Biclycloheteroaryl Compounds as P2x7 Modulators and Uses Thereof

Information

  • Patent Application
  • 20100298285
  • Publication Number
    20100298285
  • Date Filed
    March 16, 2007
    17 years ago
  • Date Published
    November 25, 2010
    13 years ago
Abstract
Bicycloheteroaryl compounds are disclosed that have a formula represented by the following: 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.
Description
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, cardiovascular 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 and 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 (P2X1-P2X6) 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 (Buisman 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′-O-(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 rP2X7 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 (1-2s) 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-terminal 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 (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 WO99/29661 disclose that certain adamantane derivatives exhibit P2X7 antagonistic activity having therapeutic efficacy in the treatment of rheumatoid arthritis and psoriasis. Similarly, WO99/29686 discloses that certain heterocyclic derivatives are P2X7 receptor antagonists and are useful as immunosuppressive agents and treating rheumatoid arthritis, asthma, septic shock and atherosclerosis. 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

Bicycloarylderivatives of formulae I-XIIIe, 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, 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 a 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 (I):









    • wherein A is CR2aR2b; 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 Z can not be N at the same time;

    • L1 is CO, SO, SO2, or substituted or unsubstituted C1-C5 alkylene;

    • n is 0, 1, 2, 3 or 4;

    • R1 is selected from substituted or unsubstituted aryl and substituted or unsubstituted heteroaryl;

    • each of R2a, R2b, R2′ and R2″ is independently selected from hydrogen, substituted or unsubstituted C1-C6 alkyl and halo; or any of R2′ and R2″ can join together to form a cycloalkyl or cycloheteroalkyl ring of 3-7 atoms;

    • R3 is selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted or unsubstituted bicycloheteroaryl, substituted or unsubstituted alkoxy; cycloalkylalkoxy, cycloheteroalkyloxy, aryloxy, heteroaryloxy, arylalkyl, and heteroarylalkyl;

    • each R4 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;

    • 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 formula I, n is 0-3.


In a further embodiment, with respect to compounds of formula I, L1 is a C1-C5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, hydroxy, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, halogen, and oxo.


In a further embodiment, with respect to compounds of formula I, L1 is a C1-C5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, hydroxy, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, halogen, and oxo.


In a further embodiment, with respect to compounds of formula I, L1 is CO.


In a further embodiment, with respect to compounds of formula I, L1 is SO2.


In a further embodiment, with respect to compounds of formula I, L1 is 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, L1 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 R3 is —NH—.


In a further embodiment, with respect to compounds of formula I, L1 is a bond, a C1-C5 alkylene group; and R3 is a heterocycloalkyl group containing —NH—.


In a further embodiment, with respect to compounds of formula I, A is CR2aR2b.


In a further embodiment, with respect to compounds of formula I, A is CH2.


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.


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 R2′and R2″ of the







group may be selected from Me, Et, halo and Cl, 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, 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 alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl and halo. In one particular embodiment, R5 is selected from Me, cyclopropyl, Cl, F and CF3.


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 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, C1-C6 alkoxy, aryl and di-C1-C6alkylamino. It should be further understood that the terms “groups” and “radicals” can be considered interchangeable when used herein.


“Acyl” refers to a radical —C(O)R20, where R20 is hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl as defined herein. Representative examples include, but are not limited to, formyl, acetyl, cylcohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl, benzylcarbonyl and the like.


“Acylamino” refers to a radical —NR21C(O)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, cyclohexylcarbonylamino, cyclohexylmethyl-carbonylamino, benzoylamino, benzylcarbonylamino and the like.


“Acyloxy” refers to the group —OC(O)R23 where R23 is hydrogen, alkyl, aryl or cycloalkyl.


“Substituted alkenyl” includes those groups recited in the 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 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(O)2— and aryl-S(O)2.


“Alkoxy” refers to the group —OR24 where R24 is alkyl. Particular alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, 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(O)2— and aryl-S(O)2—.


“Alkoxycarbonylamino” refers to the group —NR25C(O)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, n-butyl, iso-butyl, tert-butyl, n-hexyl, n-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.


“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, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)—, aryl-S(O)—, alkyl-S(O)2—, and aryl-S(O)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 to 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(O)2— and aryl-S(O)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 olefinic 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, allcoxycarbonylamino, 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(O)2— and aryl-S(O)2—.


“Alkanoyl” or “acyl” as used herein refers to the group R27—C(O)—, 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, hexylene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octal ene, 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(O)2— and aryl-S(O)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 R39 and R31 are independently selected from hydrogen, aryl and heteroaryl.


“Alkoxyamino” refers to a radical —N(H)OR32 where R32 represents an alkyl or cycloalkyl group as defined herein.


“Alkoxycarbonyl” refers to a radical —C(O)-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(O)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(O)R35 where R35 is an alkyl or cycloalkyl group as defined herein. Representative examples include, but are not limited to, methylsulfinyl, ethylsulfinyl, propylsulfinyl, butylsulfinyl 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.


