 hours. The volatiles were removed under vacuum, the residue was purified via flash chromatography (12 g of silica gel, 0-10% MeOH/CH2C12) to give a white solid.
'H NMR (CDC13) 5: 8.54 (d, J = 8.1 Hz, IH), 7.77 (dd, J = 1.3, 7.3Hz, IH), 7.51 (t, J = 7.9 Hz, IH), 7.20 (d, J = 7.6 Hz, IH), 7.09 (d, J = 7.6 Hz, IH), 5.98 (br, IH), 4.21 (dd, J = 5.5, 17.8Hz, 2H), 4.05-4.03 (m, IH), 3.55 (dd, J = 1.6, 4.8Hz, 2H), 3.36 (t, J = 6.3 Hz, 2H), 1.83-1.27 (m, 13H). MS m/z (M+H) 373.1
Method H
(Compound 2041) 2-(2-Hydroxy-ethyl)-l-oxo-l,2-dihydro-isoquinoIine-5-carboxylic acid cycloheptylmethyl-amide
-42-


a. 2-(2-hvdroxyethyl)-5-nitroisoquinolin-1 (2H)-one
5-Nitro-isochromen-l-one (3.60 g, 0.0170 mol) was suspended in MeOH (40 mL), ethanolamine (3.11 g, 0.0508 mol) was added and the reaction mixture was stirred at 70 °C for 2 h under an atmosphere of nitrogen. Cooled to room temperature and Et3N (5 mL) was added and the reaction mixture was stirred at room temperature for 2 days. Solid thus formed was filtered out (yellow solid was obtained as the desired product, 0.9 g). Filtrate was concentrated and the residue was dissolved in EtOAc, washed by water and brine, dried over Na2S04, solvent was removed, to give the product as a yellow solid (1.3 g).
b. 5-Amino-2-(2-hydroxvethvnisoquinolin-l(2H')-one
Into a 500 ml round bottom flask was combined 2-(2-hydroxyethyl)-5-nitroisoquinolin-l(2H)-one (2.0 g, 0.0085 mol) palladium on C (0.09 g, 0.0008 mol), and methanol (100 mL, 2 mol). The vessel was charged with hydrogenand evacuated three times and stirred under hydrogen at 1 atm. overnight. The mixture was filtered over Celite and the filtrate was removed under reduced pressure to yield the title compound as a light brown solid. It was taken onto the next step without further purification.
c. 2-(2-hydroxyethyl)-5-iodoisoquinolin-1 (2H)-one
5-Amino-2-(2-hydroxyethyl)isoquinolin-l(2H)-one (1.62 g, 0.00793 mol) was added to a solution of sodium nitrite (2 g, 0.03 mol) in dimethyl sulfoxide (40 mL, 0.5 mol) at 35 °C. Aqueous hydrogen iodide (4 mL, 0.03 mol) in dimethyl sulfoxide (40 mL, 0.5 mol) was added, and the reaction mixture was stirred for 1 hour. The cooled reaction mixture was neutralized with sat. aq. NaHC03 and extracted with methylene chloride (3x 50 mL). The combined methylene chloride extracts were washed with brine, dried over sodium sulfate and reduced in vacuo. The mixture was purified by column chromatography using an ethyl acetate:hexanes ( 0-100) gradient. The combined pure fractions were reduced in vacuo to yield the title compound as an off white solid.
d. N-CCvcloheptvlmethvD-1,2-dihvdro2-(2-hydroxyethyD-l-oxoisoquinoline-5-
carboxamide
Into a 5 ml microwave reaction vessel was combined 2-(2-hydroxyethyl)-5-iodoisoquinolin-l(2H)-one (0.100 g, 0.000317 mol), cycloheptylmethanamine (100 mg, 0.0008 mol), molybdenum hexacarbonyl (80 mg, 0.0003 mol), palladium acetate (7 mg, 0.00003 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (0.1 mL, 0.001 mol) and 1,4-dioxane (1 mL, 0.02 mol). The vessel was exposed to microwave heating for 15 min at 110 °C. The reaction tube was cooled to room temperature and the volatiles removed under
-43-

educed pressure. The mixture was purified directly by column chromatography using an methanol:methylene chloride ( 0-10%) gradient. The combined pure fractions were reduced in vacuo to yield a white solid which was purified again by HPLC to afford the title compound as a white solid. ]H NMR (DMSO-d6) 8: 8.55 (t, 1H), 8.31 (d,lH), 7.72 (dd, 1H), 7.517 (t, 1H), 7,446 (d, 1H), 6.76 (d, 1H), 4.88 ( t, 1H) 4.014 (t 2H), 3.66 (q, 2H) 3.123 (t,2H), 1.79-1.34 (m, 11H), 1.2615-1.149 (m, 2H)
Method J (Compound 2048) 2-(2-Hydroxy-ethyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxyIic acid (1-hydroxy-cycloheptylmethyl)-amide

a. 1.2-Dihydro-N-(( 1 -hvdroxvcvcloheptvnmethyl)-2-(2-hydroxvethvl)-1 -oxoisoqumoline-5-carboxarnide
Into a 5 ml microwave reaction vial was combined 2-(2-hydroxyethyl)-5-iodoisoquinolin-l(2H)-one (200 mg, 0.000635 mol), l-(aminomethyl)cycloheptanol (273 mg, 0.00190 mol) molybdenum hexacarbonyl (168 mg, 0.000635 mol) palladium acetate (10 mg, 0.00006 mol) l,8-diazabicyclo[5.4.0]undec-7-ene (0.285 mL, 0.00190 mol) and 1,4-dioxane (3 mL, 0.03 mol). The mixture was subjected to microwave heating at 80 °C for 10 minutes. Methanol (1.5 ml) was added and the reaction mixture was filtered through a syringe filter and the filtrate concentrated. The mixture was purified by prep HPLC using a Phenomenex C18 Axia packed xolumn at PH 12. The combined pure fractions were reduced in vacuo to yield the title compound as a white solid. LC/MS M+H= 359.3. 1H NMR (DMSO-d6) 5: 8.36-8.26 (m, 2H), 7.81-7.78 (dd, 1H) 7.52 (t, 1H), 7.48 (d, 1H) 6.813 (d, 1H), 4.892 (t, 1H), 4.325 (t, 1H), 4.016 (t, 1H), 3.66 (t,lH), 3.285 (d, 1H), 1.69-1.44 (m, 10H), 1.414-1.317 (m, 2H)
Method K
(Compound 2053) 2-(2-Hydroxy-l-hydroxymethyl-ethyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylic acid4-chloro-3-trifluoromethyl-benzylamide
-44-


a. 2-(5-Nitro-l-oxoisoquinolin-2(1H)-ylpropane-1.3-divl diacetate
5-Nitro-isochromen-l-one (4.2 g, 0.022 mol) and serinol (2.0 g, 0.022 mol) were refluxed in methanol (40 mL, 1 mol) for 1 hour. TLC showed all the starting material consumed, triethylamine (20 mL, 0.1 mol) was added to the mixture and the reaction mixture was refluxed overnight. The volatiles were removed via rotovapor, and the residue was diluted with methylene chloride (100 mL, 2 mol). Acetic anhydride (9 g, 0.09 mol) and 4-dimethylaminopyridine (30 mg, 0.0002 mol) were then added and the mixture was stirred at room temperature overnight. The volatiles were removed and the residue was purified via flash column chromatography (120 g of siical gel, 0-50% EtOAc/Hexane) to give a yellow oil. MS m/z (M+H) 349.1.
b. 2-(5-amino-1 -oxoisoquinolin-2( 1 HVvllpropane-1.3-divl diacetate
2-(5-Nitro-l-oxoisoquinolin-2(lH)-yl)propane-l,3-diyl diacetate (6.3 g, 0.018 mol) was stirred with palladium 10% wt. on calcium carbonate (0.6 g, 0.003 mol) in ethanol (100 mL, 2 mol) under hydrogen (balloon) over lh at room temperature. The catalyst was filtered, the filtrate was concentrated to dryness to give a yellow oil. MS m/z (M+H) 319.2
c. 2-(5-Iodo-l-oxoisoquinolin-2nH)-yl)propane-1.3-divl diacetate
2-(5-Amino-l-oxoisoquinolin-2(lH)-yl)propane-l,3-diyl diacetate (4.2 g, 0.013 mol) was added to a solution of sodium nitrite (4 g, 0.05 mol) in dimethyl sulfoxide (70 mL, 1 mol) at 35 °C. Aqueous hydrogen iodide (7 mL, 0.05 mol) in dimethyl sulfoxide (70 mL, 1 mol) was added, the mixture was stirred at 35 °C for 45 minutes. The cooled mixture was neutralized with sat. aq. NaHC03, extracted with CH2C12 (50 mL x 3), washed with brine, and dried over MgS04. Filtered, evaporated and purified via flash column chromatography (12 g of silica gel, 50-100% EtOAc/Hexane) to give the desired product as a yellow oil. MS m/z (M+H) 430.1.
d. 2-(5-(4-Chloro-3-(trifluoromethvl)benzvlcarbamovn-l-oxoisoquinolin-2nH')-
vDpropane-1.3-divl diacetate
A 5-mL process vial was charged with 2-(5-iodo-l-oxoisoquinolin-2(lH)-yl)propane-1,3-diyl diacetate (200 mg, 0.0004 mol), (4-chloro-3-(trifluoromethyI)phenyl) methanamine (200 mg, 0.001 mol), molybdenum hexacarbonyl (90 mg, 0.0004 mol), palladium acetate (8 mg, 0.00004 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.001 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter
-45-

cooled to room temperature, and the mixture was concentrated and dissolved in a small
volume of dichloromethane. The crude product was purified via flash column
chromatography (12 g of silica gel, 50-100% EtOAc/Hexane) to give the desired product
as a yellow oil. MS m/z (M+H) 538.9
e. N-(4-chloro-3-(triftuoromethvl)benzyl)-2-( 1,3-dihvdroxvpropan-2-vl)-1 -oxo-1,2-
dihydroisoquinoline-5-carboxamide
2-(5-(4-Chloro-3-(trifluoromethyl)benzylcarbamoyl)-1 -oxoisoquinolin-2( 1 H)-yl)propane-1,3-diyI diacetate (100 mg, 0.0002 mol), potassium carbonate (80 mg, 0.0006 mol) and methanol (2 mL, 0.05 mol) were stirred at room temperature for 2 hours. The mixture was filtered and purified via flash chromatography (12 g fo silica gel, 0-10% MeOH/CH2C12) to give a white solid. 'H NMR (DMSO-d6) 6: 9.18 (br,lH), 8.35 (d, J = 7.6 Hz, IH), 7.86-7.82 (m, 2H), 7.75-7.70 (m, 2H), 7.55-7.27 (m, 2H), 6.79 (d, J = 7.9 Hz, IH), 4.95-4.89 (m, 3H), 4.56 (d, J = 5.9 Hz, 2H), 3.75-3.71 (m, 4H), MS m/z (M+H) 455.2.

Method L (Compound 2058) 2-((R)-l-Carbamoyl-ethyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylic acid 4-chloro-3-trifluoromethyl-benzyIamide

a. (R)-2-( 1 -Amino-1 -oxopropan-2-vl)-N-(4-chloro-3-(trifluoromethyl)benzylV 1 -oxo-1.2-dihvdroisoquinoline-5-carboxamide
A 5 mL process vial was charged with (R)-2-(5-iodo-l-oxoisoquinolin-2(lH)-yl)propanamide (100 mg, 0.0003 mol) (4-chloro-3-(trifluoromethyl)phenyl)methanamine (100 mg, 0.0006 mol), molybdenum hexacarbonyl (80 mg, 0.0003 mol), palladium acetate (6 mg, 0.00003 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (100 mg, 0.0009 mol) and 1,4-dioxane (0.7 mL, 0.009 mol). The vessel was sealed under air and exposed to microwave heating at 110 °C for 15 min. After cooling to RT the mixture was concentrated, dissolved in a small amount of CH2C12, and purified via flash column chromatography (12 g of silica gel, 0-100% EtOAc/Hexane) to give the desired product as a white solid.
'H NMR (DMSO-d6) 8: 7.54 (d, J = 8.0 Hz, 1H), 6.97-6.93 (m, 2H), 6.87-6.64 (m, 4H), 6.57 (d, J = 7.9 Hz, 1H), 6.09 (d, J = 7.8 Hz, 1H), 4.66 (q, J = 7.3 Hz, 1H), 3.75 (s, 2H), 2.68-2.60 (m, 2H), 0.78 (d, J - 7.3 Hz, 3H). MS m/z (M+H) 452.0.
Method M (Compound 2059) 2-((S)-l-Carbamoyl-ethyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylic acid 4-chIoro-3-trifIuoromethyl-benzyIamide
-46-


a. (S)-2-( 1 -Amino-1 -oxopropan-2-vlVN-(4-chloro-3-(trifluoromethvl)benzvl')-1 -oxo-1,2-di hydroisoquinoline-5-carboxamide
A 5-mL process vial was charged with (S)-2-(5-iodo-l-oxoisoquinolin-2(lH)-
y])propanamide (100 mg, 0.0004 mol), (4-chloro-3-
(trifluoromethyl)phenyl)methanamine (200 mg, 0.001 mol), molybdenum hexacarbonyl (90 mg, 0.0004 mol), palladium acetate (8 mg, 0.00004 mol), 1,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.001 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 50-100% EtOAC/Hexane) to give the desired product as a white solid.
'H NMR (DMSO-d6) 8: 7.54 (d, J = 8.0 Hz, 1H), 6.97-6.93 (m, 2H), 6.87-6.64 (m, 4H), 6.57 (d, J = 7.9 Hz, 1H), 6.09 (d, J = 7.8 Hz, 1H), 4.66 (q, J = 7.3 Hz, 1H), 3.75 (s, 2H), 2.68-2.60 (m, 2H), 0.78 (d, J = 7.3 Hz, 3H). MS m/z (M+H) 452.3.
Method N (Compound 2064) 2-((R)-l-CarbamoyI-ethyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylicacid (adamantan-1 -ylmethyl)-amide

a. (R)-2-(5-Iodo-1 -oxoisoquinolin-2( 1 H)-yl)propanamide
(R)-2-(5-Amino-l-oxoisoquinolin-2(lH)-yl)propanamide (770 mg, 0.0033 mol; ) was added to a solution of sodium nitrite (900 mg, 0.01 mol;) in dimethyl sulfoxide (50 mL, 0.7 mol;) at 35 °C. Aqueous hydrogen iodide (4 mL, 0.03 mol) in dimethyl sulfoxide (50 mL, 0.7 mol; ) was added. The reaction mixture was stirred at 35 °C for 1 hour. The cooled reaction mixture was neutralized with saturated aq. Na2C03 and extracted with methylene chloride (3 x 20 mL). The combined methylene chloride extracts were washed
-47-

with brine and dried over magnesium sulfate. The solvent was removed in vacuo and the residue was chromatographed on 12g of silica gel column (0-50% EtOAc/Hexane) to give the desired product as a brown oil. MS m/z (M+H) 343.1
b. 2-((R)-1 -Carbamoyl-ethy)-1-oxo-12 -dihvdro-isoq uinoline-5-carboxylic acid (adamantan-1 -vlmethy)-amide
A 5-mL process vial was charged with (R)-2-(5-iodo-l-oxoisoquinolin-2(lH)-yl)propanamide (250 mg, 0.00073 mol; ) , 1-adamantanemethylamine (400 mg, 0.002 mol; ) , molybdenum hexacarbonyl (200 mg, 0.0007 mol; ), palladium acetate (20 mg, 0.00007 mol; ), l,8-diazabicyclo[5.4.0]undec-7-ene (300 mg, 0.002 mol; ) and 1,4-dioxane (3 mL, 0.04 mol; ). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 50-100% etOAC/Hexane) to give the desired product as a white solid. 1H NMR (CDC13) 5: 8.51 (d, J = 7.8 Hz, 1H), 7.78 (dd, J = 1.3, 7.3 Hz, 1H), 7.51 (t, 8.0 Hz, 1H), 7.28 (d, J = 7.8 Hz,lH), 7.10 (d, J = 7.8 Hz, 1H), 6.3 (br, 1H), 5.93 (br, 1H), 5.73 (q, J = 7.2 Hz, 1H), 5.33 (br, 1H), 3.26-3.16 (m, 2H), 2.09-2.02 (m, 3H), 1.76-1.57 (m, 15H). MS m/z (M+H) 408.0

Method O (Compound 2065) 2-((S)-l-Carbamoyl-ethyl)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylic acid (adamantan-l-ylmethyl)-amide

a. (S)-2-(5-lodo-1 -oxoisoqumoun-2(1h)-ylpropanamide
(S)-2-(5-Amino-l-oxoisoquinolin-2(lH)-yl)propanamide (320 mg, 0.0014 mol; ) was added to a solution of sodium nitrite (400 mg, 0.006 mol;) in dimethyl sulfoxide (20 mL, 0.3 mol;) at 35 °C. Aqueous hydrogen iodide (2 mL, 0.01 mol) in dimethyl sulfoxide (20 mL, 0.3 mol; ) was added. The reaction mixture was stirred at 35 °C for 1 hour. The cooled reaction mixture was neutralized with saturated aq. Na2C03 and extracted with methylene chloride (3 x 20 mL). th ecombined methylene chloride extracts were washed with brine and dried over magnesium sulfate. The solvent was removed in vacuo and the
-48-

residue was chromatographed on 25g of silica gel column (0-50% EtOAc/Hexane) to give the desired product as a yellow solid. MS m/z(M+H) 343.1
b. 2-((S)-1 -Carbamoyl-ethvP-1 -oxo-1.2 -dihvdro-isoq uinoKne-5-carboxvlic acid (adamantan-1 -ylmethy)-amide
A 5-mL process vial was charged with (S)-2-(5-iodo-l-oxoisoquinolin-2(lH)-yl)propanamide (90 mg, 0.0003 mol;), 1-adamantanemethylamine (90 mg, 0.0005 mol;) , molybdenum hexacarbonyl (90 mg, 0.0004 mol; ), palladium acetate (8 mg, 0.00004 mol;), l,8-diazabicyclo[5.4.0]undec-7-ene (80 mg, 0.0005 mol;) and 1,4-dioxane (2 mL, 0.02 mol; ). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 50-100% EtOAC/Hexane) then Prep. HPLC to give the desired product as a white solid. 'H NMR (CDC13) 8: 8.51 (d, J = 7.8 Hz, 1H), 7.78 (dd, J = 1.3, 7.3 Hz, 1H), 7.51 (t, 8.0 Hz, 1H), 7.28 (d, J = 7.8 Hz,lH), 7.10 (d, J = 7.8 Hz, 1H), 6.3 (br, 1H), 5.93 (br, 1H), 5.73 (q, J = 7.2 Hz, 1H), 5.33 (br, 1H), 3.26-3.16 (m, 2H), 2.09-2.02 (m, 3H), 1.76-1.57 (m, 15H). MS m/z(M+H) 408.1
Method P (Compound 2069) 2-((R)-2-Hydroxy-l-methyl-ethyI)-l-oxo-l,2-dihydro-isoquinoline-5-carboxylic acid cycloheptylmethyl-amide

a. Acetic acid (RV2-(5-iodo-l-oxo-lH- isoquinolin-2-yl)-propvi ester
Acetic acid (R)-2-(5-amino-l-oxo-lH -isoquinolin-2-yl)-propyl ester (790 mg, 0.0030 mol) was added to a solution of sodium nitrite (800 mg, 0.01 mol) in dimethyl sulfoxide (10 mL, 0.1 mol) at room temperature. Aqueous hydrogen iodide (4 mL, 0.03 mol) in dimethyl sulfoxide (10 mL, 0.1 mol) was added. The reaction mixture was stirred at room temperature for 1 hour. The cooled reaction mixture was neutralized with saturated aq. Na2C03 and extracted with methylene chloride (3 x 100 mL). The combined methylene chloride extracts were washed with brine and dried over magnesium sulfate. The solvent
-49-

was removed in vacuo and the residue was chromatographed on 40 of silica gel column (0-50% EtOAc/Hexane) to give the desired product as a brown oil. MS m/z (M+H) 371.9
b. (R)-2-(5-(cvcloheptvlmethvIcarbamoyl)-1 -oxoisoquinolin-2( 1 HVy)propyl acetate
A 5 mL process vial was charged with Acetic acid (R)-2-(5-iodo-l-oxo-lH- isoquinolin-2-yl)-propyl ester (200 mg, 0.0005 mol) cycloheptylmethanamine (100 mg, 0.001 mol), molybdenum hexacarbonyl (100 mg, 0.0005 mol), palladium acetate (10 mg, 0.00005 mol), l,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.002 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating at 110 °C for 15 min. After cooling to RT the mixture was concentrated, dissolved in a small amount of CH2C12, purified via flash column chromatography (12 g of silica gel, 0-50% EtOAc/Hexane) to give the desired product as a white solid. MS m/z (M+H) 399.1.
c. (R)-N-(CvcIoheptvlmethyD-2-( 1 -hvdroxypropan-2-vD-1 -oxo-1.2-dihvdroisoquinoline-
5-carboxamide
(R)-2-(5-(CycloheptyImethylcarbamoyl)-1 -oxoisoquinolin-2( 1 H)-yl)propyl acetate (170 mg, 0.00043 mol) and Potassium carbonate (100 mg, 0.0008 mol) were stirred in methanol (5 mL, 0.1 mol) at room temperature overnight. The volatiles were removed under vacuo and the residue was purified via flash chromatography (12 g of silica gel, 0-10% MeOH/CH2Cl2) to give a white solid.
'H NMR (CDC13) 5: 8.46 (d, J = 8.1 Hz, 1H), 7.70 (dd, J = 1.2, 7.3 Hz, 1H), 7.43 (t, J = 8.0 Hz, 1H), 7.22 (d, J = 7.84, 1H), 7.02 (d, J = 7.8 Hz, 1H), 6.09 (br, 1H), 5.13-5.12 (m, 1H), 3.95-3.82 (m, 2H), 3.35 (t, J = 6.3 Hz, 2H), 1.83-1.27 (m, 16H). MS m/z (M+H) 355.1.
Method Q (Compound 2070) 2-CyclopropyI-l-oxo-l,2-dihydro-isoquinoline-5-carboxyIic acid cycloheptylmethyl-amide

a. 2-Cvclopropvl-5-nitro-2H-isoquinolin-1 -one
5-Nitro-isochromen-l-one (5 g, 0.03 mol), cyclopropylamine (2 g, 0.04 mol) were refluxed in methanol (50 mL, 1 mol) for 2 hours and then room temperature stirring overnight. The resulting yellow solid was collected via filtration. The volatiles were removed under vacuum, the residue was purified via flash chromatography (120 g of silica gel, 0-20% EtOAc/Hexane) to give a yellow solid. MS m/z (M+H) 231.3.
b. 5-Amino-2-cvclopropvlisoquinolin-1 (2H)-one
-50-

To a suspension of 2-cyclopropyl-5-nitro-2H-isoquinolin-l-one (3.7 g, 0.015 mol) in Ethanol (80 mL, 1 mol) was added ammonium chloride (8 g, 0.2 mol) in Water (80 mL, 4 mol) and the reaction heated at 85 °C and then iron (4 g, 0.06 mol) was added. The reaction started turning dark and became completely brown.The reaction was heated for lh. LC/MS showed no starting material left and only one peak which to give the desired MW. The reaction was removed from the oil bath and 150 ml of methylene chloride was added in the flask. The mixture layers were separated and aqueous layer was extracted with CH2C12 (100 mL x3).The combined organic layers were washed once with brine. The organic layer was collected, dried over Na2S04 and reduced in vacuo to produce a yellow-orange solid.
1H-NMR (400MHz, DMSO-d) 8 7.41 (d, J= 7.95 Hz, 1H), 7.19-7.13 (m, 2H), 6.84 (d, 7.83 Hz, 1H), 6.69 (d, J= 7.78 Hz, 1H), 5.64 (s, 2H), 3.34-3.28 (m, 1H), 1.00-0.95 (m, 2H), 0.84-0.80 (m, 2H). MS m/z (M+H) 201.3.
c. 2-Cvclopropyl-5-iodoisoquinolin-1 (2H)-one
5-Amino-2-cyclopropylisoquinolin-l(2H)-one (2.0 g, 0.0095 mol) was added to a solution of sodium nitrite (3 g, 0.04 mol) in dimethyl sulfoxide (100 mL, 2 mol) at room temperature. Aqueous hydrogen iodide (10 mL, 0.08 mol) in dimethyl sulfoxide (100 mL, 2 mol) was added. The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was neutralized with saturated aq. Na2C03 and extracted with methylene chloride (3 x 200 mL). The combined methylene chloride extracts were washed with brine and dried over magnesium sulfate. The solvent was removed in vacuo and the residue was chromatographed on 120 g of silica gel column (0-25% EtOAc/Hexane) to give the desired product as a yellow solid. MS m/z (M+H) 312.2.
d. N-(Cvcloheptvlmethyn-2-cyclopropvl-l-oxo-l,2-dihvdroisoquinoline-5-carboxamide
A 5-mL process vial was charged with 2-cyclopropyl-5-iodoisoquinolin-l(2H)-one (100 mg, 0.0004 mol), cycloheptylmethanamine (100 mg, 0.001 mol), molybdenum hexacarbonyl (90 mg, 0.0004 mol), palladium acetate (8 mg, 0.00004 mol), 1,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.001 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 30% EtOAC/Hexane) to give the desired product as a white solid.
!H NMR (CDC13) 5: 8.48 (d, J = 8.0 Hz, 1H), 7.69 (dd, J = 1.1, 7.3 Hz, 1H), 7.43 (t, J = 7.9 Hz, 1H), 7.16 (d, J = 7.8 Hz, 1H), 6.93 (d, J = 7.8 Hz, 1H), 6.02 (br, 1H), 3.37-3.32 (m, 3H), 1.83-0.94 (m, 17H). MS m/z (M+H) 339.3.
Method R (Compound 2072) 2-Cyclopropyl-l-oxo-1,2-dihydro-isoquinoline-5-carboxylic acid 3,4-dichloro-benzylamide
-51 -


a. 2-Cvclopropyl-N-(3.4-dichlorobenzvD-l-oxo-1.2-dihydroisoquinoline-5-carboxamide
A 5-mL process vial was charged with 2-cycIopropyl-5-iodoisoquinolin-l(2H)-one (100 mg, 0.0004 mol), 3,4-dichloro-benzylamine (200 mg, 0.001 mol), molybdenum hexacarbonyl (90 mg, 0.0004 mol), palladium acetate (8 mg, 0.00004 mol), 1,8-diazabicyclo[5.4.0]undec-7-ene (200 mg, 0.001 mol) and 1,4-dioxane (2 mL, 0.02 mol). The vessel was sealed under air and exposed to microwave heating for 15 min at 110 °C. The reaction tube was thereafter cooled to room temperature, and the mixture was concentrated and dissolved in a small volume of dichloromethane. The crude product was purified via flash column chromatography (12 g of silica gel, 50% EtOAC/Hexane) and then prep. HPLC to give the desired product as a white solid.
'H NMR (CDC13) 5: 8.47 (d, J =7.9 Hz, 1H), 7.72 (d, J = 7.2 Hz, 1H), 7.49 (d, J =1.9 Hz, 1H), 7.45-7.39 (m, 2H), 7.26-7.23 (m, 1H), 7.16 (d, J = 7.8 Hz, 1H), 6.93 (d, J = 7.7 Hz, 1H), 6.52 (br, 1H), 4.64 (d, J = 6.0 Hz, 2H), 3.33 (br, 1H), 1.17-1.12 (m, 2H), 0.91-0.86 (m, 2H). MS m/z(M+H) 387.2
Example 1
The P2X7 receptor is strongly expressed in macrophage-derived cell lines, including, but not limited to, J774 (mouse macrophage line, American Type Culture Collection (ATCC), Rockville, MD, ATCC TIB-67), P388 (mouse cell line, ATCC CCL-46), P815 (mouse mast cell mastocytoma-derived line, ATCC TIB-64), THP-1 (Human monocyte-derived cell line, ATCC TIB202) and U937 (human cell line derived from histiocytic lymphoma, induceable to monocyte differentiation, ATCC CRL-1593.2) and in isolated macrophage cultures. Human or non-human animal macrophages are isolated using the procedure noted below.
The P2Z/ P2X7 receptor can be characterized by measuring channel opening, for instance ion flux, and/or by assessing pore formation, including by monitoring dye uptake or cell lysis in cells naturally expressing this receptor. Compounds such as ATP, 2' and 3*-(0)-(4-benzoyl benzoyl) ATP (BzATP) effect the formation of pores in the plasma membrane of these cells, particularly at low extracellular divalent ion concentrations (Buisman et al, Proc. Natl. Acad. Sci. USA 85:7988 (1988); Zambon et al, Cell. Immunol 156:458 (1994); Hickman et al Blood 84:2452 (1994)). Large molecular size dyes, including propidium dye YO-PRO-1, can be seen entering macrophage-derived cell lines during cell recordings (Hickman et al, Blood 84:2452 (1994); Wiley et al, Br J Pharmacol 112:946 (1994); Steinberg et al, J Biol Chem 262:8884 (1987)). Ethidium bromide (a fluorescent DNA probe) can also be monitored, where an increase in the fluorescence of intracellular DNA-bound ethidium bromide is observed. Expression of recombinant rat or human rP2X7 in cells, including HEK293 cells, and in Xenopus oocytes demonstrates influx and pore formation by whole cell recordings and YO-PRO-1 fluorescence
-52-

(Suprenant et al, Science 272:735 (1996); Rassendren et al, J Biol Chem 272:5482 (1997)).
The compounds of the invention may be tested for antagonist activity at the P2X7 receptor. Tests to be performed include and are selected from: (i) electrophysiological experiments; (ii) YO-PROl fluorescence; (iii) ethidium bromide fluorescence; and (iv) IL-lp release from stimulated macrophages, including as described below. Compounds can be tested in vivo in animal models including for inflammation models (e.g. paw edema model, collagen-induced arthritis, EAE model of MS).
Isolation of Human Macrophages
Monocyte-derived human or non-human animal macrophage cultures are prepared as described by Blanchard et al (Blanchard et al, J Cell Biochem 57:452 (1995); Blanchard et al, J Immunol 147:2579 (1991)). Briefly, monocytes are isolated from leukocyte concentrates obtained from a healthy volunteer. Leukocytes are suspended in RPMI 1460 medium (Life Techologies, Inc.) with 20% serum (human for human cells), 2mM glutamine, 5mM HEPES, and lOOug/ml streptomycin. Cells are allowed to adhere to culture flasks for l-2h, after which nonadherent cells are washed away. Adherent cells are cultured for 7-14d in this medium plus interferon-? (human for human cells) (1000 units/ml). Macrophages are recovered from the culture flask by pipetting with cold phosphate-buffered saline and plated onto glass coverslips for electrophysiological or other experiments carried out 12-24h later.
Example 2
Electrophysiological Experiments Whole cell recordings are made using the EPC9 patch-clamp amplifier and Pulse acquisition programs (HEKA, Lambrecht, Germany). Whole-cell recordings are obtained from cells, e.g. J774A.1 cells (American Type Culture Collection, Rockville, MD, ATCC TIB-67)); agonists are applied for periods of 1 to 3 s by a fast-flow U-tube delivery system [E.M. Fenwick, A. Marty, E. Neher, J. Physiol, (London) 331, 577 (1982)]. The internal pipette solution is 140 mM cesium-aspartate or potassium-aspartate, 20 mM NaCl, 10 mM EGTA, and 5 mM Hepes; normal external solution is 145 mM NaCl, 2 mM KC1, 2 mM CaCl2, 1 mM MgCl2i 10 mM Hepes, and 12 mM glucose. Low divalent external solution is nominally magnesium-free with 0.3 mM CaCl2. Concentration-response curves are constructed in low divalent solution by recording currents in response to 1 s applications of agonist at 8 min intervals with normal external solution present for 6 min before each application. This protocol is necessary to prevent the development of sustained inward currents.
Reversal potentials (Erev) are obtained by application of ATP (300 uM) or BzATP (30 uM)(controls), or the compound being tested, while the membrane is held at various potentials or by application of voltage ramps from -120 to 30 or 50 mV. Permeability ratios are calculated from Erev by first computing a (= PNH/PIC where P is permeability) for internal (i) and external (o) concentrations [Na]| = 20 mM, [Na]0 = 145 mM, [K]0 = 0 mM, and [K], = 140 mM from a = ([\45/Qxp(EKVFIRT)] - 20)/140 (where F is the Faraday, R is the gas constant, and T is the absolute temperature). Other PX/PN3 values, when PC]0 = 145 mM, [Na], = 20 mM, [K], = 140 mM, and [Na]0 - [K]0 = [X]! = 0 mM, are computed from Px/PNa = [(exp)ErejF/RT)] (20 + 140a))/145. In order of size, X is
-53-

cesium, methylamine, tris(hydroxymethyl)-aminomethane, tetraethylammonium, and N-methyl-D-glucamine. The internal solution also contains 10 mM EGTA and 5 mM Hepes. External solutions also contain 10 mM glucose and normal or low concentrations of divalent cations; pH is maintained at 7.3 with HC1, histidine, or Hepes as required, and the osmolality of all solutions is 295 to 315.
Example 3
YO-PROl Fluorescence The Photonics Imaging (IDEA) system for microscopic fluorescence measurements (Photonics, Planegg, Germany) is used. Coverslips are placed at the stage of a Zeiss Axiovert 100 or equivalent inverted microscope and viewed under oil immersion with a 40X Fluor objective. YO-PRO-1 (10 uM; Molecular Probes, Eugene, OR) is added to the supervision fluid during electrophysiological recordings 3 to 6 min before switching to low divalent solution and washed out upon switching back to normal divalent solution, after which the fluorescent lamp is turned on and cells are examined with a fluorescein isothiocyanate filter. YO-PROl fluorescence is measured using 491/509 nm excitation/emission wavelengths. Images are obtained at 5-20s intervals during continuous superfusion (2ml/min) with YO-PROl and varying concentrations of control ATP, BzATP or compound to be tested. For each experiment, the time course of YO-PROl fluorescence obtained for 10-20 individual cells and then averaged to obtain the mean fluorescence signal. Results were expressed as mean signal at 3 min for rP2X7 and the signal at 10 min is used for P2X7 and human macrophage cells. All experiments are carried out at room temperature.
Example 4
Ethidium Bromide Compounds of the invention are tested for antagonist activity at the P2X7 receptor by monitoring Ethidium Bromide entering P2X7 receptor-expressing cells on pore formation. The test is performed in 96-well flat bottomed microtitre plates, the wells being filled with 250 ui of test solution comprising 200 ul of a suspension of P2X7- expressing cells (e.g. THP-1 cells, J774 cells, etc.)(2.5 xlO6 cells/ml) containing 10"*M ethidium bromide, 25 ul of a high potassium buffer solution containing 10"5M BzATP, and 25 ul of a high potassium buffer solution containing test compound. The plate is covered with a plastic sheet and incubated at 37° C for one hour. The plate is then read in a Perkin-Elmer fluorescent plate reader, excitation 520 nm, emission 595 nm, slit widths; Ex 15 nm, EM 20 nm. For the purposes of comparison, BzATP (a P2X7 receptor agonist) and pyridoxal 5-phosphate (a P2X7 receptor agonist) are used separately in the test as controls. From the readings obtained, a pIC5o figure is calculated for each test compound. This figure is the negative logarithm of the concentration of test compound necessary to reduce the BzATP agonist activity by 50%.
Example 5 IL-ip Release This Example demonstrates the testing of the compounds of this invention for efficacy as inhibitors of P2X7-mediated release of IL-lfl from human macrophages activated by the Alzheimer's beta amyloid peptide 1-42.
-54-

Cell isolation
Monocytes are isolated from peripheral blood mononuclear cells (PBMCs) as follows. Whole blood is layered directly onto Histopak 1077-1 columns (Sigma Biochemicals) and centrifuged at 800xg for 15 minutes. The PBMC band of cells is removed to a fresh 50 ml culture tube and diluted 1:1 with wash buffer (Phosphate buffered saline, pH 7.4 containing 2 mM EDTA and 5 mg/ml BSA) followed by centrifugation at 800xg for 5 minutes. Cells are then washed by sequential resuspension of the cell pellet in wash buffer .and centrifugation at 600xg for 5 minutes. The wash process is repeated until the supernatent is clear of contaminating platelets (generally, 5 to 6 washes). Monocytes are then purified from the PBMCs by negative selection using a monocyte isolation kit (Miltenyi Biotec, Inc.) that contains antibodies to non-monocytic cells, running the cells over a magnetic column to remove antibody-bound cells, and collecting the flow through volume of monocytes. Monocytes are washed once with wash buffer and seeded at 100,000 cells per well in 100 ul serum-free RPMI 1640 in 96-well plates and incubated for 1 hour at 37° C in a 5% C02/95% humidified tissue culture incubator. After 1 hour, the medium is replaced with 100 ul complete culture medium (RPMI 1640, 10% human serum-type AB (heat inactivated), 25 mM HEPES, 2 mM glutamine, 50 U/ml each of penicillin and streptomycin) and incubated overnight (16 hours).
Dosing regimen
The next day, the culture medium is replaced with 100 fresh complete culture medium in the absence or presence of human beta amyloid 1-42 peptide (5 uM) and incubated at 37° C in a 5% C02/95% humidified tissue culture incubator for 5 hours. Medium is then removed and discarded. Each well is washed once with Hanks buffered saline (HBSS) containing 1 mM CaCl2 followed by the addition of 80 of HBSS/CaCl2-inhibiting compound of the present invention (lOx stock in HBSS/CaCl2 for a final concentration of 23 nM and 206 nM) and incubated 15 minutes in the tissue culture incubator followed by the addition of either 10 ul of HBSS/CaCl2or 10 ul of benzoyl ATP (BzATP; 3 mM stock in HBSS/ CaCl2 for a 300 uM final concentration) and incubated for a further 30 minutes in the tissue culture incubator. Medium is then removed to new 96-well plates for storage at -70° C until the IL-16 content was quantitated by ELISA (from R&D Systems). The cells are washed once with HBSS/CaCl2 followed by lysing the cells with 100 \i\ ice cold lysis buffer (100 mM Tris, pH 7.6, 1% Triton X-100, and 1 tablet per 30 ml Complete TM protease inhibitor from Roche Biochemicals, Inc). Cell lysates are stored at -70° C until the IL- is quantitated by ELISA.
Example 6 In Vivo Animal Models A. This example illustrates the efficacy of the compounds of this invention in the treatment of multiple sclerosis.
As described herein, experimental autoimmune encephalomyelitis (EAE) model is used to show such an efficacy. The following procedures are employed in this model.
Animals
SJL/J female mice, 8 wks. old, are obtained from Jackson Laboratories.
-55-

Antigens
Myelin Proteolipid Protein (PLP 139-151) (HSLGKWLGHPDKF) (Cat # H-2478) is obtained from BACHEM, Bioscience, Inc., 3700 Horizon Dr., King of Prussia, Pa. 19406, 1-610-239-0300 (phone), 1-610-239-0800 (fax).
Complete Freund's Adjuvant H37 Ra [1 mg/ml Mycobacterium Tuberculosis H37 Ra] is obtained from Difco 1-800-521-0851 (Cat # 3114-60-5, 6X10 ml).
Mycobacterium Tuberculosis is also obtained from Difco, 1-800-521-0851 (Cat #3114-33-8,6.times.l00mg).
Pertussis Toxin
Bordetella Pertussis, (Lyophilized powder containing PBS and lactose) is obtained from List Biological Laboratories, 1-408-866-6363 (Product #180, 50 ug).
Induction of EAE in Mice
PLP139-151 peptide is dissolved in H20:PBS (1:1) solution to a concentration 7.5 mg/10 ml (for 75 fig PLP per group) and emulsified with an equal volume of CFA supplemented with 40 mg/10 ml heated-killed mycobacterium tuberculosis H37Ra. Mice are injected s.c. with 0.2 ml of peptide emulsion in the abdominal flank (0.1 ml on each side). On the same day and 72 hours later, mice are injected i.v. with 100% of 35 ng and 50 ng of Bordetella Pertussis toxin in saline respectively.
Clinical Assessment
STAGE 0: Normal
STAGE 0.5: Partial limp tail
STAGE 1: Complete Limp Tail
STAGE 2: Impaired righting reflex
STAGE 2.5: Righting reflex is delayed (Not weak enough to be stage 3).
STAGE 3: Partial hind limb paralysis
STAGE 3.5: One leg is completely paralyzed, and one leg is partially paralyzed,
STAGE 4: Complete hind limb paralysis
STAGE 4.5: Legs are completely paralyzed and Moribund
STAGE 5: Death due to EAE
Clinical Courses of EAE
Acute phase: First clinical episode (Day 10-18)
Remission: Phase of clinical improvement following a clinical episode; characterized by a reduction (>=one grade) in clinical score for at least two days after the peak score of acute phase or a disease relapse.
Relapse: Increase of at least one grade in clinical score for at least two days after remission has been attained.
The animals treated with the compounds of this invention generally would be expected to show improvements in clinical scores.
-56-

B. This Example illustrates a protocol for determining the efficacy of the compounds of the present invention for the treatment of stroke using an animal model.
Male Sprague Dawley rats (Charles River) weighing 280-320 g are given free access to food and water and acclimatized for a minimum of 4 days before use in experiments. All rats for use in studies are to be fasted beginning at 3;00 pm the day prior to surgery but given free access to water. Prior to surgery each rat is weighed. The rat is initially induced with 5% isoflurane (Aerrane, Fort Dodge), combined with 30% 02, 70% N20 for 2-5 minutes. The rat is then placed on a circulating water-heating pad and into a nose cone for spontaneous respiration of anesthetic gases. The isoflurane is reduced to 2%. A rectal probe is inserted and body temperature maintained at 36.5-37.5° C. The hair is clipped at all surgical sites and these regions will then be scrubbed with Betadine.
Surgical Procedure
A temporalis muscle probe is placed into the right temporalis muscle and "brain" temperature" is monitored. A midline neck incision is made in the upper thorax of the rat. Careful dissection, isolation and retraction of the sternomastoideus, digastricus, and sternohyoideus muscles is made to expose the right common, internal and external carotid arteries. The right common carotid artery is isolated with a 5-0 silk suture. During surgery the suture is released allowing reperfusion every 2-4 minutes. The right external caTotid and superior thyroid arteries are also isolated and the superior thyroid is cauterized, while the external carotid is ligated distally with a 5-0 silk suture. Another 5-0 silk suture is loosely tied around the external carotid artery. The occipital artery is isolated, ligated and incised. The internal carotid is isolated.
With the common and external carotid arteries immobilized, an aneurysm clip is placed onto the internal carotid artery. A small incision is made at the distal end of the external carotid. A 3-0 nylon suture coated with1 poly-L-lysine is then inserted into the external carotid and up into the common carotid artery. The loosely tied 5-0 silk suture around the external carotid is now gently tightened around the filament. The external carotid artery is then incised and the remaining piece of the external carotid artery with the filament is rotated so that the filament may be inserted into the internal carotid artery the length of insertion depending on the weight and rat strain. In Sprague Dawley rats the monofilament is inserted 18-19 mm (18 mm for rats weighing The external jugular vein will be cannulated with PE 50 tubing for I.V. administration of compounds. The cannula will be exteriorized at the previously shaven, scruff of the neck and sutured in place. The wound will be closed by means of suture. The right femoral artery is catheterized for blood gas and glucose determination during surgery.
Two hours after the insertion of the monofilament suture the rats are re-anesthetized with the same anesthetic combination used initially and placed back into the nose cone with the reduction of isoflurane concentration to 2%. The neck incision is reopened to expose the external carotid artery. The restoration of blood flow is accomplished by completely withdrawing the intraluminal suture from the carotid arteries. The incision is then closed with 3-0 silk in an interrupted stitch.
-57-

Compound Administration
Five groups of 15 animals are subjected to the above methodology. Compounds are infused (I.V.) at various doses (dose response) over different time period's post MCAo. A pre-determined concentration is infused over a pre-selected time period beginning at various intervals post MCAo. Vehicle-treated controls receive an infusion of normally 0.9 ml/hr. A positive control compound is run at the same time.
Neurological Tests
Prior to surgery, 2 hours following the onset of ischaemia and 24 hours after ischaemia a battery of neurological tests are performed. The postural reflex test, which is designed to examine upper body posture, when the rat is suspended by the tail above a flat surface. A normal rat will extend the entire body and both forelimbs towards the surface. Rats with an infarction will consistently flex the contralateral limb and show signs of body rotation. The rats respond to a gentle lateral push with a finger behind the shoulders. A normal rat would resist such a push, whereas a rat with an infarction will not. The elicited forelimb placing in response to visual and tactile stimuli. The animal is held by the body so that the lateral or dorsal forepaw surface is placed against a bench. This test is repeated but on this occasion obstructing the view of the rat.
Upon completion of each experiment, all animals are deeply anaesthetized with isoflurane (5%), euthanized by decapitation, and the brains removed, the extent and location of the ischaemic damage is verified histologically by means of tetrazolium chloride. C. This Example illustrates the anti-inflammatory activity of the compounds of this invention using a model of 2,4-dinitrobenzenesulfonic acid (DNBS) induced distal colitis (a model of inflammatory bowel disease).
Test Substance and Dosing Pattern
A compound of this invention is dissolved in vehicle of 2% Tween 80 in distilled water for oral administration at a dose of 50 mg/kg or dissolved in vehicle of 2% Tween 80 and 0.9% NaCl for intraperitoneal injection at 30 mg/kg. The dose is given once daily for 7 consecutive days. Dosing volume is 10 ml/kg. DNBS was challenged 2 hours after dosing on the second day.
Animals
In these studies, male Wistar, Long Evans rats provided by animal breeding center of MDS Panlabs Taiwan, Ltd. and Balb/cByJ derived male mice (weighing 20±2 gms), provided by National Laboratory Animals Breeding Research center (NALBRC, Taiwan), may be used. Space allocation of 6 animals may be 45x23x15 cm. Animals are housed in APEC® cages (Allentown Caging, Allentown, NJ. 08501, USA) in a positive pressure isolator (NuAire®, Mode: Nu-605, airflow velocity 50±5 ft/rnin, HEPA Filter) and maintained in a controlled temperature (22°C -24°C) and humidity (60%-80%) environment with 12 hours light dark cycles for at least one week in MDS Panlabs Taiwan laboratory prior to being used. Free access to standard lab chow for rats (Fwusow Industry Co., Limited, Taiwan) and tap water is granted. All aspects of this work including housing, experimentation and disposal of animals would be performed in general accordance with the International Guiding Principles for Biomedical Research Involving Animals (CIOMS Publication No. ISBN 92 90360194, 1985).
-58-

Chemicals
DNBS is obtained from TCI,. Tokyo, Japan, ethanol is from Merck, Germany and Sulfasalazine is purchased from Sigma, USA.
Equipment
Electriconic scale (Tanita, model 1140, Japan), Electriconic scale (Sartorius, R160P, Germany), Glass syringe (2 ml, Mitsuba, Japan), Rat oral needle, Hypodermic needle (25G.times.l"TOP Corporation, Japan), Stainless Scissors (Klappenclear, Germany), Stainless Forceps (Klappenclear, Germany).
Method
Groups of 3 Wistar derived male rats weighing 180±20 gms are used. Distal colitis is induced by intra-colonic instillation of DNBS (2,4-dinitrobenzene sulfonic acid, 30 mg in 0.5 ml ethanol 30%) after which, 2 ml of air is gently injected through the cannula to ensure that the solution remains in the colon. Test substance is administered orally (PO) at a dose of 50 mg/kg or intraperitoneally (IP) at 30 mg/kg once daily for 7 consecutive days. DNBS is instillated into the distal colon of each animal 2 hours after dosing on the second day. The control group is similarly treated with vehicle alone and sulfasalazine (300 mg/kg, PO) is used as reference agent. Animals are fasted 24 hours before DNBS challenge and 24 hours after the final treatment when they are sacrificed and each colon is removed and weighed. During the experiments, presence of diarrhea is recorded daily. When the abdominal cavity is opened before removal of the colon, adhesions between the colon and other organs are noted. After weighing the colon, the extent of colonic ulceration is observed and noted as well. Colon-to-body weight ratio is then calculated for each animal according to the formula: Colon (g)/BWxl00%. The "Net" increase in ratio of Vehicle-control +DNBS group relative to Vehicle-control group is used as a base value for comparison with test substance treated groups and expressed as % decrease in inflammation. A 30 percent or more (30%) decrease in "Net" colon-to-body weight ratio for each test substance treated group relative to the "Net" vehicle+DNBS treated group is considered significant.
D. This Example illustrates the anti-inflammatory activity of the present compounds using a model of carrageenan induced paw edema (a model of inflammation, carrageenan).
Test Substance and Dosing Pattern
A compound of this invention is dissolved in vehicle of 2% Tween 80/0.9% NaCI and administered intraperitoneally at a dose of 30 mg/kg 30 minutes before carrageenan (1% 0.1 ml/paw) challenge. Dosing volume is 10 ml/kg.
Animals
Animals are conditioned in accordance with the procedures set forth in the previous Example.
Chemicals
Carrageenan is obtained from TCI, Japan; Pyrogen free saline is from Astar, Taiwan; and Aspirin is purchased from ICN BioMedicals, USA.
-59-

Equipment
Glass syringe (1 ml and 2 ml Mitsuba, Japan), Hypodermic needle 24Gxl" (Top Corporation, Japan), Plethysmometer #7150 (UGO Basile, Italy), and Water cell 25 mm Diameter, #7157 (UGO Basile, Italy).
Method
Test substance (Example) is administered IP (30 mg/kg) to groups of 3 Long Evans derived male overnight fasted rats weighing 150±20 gms 30 minutes before right hind paw injection of carrageenan (0.1 ml of 1% suspension intraplantar). Hind paw edema, as a measure of inflammation, is recorded 3 hours after carrageenan administration using a plethysmometer (Ugo Basile Cat. #7150) with water cell (25 mm diameter, Cat. #7157). Reduction of hind paw edema by 30 percent or more ( 30%) indicated significant acute anti-inflammatory activity.
E. This Example illustrates the anti-inflammatory activity of the present compounds using a model of Balb/c mice subjected to monoclonal antibody (mAb) type II collagen induced arthritis.
Test Substance and Dosing Pattern
A compound of this invention is dissolved in vehicle of 2% Tween 80/0.9% NaCl, at doses of 50 or 30 and administered orally (50 mg/kg) or intraperitoneally at 30 mg/kg once daily for 3 consecutive days after monoclonal antibody of collagen was injected, Dosing volume is 20 ml/kg.
Animals
Animals are conditioned in accordance with the procedures set forth in the previous Example.
Chemicals
Lipopolysaccharide is obtained from Sigma, USA; Indomethacin is from Sigma, USA; Arthrogen-CIA.TM Monoclonal Antibodies D8, F10, DI-2G and A2 are obtained from IBL, Japan; Phosphated-Buffer Saline is purchased from Sigma, USA; and Tween 80 is from Wako, Japan.
Equipment
Plethysmometer (Ugo Basile, Italy) and Water Cell (Ugo Basile, Italy).
Method
Groups of 5 Balb/cByJ mice strain, 6-8 weeks of age, are used for the induction of arthritis by monoclonal antibodies (mAbs) responding to type II collagen, plus lipopolysaccharide (LPS). The animals are administered intravenously with a combination of 4 different mabs in a total of 4 mg/mouse at day 0, and followed by intravenous 25 of LPS 72 hours later (day 3). From day 3, one hour after LPS administration, ML-659 at 50 mg/kg (PO) or 30 mg/kg (IP) and vehicle (2% Tween 80/0.9% NaCI, PO) as well as the positive control indomethacin, 3 mg/kg (PO) are administrated once daily for 3 consecutive days. A plethysmometer (Ugo Basile Cat #7150) with water cell (12 mm diameter) is used for the measurement of increase in volume of the two hind paws at day 0, 5, 7, 10, 14, and 17. The percent inhibition of increase in volume is calculated by the following formula:
-60-

Inhibition (%): [l-(Tn-To)/(Cn-Co)]x100 Where:
Co (Cn): volume of day 0 (day n) in vehicle control To (Tn): volume of day 0 (day n) in test compound-treated group
The reduction of both of two hind paws edema by more than 30% is considered significant.
Example 7
Neuropathic Pain Model This example illustrates the analgesic activity of the compounds of this invention using a Sciatic Nerve ligation model of mononeuropathic pain
Test system
Adult male Sprague Dawley (SD) rats weighing 250-300 gin (Charles River Laboratories, San Diego, CA) are used. The animal room is lighted artificially at a 12-hr light-dark cycle (from 7:00 A.M. to 7:00 P.M) with water and food supply ad libitum. Animals are allocated randomly into groups.
Model induction
Sciatic nerve ligation (SNL, Seltzer's model):
Under anesthesia with pentobarbital (50 mg/kg, i.p.) and aseptic techniques, the selective nerve injury is created by tightly ligating the selective portion of the common sciatic nerve according to the method of Seltzer (1990). Briefly, the high-thigh level of the left sciatic nerve is exposed after skin incision and blunt separation of muscles at a site near the trochanter just distal to the point at which the posterior biceps semitendious nerve nerve branches from the common sciatic nerve. The nervtf is then fixed in this position with fine forceps by pinching the epineurium on its dorsal aspect, taking care not to press the nerve against underlying structures. An 8-0 silicon-treated silk suture is inserted into the nerve with a curved, reversed-cutting mini-needle, and tightly ligated so that the dorsal - Vi of the nerve is trapped in the ligature. The muscles are sutured in layers, and the skin closed with wound clips. Animals are then returned to their home cages. Rats exhibiting postoperative neurological deficits or poor grooming are excluded from the experiments.
Equipment
The following equipment is used in the current studies: von Frey filament set (Touch-test Sensory Evaluator, North Coast Medical Inc., Morgan Hill, CA).
Statistical Methods:
Within each experiment mean, standard error of the mean (SEM) and statistical significance are calculated using the average, standard error of the mean and unpaired, two-tailed t-Test functions, respectively, using Microsoft Excel®. Statistical significance of effects observed between individual experiments is determined, using Prism (GraphPad Software Inc., San Diego, CA) for the one-way or two-way analysis of variance (ANOVA) function. Statistical analyses are performed with a confidence limit of 0.95 and a significance level of 0.05.
-61-

Example 8 Pore Formation
THP-1 cells (ATCC Cat # 285-IF-lOO) are plated in 96 well plates at a concentration of 200,000 cells per well and allowed to differentiate in RPMI-1640 media (ATCC Cat # 30-2001) containing 10% FBS, 100 IU/mL penicillin, 100 ug/mL streptomycin, 100 ng/mL LPS and 100 ng/mL IFN-y for 16 hours. Following differentiation, the cells are pretreated with the compound of interest at the appropriate concentration for 30 minutes in RPMI-1640 media containing 100 IU/mL penicillin, 100 ug/mL streptomycin. The pretreatment media is then replaced with assay buffer (20 mM HEPES, 10 mM d-glucose, 118 mM NMDG, 5 mM KC1, 0.4 mM CaCl2) containing 5 uM Yo-Pro 1 (Molecular Probes Cat # Y3603) and the compound of interest at the appropriate concentration and the cells are incubated for an additional 10 minutes. 2',3'-0-(4-benzoylbenzoyl)-adenosine 5'-triphosphate (Sigma Aldrich Cat# B6396) is then added to a final concentration of 40 uM and fluoroscence readings measured at 491/509 excitation/emission every minute for 50 minutes using a Tecan Safire plate reader. During this time temperature is maintained at of 37°C. Background adjusted fluorescence levels between drug treated and non-treated cells are used to calculate the percent inhibition.
Example 9 IL-1R Release Assay THP-1 cells (ATCC Cat # 285-IF-lOO) are plated in 96 well plates at a concentration of 200,000 cells per well and allowed to differentiate in RPMI-1640 media (ATCC Cat # 30-2001) containing 10% FBS, 100 IU/mL penicillin, 100 ug/mL streptomycin, 100 ng/mL LPS and 100 ng/mL IFN-y for 16 hours. Following differentiation, the cells are treated for an additional 2 hours in RPMI-1640 media containing 100 IU/mL penicillin, 100 ug/mL streptomycin and fresh LPS at 100 ng/mL. The cells are then pretreated for 30 minutes with the compound of interest at the appropriate concentration in RPMI media containing 100 IU/mL penicillin, 100 ug/mL streptomycin. Following the pretreatment 2',3'-0-(4-benzoylbenzoyl)-adenosine 5'-triphosphate (Sigma Aldrich Cat # B6396) is added to a final concentration of 250 uM and the cells are incubated for an additional 45 minutes. 30 uL of cell supernatant is then collected and IL-1B levels determined via ELISA (R&D systems Cat. # HSLB50) according to manufacturer's recommendations using the Tecan Safire plate reader. Background adjusted IL-16 levels of drug treated and non-treated cells are used to calculate the percent inhibition.
The synthetic and biological examples described in this application are offered to illustrate this invention and are not to be construed in any way as limiting the scope of this invention. In the examples, all temperatures are in degrees Celsius (unless otherwise indicated). The compounds that have been prepared in accordance with the invention along with their biological activity data are presented in following Table. The syntheses of these representative compounds are carried out in accordance with the methods set forth above.
Exemplary Compounds of the Invention
The following compounds have been or can be prepared according to the synthetic methods described above. For the purpose of Table 1 below, activity of each compound,
-62-

which can be determined using the IL-lp assay method described in Example 9, is expressed as follows:
"+" compound exhibited 0-25% inhibition at 0.3 uM concentration
"++" compound exhibited 26-50% inhibition at 0.3 uM concentration
"+++" compound exhibited 51-75% inhibition at 0.3 uM concentration
"++++" compound exhibited 76% or greater inhibition at 0.3 uM concentration
Compounds with a percent inhibition represented by "++++" are of particular interest.
TABLE 1: IL-lp % Inhibition of Exemplary Compounds

ID Structure MW MS (obs) IL-lp % Inhib. @ 0.3 uM
2001 i 406.91 +
2002 274.37 +
2003 282.35 +
2004 364.49 +
2005 416.57 417.82 ++++
2006 365.48 366.60 +
-63-

ID Structure ■Mwy MS (obs) IL-lp % Inhib.
@0.3uM ^
2007 365.48 366.63 +
2008 398.94 399.32 +
2009 398.94 399.32 +
2010 365.48 366.66 +
2011 398.94 399.34 +
2012 394.52 395.60 +
2013 378.52 379.73 +
2014 442.58 443.64 +
2015 421.54 422.67 +

ID Structure MW ' MS (obs) IL-lp %llnhib;@0.3uM T"






2016 394.52 394.90 +
2017 408.54 409.41 +
2018 454.61 455.49 +
2019 452.55 451.46 +-H-
2020 K
474.60 475.66 ++++
2021 434.56 435.90 ++
2022 434.56 435.41 +
2023 1 446.59 447.72 ++
-65-

ID Structure MW MS (obs) IL-lp % Inhib. @ 0.3 uM





2024 430.59 431.84 ++++
2025 451.01
451.37 ++
2026 434.56 435.75 ++++
2027 460.57 461.72 +++
2028 336.43 ++++
2029 352.43
2030 314.38 315.20 +++
2031 i 422.50 422.50 ++++
2032 328.41 329.20 ++-H-
-66-

ID Structure MW MS (obs) IL-lp % Inhib. @ 0.3 uM

2033 462.54 463.50 ++++
2034 412.53 413.40 -H-+
2035 400.52 401.00 ++++
2036 446.54 447.50 ++-H-
2037 374.48 375.10 +++
2038 412.53 413.40 +
2039 372.46 373.10 -H-+
2040 372.46 373.30 -H-H-
-67-


-68-


-69-


-70-


-71 -





-72-



IC50 Determinations
The compounds set forth in Table 1 were tested for activity in a cellular model as described herein. Specifically, cells were pretreated with differing amounts of the compound under test and released IL-lβ determined as in Example 9, above. Measurements were made and IC50 values, presented in Table 2, below, were determined by fitting the data to a four parameter logistic equation using GraphPad Prism software (GraphPad Software, Inc). The equation may be expressed by the following formula:
Y=Bottom + (Top-Bottom)/(l+10^ ((LogEC50-X)*HillSlope)) Where X is the logarithm of concentration,Y is the response and Y starts at Bottom and goes to Top with a sigmoid shape.
TABLE 2: IL-1β ICS0 for Exemplary Compounds
















-73-


-74-

Half-life in human liver microsomes (HLM)
Test compounds (1 μM) are incubated with 3.3 mM MgCl2 and 0.78 mg/mL HLM (HL101) in 100 mM potassium phosphate buffer (pH 7.4) at 37°C on the 96-deep well plate. The reaction mixture is split into two groups, a non-P450 and a P450 group. NADPH is only added to the reaction mixture of the P450 group. An aliquot of samples of P450 group is collected at 0, 10, 30, and 60 min time point, where 0 min time point indicated the time when NADPH is added into the reaction mixture of P450 group. An aliquot of samples of non-P450 group is collected at -10 and 65 min time point. Collected aliquots are extracted with acetonitrile solution containing an internal standard. The precipitated protein is spun down in centrifuge (2000 rpm, 15 min). The compound concentration in supernatant is measured by LC/MS/MS system.
The half-life value is obtained by plotting the natural logarithm of the peak area ratio of compounds/ internal standard versus time. The slope of the line of best fit through the points yields the rate of metabolism (k). This is converted to a half-life value using following equations:
Half-life = In 2 /k
The results of the tests and corresponding T1/2 values are set forth in Table 3, below.
TABLE 3: T-Half Life In Hours For Exemplary Compounds







-75-


Pharmacokinetic Evaluation of compounds following Intravenous and oral administration in rats.
Male Sprague-Dawley rats are acclimatized for at least 24 hours prior to experiment initiation. During the acclimation period, all animals receive food and water ad libitum. However, food but not water is removed from the animal's cages at least 12 hours before initiation of the experiment. During the first 3 hours of experimentation, the animals receive only water ad libitum. At least three animals each are tested for intravenous and oral dosage. For intravenous formulation, compounds were dissolved (0.25 to 1 mg/mL) in a mixture of 3% dimethyl sulfoxide, 40% PEG 400 and the rest percentage of 40% Captisol in water (w/v). The animals are weighed before dosing. The determined body weight is used to calculate the dose volume for each animal.
Dose volume (mL/kg) = 1 mg/kg/formulation concentration (mg/mL) In instances where the formulation concentrations were less than 0.5 mg/mL, the dosing volume is about 2 mL/kg.
For oral formulation, compounds of this invention are suspended (0.5 to 0.75 mg/mL) in a mixture of 5% of 10% Tween 80 in water (v/v) and 95% of 0.5 % methyl cellulose in water (w/v). PO rats are typically dosed through oral gavage following the same dose volume formula as IV to achieve a dose level of 1 to 5 mg/kg. For IV dosing, blood samples are collected (using a pre-heparinized syringe) via the jugular vein catheter at 2, 5, 15, 30, 60, 120, 180, 300, 480, and 1440 minutes post dosing. For PO dosing, blood samples are collected (using a pre-heparinized syringe) via the jugular vein catheter before dosing and at 5, 15, 30, 60, 120, 180, 300, 480, and 1440 minutes post dosing. About 250 uL of blood is obtained at each time point from the animal. Equal volumes of 0.9% normal saline are replaced to prevent dehydration. The whole blood samples are maintained on ice until centrifugation. Blood samples are then centrifuged at 14,000 rpm for 10 minutes at 4°C and the upper plasma layer transferred into a clean vial and stored at -80°C. The resulting plasma samples are then analyzed by liquid chromatography-tandem mass spectrometry. Following the measurement of plasma samples and dosing solutions, plasma concentration-time curve is plotted. Plasma exposure is calculated as76
the area under the concentration-time curve extrapolated to time infinite (AUCnf). The AUCinf is averaged and the oral bioavailability (%F) for individual animal is calculated as:
AUCinf (PO)/AUCinf (IV), normalized to their respective dose levels.
The %F can be reported as the mean %F of all animals dosed orally with the compound
of the invention at the specified level.
From the foregoing description, various modifications and changes in the compositions and methods of this invention will occur to those skilled in the art. All such modifications coming within the scope of the appended claims are intended to be included therein.
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.
The chemical names of compounds of invention given in this application are generated using Open Eye Software's Lexichem naming tool, Symyx Renaissance Software's Reaction Planner or MDL's ISIS Draw Autonom Software tool and not verified.












-77-

WE CLAIM:


1. A bicycloheteroaryl compound having a formula:
wherein
A is CR2aR2b or CO; B and Y are independently selected from CR2a and CR2aR2b;
W, W and Z are independently selected from CR4 and N, provided that all three
of W, W and 2 are not N at the same time;
L1 is a bond, SO, S02 or substituted or unsubstituted C1--C5 alkylene;
n is 0, 1,2,3 or 4;
R1 is selected from a substituted or unsubstituted 3-13 membered cycloalkyl,
heterocycloalkyl, aryl and heteroaryl ring;
each of R2a, R2b, R2 and R2' is independently selected from hydrogen, halo, and
substituted or unsubstituted C1-C6 alkyl; or any of R2 and R2 join together to form
a cycloalkyl or cycloheteroalkyl ring of 3-7 atoms;
R3 is selected from hydrogen, a hydrogen bond donor group, substituted or
unsubstituted cycloalkyl, substituted or unsubstituted alkyl, substituted or
unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or
unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted
or unsubstituted bicycloheteroaryl;
R4 is independently selected from H, alkyl, substituted alkyl, acyl, substituted acyl,
substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino,
substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy,
alkoxycarbonyl, substituted alkoxycarbonyl, substituted or unsubstituted
alkylarylamino, arylalkyloxy, substituted arylalkyloxy, amino, aryl, substituted
aryl, arylalkyl, substituted or unsubstituted sulfoxide, substituted or unsubstituted
sulfone, substituted or unsubstituted sulfanyl, substituted or unsubstituted
aminosulfonyl, substituted or unsubstituted arylsulfonyl, sulfuric acid, sulfuric
acid ester, substituted or unsubstituted dihydroxyphosphoryl, substituted or
unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or
unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl,
substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted
dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl,
substituted or unsubstituted heteroalkyl, hydroxy, nitro, and thio;
and the dotted bond is a single or a double bond;
or a pharmaceutically acceptable salt, solvate or prodrug thereof;
and stereoisomers, isotopic variants and tautomers thereof.
2. A compound according to Claim 1 wherein A is CR2aR2b.
3. A compound according to Claim 1 wherein A is CH2.





















-78-

4. A compound according to Claim 1 wherein A is CO.
5. A compound according to Claim 1 wherein each of B and Y is CR2aR2b; and the dotted bond is a single bond.
6. A compound according to Claim 1 wherein each of B and Y is CH2; and the dotted bond is a single bond.
7. A compound according to Claim 1 wherein each of B and Y is CR2a; and the dotted bond is a double bond.
8. A compound according to Claim 1 wherein each of B and Y is CH; and the dotted bond is a double bond.
9. A compound according to Claim 1 wherein each of R2' and R2 is H.
10. A compound according to Claim 1 wherein one of R2 and R2 is independently Me and the other is H.
11. A compound according to Claim 1 wherein each of R2 and R2 is Me.
12. A compound according to Claim 1 wherein n is 0,1, or 2.
13. A compound according to Claim 1 wherein R] is substituted or unsubstituted aryl.
14. A compound according to Claim 1 wherein R1 is substituted or unsubstituted phenyl or substituted or unsubstituted naphthyl.
15. A compound according to Claim 1 wherein R* is substituted or unsubstituted
heteroaryl.
16. A compound according to Claim 1 wherein R1 is substituted or unsubstituted pyridyl, substituted or unsubstituted quinoline, substituted or unsubstituted benzodioxole, substituted or unsubstituted benzodioxane, substituted or unsubstituted benzofuran, substituted or unsubstituted benzothiophene, and substituted or unsubstituted benzodioxepine.
17. A compound according to Claim 1 wherein R1 is substituted or unsubstituted adamantyl.
18. A compound according to Claim 1 wherein R1 is substituted or unsubstituted cyclopropyl, cyclopentyl, cyclohexyl or cycloheptyl.
19. A compound according to claim 1 wherein the compound is according to formula II, III or IV:
20. -79-


wherein
W is CR4; Z is CR4;
L1, R1, R2', R2", R3 and R4 are as in claim 1;
and R5 is selected from H, alkyl, substituted alkyl, acyl, substituted acyl,
substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino,
substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy,
alkoxycarbonyl, substituted alkoxycarbonyl, substituted or unsubstituted
alkylarylamino, arylalkyloxy, substituted arylalkyloxy, amino, aryl, substituted
aryl, arylalkyl, substituted or unsubstituted sulfoxide, substituted or unsubstituted
sulfone, substituted or unsubstituted sulfanyl, substituted or unsubstituted
aminosulfonyl, substituted or unsubstituted arylsulfonyl, sulfuric acid, sulfuric
acid ester, substituted or unsubstituted dihydroxyphosphoryl, substituted or
unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or
unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl,
substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted
dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl,
substituted or unsubstituted heteroalkyl, hydroxy, nitro, and thio;
or a pharmaceutically acceptable salt, solvate or prodrug thereof;
and stereoisomers, isotopic variants and tautomers thereof.
20. A compound according to Claim 19 wherein each of R2 and R2 is H.
21. A compound according to Claim 19 wherein R2 is CI or F; and R2 is H.
22. A compound according to Claim 19 wherein R2 is Me; and R2 is H.
23. A compound according to Claim 19 wherein R1 is substituted or unsubstituted adamantyl.
24. A compound according to Claim 19 wherein R1 is substituted or unsubstituted cyclohexyl or cycloheptyl.
25. A compound according to Claim 19 wherein R1, is selected from substituted or unsubstituted phenyl.
26. A compound according to Claim 19 wherein R1 is selected from substituted or unsubstituted naphthalene.
-80-


A compound according to claim 1 wherein the compound is according to formula V, VI or VII:
wherein
W is CR4; Z is CR4;
L1 R1, R2, R2, R3 and R4 are as in claim 1;
R5 is selected from H, alkyl, substituted alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy, alkoxycarbonyl, substituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, arylalkyloxy, substituted arylalkyloxy, amino, aryl, substituted aryl, arylalkyl, substituted or unsubstituted sulfoxide, substituted or unsubstituted sulfone, substituted or unsubstituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted arylsulfonyl, sulfuric acid, sulfuric acid ester, substituted or unsubstituted dihydroxyphosphoryl, substituted or unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroalkyl, hydroxy, nitro, and thio;
each R4a is selected from hydrogen, alkyl, substituted alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxy, aryloxy, alkoxycarbonyl, substituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, arylalkyloxy, substituted arylalkyloxy, amino, aryl, substituted aryl, arylalkyl, substituted or unsubstituted sulfoxide, substituted or unsubstituted sulfone, substituted or unsubstituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted arylsulfonyl, sulfuric acid, sulfuric acid ester, substituted or unsubstituted dihydroxyphosphoryl, substituted or unsubstituted aminodihydroxyphosphoryl, azido, carboxy, substituted or unsubstituted carbamoyl, cyano, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloheteroalkyl, substituted or unsubstituted dialkylamino, halo, heteroaryloxy, substituted or unsubstituted heteroaryl, substituted or unsubstituted heteroalkyl, hydroxy, nitro, and thio; and m is selected from 0-5;
or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.
A compound according to Claim 27 wherein each of R2 and R2 is H.
A compound according to Claim 27 wherein R2 is CI; and R2 is H.
A compound according to Claim 27 wherein R2 is Me; and R2 is H.
-81-

31. A compound according to Claim 27 wherein R2' is F; and R2 is H.
32. A compound according to Claim 27 wherein R2' is Et; and R2 is H.
33. A compound according to claim 1 wherein the compound is according to formula VIII. IX or X:
wherein
W is CR4; Z is CR4;
L2 ,R3 and R4 are as in claim 1; m, R4a, and R5 are as in claim 27; R2 is H or Me;
Cy is adamantyl, cyclohexyl or cycloheptyl; and R4b is independently selected
from C1-C4 alkyl and hydroxy;
or a pharmaceutically acceptable salt, solvate or prodrug thereof;
and stereoisomers, isotopic variants and tautomers thereof.
34. A compound according to either of claims 27 or 33 wherein m is 1, 2 or 3.
35. A compound according to either of claims 27 or 33 wherein m is 1.
36. A compound according to either of claims 27 or 33 wherein each R4a is independently selected from Me, Et, Ph, CI, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH=CH-C02H, SOMe, S02Me, SO3H, S03Me, and pyridyl.
37. A compound according to any one of claims 1-36 wherein L1 is a C1-C5 alkylene group.
38. A compound according to any one of claims 1-36 wherein L1 is C1-C5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, oxo, aryl, hydroxyl, and hydroxyalkyl
39. A compound according to any one of claims 1-36 wherein L1 is a C1-C5 alkylene group substituted with two alkyl groups and wherein any two alkyl groups on the same carbon atom can join together to form a cycloalkyl or cycloheteroalkyl ring
of 3-7 atoms.
40. A compound according to any one of claims 1-39 wherein R3 is selected from hydroxyl, amino, alkylamino or carbamoyl.
41. A compound according to any one of claims 1-36 wherein L1 is CH2 and Rl is substituted or unsubstituted aryl or heteroaryl.
-82-

42.

A compound according to any one of claims 1-36 wherein Lt is CH2 and R1 is phenyl or pyridyl substituted with one or more substituents independently selected from halo, hydroxyl, amino, cyano, sulfo, sulfanyl, sulfinyl, amido, carboxy, ester, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, and sulfonamide.



43.

A compound according to any one of claims 1-36 wherein L1 is CH2 and R1 is phenyl or pyridyl substituted with one or more substituents independently selected from Me, Et, Ph, C1, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH=CH-CO2H, SOMe, S02Me, S03H, S03Me, and pyridyl.



44.

A compound according to any one of claims 1-36 wherein R3 is a hydrogen bond donor group.



45.

A compound according to any one of claims 1-36 wherein R3 is selected from hydroxyl, amino, alkylamino or carbamoyl.

46. A compound according to any one of claims 1-36 wherein the group -LrR is selected from

47. A compound according to any one of claims 1-36 wherein the group -L(-R is selected from


48. A compound according to any one of claims 1-36 wherein the group -L1-R3 is selected from



v
50. A compound according to any one of claims 1-48 wherein R is H.
51. A compound according to claim 1 wherein the compound is according to formula XIa, Xlb, XIc, Xld, Xle, Xlf, Xlg, Xlh or XIj:

wherein m and R are as in claim 27; and R5 is H, alkyl, cycloalkyl or halo.
52. A compound according to claim 1 wherein the compound is according to formula XlIa,XIIb,orXIc:
-84-


wherein m and R4a are as in claim 27; R5 is H, alkyl, cycloalkyl or halo; and R2d is selected from hydrogen, alkyl, hydroxyalkyl, and substituted or unsubstituted phenyl.
Xllla , xillb
53. A compound according to claim 52 wherein R2d is methyl, i-Pr or hydroxymethyl.
54. A compound according to claim 52 wherein R2d is phenyl.
55. A compound according to claim 1 wherein the compound is according to formula XIIa, XIIIb, XIIIc, or Xllld:


wherein m and R4a are as in claim 27; R5 is H, alkyl, cycloalkyl or halo.
56. A compound according to any one of claims 51-55 wherein m is 1, 2 or 3.
57. A compound according to any one of claims 51 -55 wherein m is 1 or 2.
58. A compound according to any one of claims 51-55 wherem m is 2.
59. A compound according to any one of claims 56-58 wherein each R4a is independently selected from Me, Et, Ph, C1, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH=CH-C02H, SOMe, S02Me, S03H, S03Me, and pyridyl.
60. A compound according to any one of claims 51-55 wherein m is 1 and R4a is CF3.
61. A compound according to any one of claims 51-55 wherein m is 2 and R4a is F and CF3.
-85-

62. A compound according to any one of claims 51-55 wherein m is 2 and R a is F and C1.
63. A compound according to any one of claims 1-18 wherein W is CR4.
64. A compound according to any one of claims 1'50 wherein each of W and Z is independently CH.
65. A compound according to any one of claims 1-18 wherein W is N.
66. A compound according to any one of claims 1-18 wherein W is N and Z is CH.
67. A compound according to any one of claims 19-68 wherein R5 is H.
68. A compound according to any one of claims 19-68 wherein R5 is Me, cyclopropyl, CI, F, or CF3.
69. A compound according to claim 1 wherein the compound is selected from compounds listed in Table 1.
70. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound of any of the preceding claims.
71. The pharmaceutical composition of claim 70, wherein the carrier is a parenteral carrier.
72. The pharmaceutical composition of claim 70, wherein the carrier is an oral carrier.
73. The pharmaceutical composition of claim 70, wherein the carrier is a topical carrier.
74. A method for preventing, treating or ameliorating in a mammal a disease or condition that is causally related to the aberrant activity of the P2X7 receptor in vivo, which comprises administering to the mammal an effective disease-treating or condition-treating amount of a compound according to any of claims 1-69, or a pharmaceutical composition according to claim 70.
75. The method of claim 74, wherein the disease or condition is a pain condition.
76. The method of claim 74, wherein the disease or condition is an autoimmune disease.-86-

77. The method of claim 74, wherein the disease or condition is an inflammatory disease or condition.
78. The method of claim 74, wherein the disease or condition is a neurological or neurodegenerative disease or condition.
79. A method for preventing, treating or ameliorating in a mammal a disease or condition selected from: pain including acute, inflammatory and neuropathic pain, chronic pain, dental pain and headache including migraine, cluster headache and tension headache, Parkinson's disease, multiple sclerosis; diseases and disorders which are mediated by or result in neuroinflammation, traumatic brain injury and encephalitis; centrally-mediated neuropsychiatric diseases and disorders, depression mania, bipolar disease, anxiety, schizophrenia, eating disorders, sleep disorders and cognition disorders; epilepsy and seizure disorders; prostate, bladder and bowel dysfunction, urinary incontinence, urinary hesitancy, rectal hypersensitivity, fecal incontinence, benign prostatic hypertrophy and inflammatory bowel disease; respiratory and airway disease and disorders, allergic rhinitis, asthma and reactive airway disease and chronic obstructive pulmonary disease; diseases and disorders which are mediated by or result in inflammation, arthritis, rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus, psoriasis; obesity; lipid disorders; cancer; blood pressure; spinal cord injury; and renal disorders which comprises administering to the mammal an effective disease-treating or condition-treating amount of a compound according to any of claims 1-69, or a pharmaceutical composition of claim 70.
80. The method of claim 79, wherein the disease or condition is Parkinson's disease.
81. The method of claim 79, wherein the disease or condition is rheumatoid arthritis.
82. The method of claim 79, wherein the disease or condition is traumatic brain injury.
83. The method of claim 79, wherein the disease or condition is osteoarthritis.
84. The method of claim 79, wherein the disease or condition is pain.
85. The method of claim 79, wherein the disease or condition is neuropathic pain.
86. A method of treating a mammal suffering from at least one symptom selected from the group consisting of symptoms of exposure to capsaicin, symptoms of burns or irritation due to exposure to heat, symptoms of burns or irritation due to exposure to light, symptoms of burns, bronchoconstriction or irritation due to exposure to tear gas, and symptoms of burns or irritation due to exposure to acid, which comprises administering to the mammal an effective disease-treating or
-87-

condition-treating amount of a compound according to any of claims 1-69, or a pharmaceutical composition of claim 70.
The method of claim 85, wherein the pain is associated with a condition selected from the group consisting of postmastectomy pain syndrome, stump pain, phantom limb pain, oral neuropathic pain, Charcot's pain, toothache, venomous snake bite, spider bite, insect sting, postherpetic neuralgia, diabetic neuropathy, reflex sympathetic dystrophy, trigeminal neuralgia, osteoarthritis, rheumatoid arthritis, fibromyalgis, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, bilateral peripheral neuropathy, causalgia, sciatic neuritis, peripheral neuritis, polyneuritis, segmental neuritis, Gombault's neuritis, neuronitis, cervicobrachial neuralgia, cranial neuralgia, egniculate neuralgia, glossopharyngial neuralgia, migranous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, mandibular joint neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, red neuralgia, Sluder's neuralgia splenopalatine neuralgia, supraorbital neuralgia, vidian neuralgia, sinus headache, tension headache, labor, childbirth, intestinal gas, menstruation, cancer, and trauma.
A compound according to any one of claims 1-69, for use as a pharmaceutical.
A compound according to any one of claims 1-69, for use as a pharmaceutical in the treatment or prevention of a disease or condition selected from: pain including acute, inflammatory and neuropathic pain, chronic pain, dental pain and headache including migraine, cluster headache and tension headache, Parkinson's disease, multiple sclerosis; diseases and disorders which are mediated by or result in neuroinflammation, traumatic brain injury, encephalitis; centrally-mediated neuropsychiatric diseases and disorders, depression mania, bipolar disease, anxiety, schizophrenia, eating disorders, sleep disorders and cognition disorders; epilepsy and seizure disorders; prostate, bladder and bowel dysfunction, urinary incontinence, urinary hesitancy, rectal hypersensitivity, fecal incontinence, benign prostatic hypertrophy and inflammatory bowel disease; respiratory and airway disease and disorders, allergic rhinitis, asthma and reactive airway disease and chronic obstructive pulmonary disease; diseases and disorders which are mediated by or result in inflammation, arthritis, rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus, psoriasis; obesity; lipid disorders; cancer; blood pressure; spinal cord injury; and renal disorders.
Use of a compound according to any of claims 1-69, in the manufacture of a medicament for the treatment or prevention of a disease or condition selected from: pain including acute, inflammatory and neuropathic pain, chronic pain, dental pain and headache including migraine, cluster headache and tension headache, Parkinson's disease, and multiple sclerosis; diseases and disorders which are mediated by or result in neuroinflammation, traumatic brain injury, and encephalitis; centrally-mediated neuropsychiatric diseases and disorders, depression mania, bipolar disease, anxiety, schizophrenia, eating disorders, sleep
-88-

disorders and cognition disorders; prostate, bladder and bowel dysfunction, urinary incontinence, urinary hesitancy, rectal hypersensitivity, fecal incontinence, benign prostatic hypertrophy and inflammatory bowel disease; respiratory and airway disease and disorders, allergic rhinitis, asthma and reactive airway disease and chronic obstructive pulmonary disease; diseases and disorders which are mediated by or Tesult in inflammation, arthritis, rheumatoid arthritis and osteoarthritis, myocardial infarction, various autoimmune diseases and disorders, uveitis and atherosclerosis; itch / pruritus, psoriasis; obesity; lipid disorders; cancer; blood pressure; spinal cord injuryconditions resulting from or related to immune dysfunction; and renal disorders.
Dated this 16th day of September, 2008

HIRAL CHANDRAKANT JOSHI
AGENT FOR
RENOVIS, INC.




























89-

Documents:

1976-MUMNP-2008-ABSTRACT(GRANTED)-(28-2-2012).pdf

1976-mumnp-2008-abstract.doc

1976-mumnp-2008-abstract.pdf

1976-MUMNP-2008-CANCELLED PAGES(24-11-2011).pdf

1976-MUMNP-2008-CLAIMS(AMENDED)-(24-11-2011).pdf

1976-MUMNP-2008-CLAIMS(GRANTED)-(28-2-2012).pdf

1976-mumnp-2008-claims.doc

1976-mumnp-2008-claims.pdf

1976-mumnp-2008-correspondence(16-9-2008).pdf

1976-MUMNP-2008-CORRESPONDENCE(22-12-2009).pdf

1976-MUMNP-2008-CORRESPONDENCE(25-8-2009).pdf

1976-MUMNP-2008-CORRESPONDENCE(5-3-2012).pdf

1976-MUMNP-2008-CORRESPONDENCE(IPO)-(29-2-2012).pdf

1976-mumnp-2008-description(complete).doc

1976-mumnp-2008-description(complete).pdf

1976-MUMNP-2008-DESCRIPTION(GRANTED)-(28-2-2012).pdf

1976-MUMNP-2008-FORM 1(24-11-2011).pdf

1976-mumnp-2008-form 1.pdf

1976-mumnp-2008-form 18.pdf

1976-MUMNP-2008-FORM 2(GRANTED)-(28-2-2012).pdf

1976-MUMNP-2008-FORM 2(TITLE PAGE)-(GRANTED)-(28-2-2012).pdf

1976-mumnp-2008-form 2(title page).pdf

1976-mumnp-2008-form 2.doc

1976-mumnp-2008-form 2.pdf

1976-MUMNP-2008-FORM 26(5-3-2012).pdf

1976-MUMNP-2008-FORM 3(24-11-2011).pdf

1976-mumnp-2008-form 3.pdf

1976-mumnp-2008-form 5.pdf

1976-MUMNP-2008-FORM PCT-IPEA-409(24-11-2011).pdf

1976-MUMNP-2008-GENERAL POWER OF ATTORNEY(22-12-2009).pdf

1976-mumnp-2008-pct-isa-210.pdf

1976-MUMNP-2008-PETITION UNDER RULE 137(24-11-2011).pdf

1976-MUMNP-2008-PETITION UNDER RULE 137-(24-11-2011).pdf

1976-MUMNP-2008-REPLY TO EXAMINATION REPORT(24-11-2011).pdf

1976-mumnp-2008-wo international publication report a2.pdf

abstract1.jpg


Patent Number 251162
Indian Patent Application Number 1976/MUMNP/2008
PG Journal Number 09/2012
Publication Date 02-Mar-2012
Grant Date 28-Feb-2012
Date of Filing 16-Sep-2008
Name of Patentee RENOVIS, INC.
Applicant Address 2 CORPORATE DRIVE, SOUTH SAN FRANCISCO, CA 94080
Inventors:
# Inventor's Name Inventor's Address
1 KELLY, MICHAEL, G. 790 SAN DOVAL PLACE, THOUSAND OAKS, CA 91360
2 KINCAID, JOHN 2201 BRIDGEPOINTE PKWY. SAN MATEO, CA 94404
3 GOWLUGARI, SUMITHRA 2021 S DELEWARE ST., APT #2, SAN MATEO CA 94403
4 FANG, YUNFENG 12029 WORLD TRADE DRIVE #4, SAN DIEGO, CA 92128,
5 CAO, YEYU 1131 COMPASS LANE #110, FOSTER CITY, CA 94404
6 KAUB, CARL 120 WEST THIRD AVENUE, SAN MATEO, CA 94404
PCT International Classification Number A61K31/4704
PCT International Application Number PCT/US2007/006685
PCT International Filing date 2007-03-16
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/782,776 2006-03-16 U.S.A.
2 60/831,416 2006-07-17 U.S.A.
3 60/846,993 2006-09-25 U.S.A.
4 60/783,121 2006-03-16 U.S.A.
5 60/918,086 2007-03-15 U.S.A.