“Aminocarbonyl” refers to the group —C(O)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(O)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(O)NR39R39 where each R39 is independently hydrogen, alkyl, aryl or cycloalkyl, 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(O)—O-aryl where aryl is as defined herein.


“Arylsulfonyl” refers to a radical —S(O)2R41 where R41 is an aryl or heteroaryl group as defined herein.


“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, benzomorphan, 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(O)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(O)OH.


“Carboxyamino” refers to the radical —N(H)C(O)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(O)2— and aryl-S(O)2—.


“Cycloalkoxy” refers to the group —OR43 where R43 is cycloalkyl. Such cycloalkoxy groups include, by way of example, cyclopentoxy, cyclohexoxy and the like.


“Cycloalkenyl” refers 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 olefinic 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(O)2— and aryl-S(O)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 olefinic 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 —NR44R45 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 —NO2.


“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, —O, ═O, —OR46, —SR46, —S, ═S, —NR46R47, —NR46, —CX3, —CF3, —CN, —OCN, —SCN, —NO, —NO2, ═N2, —N3, —S(O)2O, —S(O)2OH, —S(O)2R46, —OS(O2)O, —OS(O)2R46, —P(O)(O)2, —P(O)(OR46)(O)(OR46)(OR47), —C(O)R46, —C(S)R46, —C(O)OR46, —C(O)NR46R47, —C(O)O, —C(S)OR46, —NR48C(O)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)R50 or —S(O)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, NR54SO2R57, COOalkyl, COOaryl, CONR54R55, CONR54OR55, NR54R55, SO2NR54R55, S-alkyl, S-alkyl, SOalkyl, SO2alkyl, Saryl, SOaryl, SO2aryl; 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 hctcroaromatic 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, β-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochrotnene, 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 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.


Examples of representative heteroaryls include the following:







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:







wherein each X is selected from CR582, NR58, O and S; and each Y is selected from NR58, O and S; and R58 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(O)2— and aryl-S(O)2—. Substituting groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea derivatives.


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, O 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:







wherein each X is selected from C—R582 NR58, 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,


—NO2, —NH2, —NHR59, —N(R59)2,


—NRCOR, —NR59SOR59, —NR59SO2R59, OH, CN,


—CO2H,


—R59—OH, —O—R59, —COOR59,


—CON(R59)2, —CONROR59,


—SO3H, —R59—S, —SO2N(R59)2,


—S(O)R59, —S(O)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 R59a is alkyl, cycloalkyl, acyl, aryl, or heteroaryl.


“Dihydroxyphosphoryl” refers to the radical —PO(OH)2.


“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(O)2— and aryl-S(O)2—.


“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(O2)—. “Substituted sulfonyl” refers to a radical such as R61—(O2)S— wherein R61 is any substituent described herein. “Aminosulfonyl” or “Sulfonamide” refers to the radical H2N(O2)S—, and “substituted aminosulfonyl” or “substituted sulfonamide” refers to a radical such as R622N(O2)S— wherein each R62 is independently any substituent described herein.


“Sulfone” refers to the group —SO2R63. 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.


“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-methylbicyclo[2.2.2]-oct-2-ene-1-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, tetraalkylammonium, 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, tetraalkylammonium 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, 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-C12 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 (13C), 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 13C, or any nitrogen may be 15N, 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 and ready means of detection. Further, compounds may be prepared that are substituted with positron emitting isotopes, such as 11C, 18F, 15O and 13N, 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 their 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 levorotatory (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 π 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 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; 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 Z can not be N at the same time;

    • L1 is CO, SO, SO2, substituted or unsubstituted C1-Cs alkylene;

    • n is 0, 1, 2, 3 or 4;

    • R1 is selected from substituted or unsubstituted aryl and substituted or unsubstituted heteroaryl;

    • each of R2a, R2b, R2′ and R2″ is independently selected from hydrogen, substituted or unsubstituted C1-C6 alkyl and halo; or any of R2′ and R2″ can join together to form a cycloalkyl or cycloheteroalkyl ring of 3-7 atoms;

    • R3 is selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted or unsubstituted bicycloheteroaryl, substituted or unsubstituted alkoxy; cycloalkylalkoxy, cycloheteroalkyloxy, aryloxy, heteroaryloxy, aryl alkyl, and heteroarylalkyl;

    • each R4 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, carboxyl, 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 formula I, n is 0-4.


In a further embodiment, with respect to compounds of formula I, L1 is a C1-C5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, hydroxy, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, halogen, and oxo.


In a further embodiment, with respect to compounds of formula I, L1 is a C1-C5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, hydroxy, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, halogen, and oxo.


In a further embodiment, with respect to compounds of formula I, L1 is CO.


In a further embodiment, with respect to compounds of formula I, L1 is SO2.


In a further embodiment, with respect to compounds of formula I, A is CR2aR2b.


In a further embodiment, with respect to compounds of formula I, A is CH2.


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.


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 R2′ and R2″ of the







group may be selected from Me, Et, halo and Cl, 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 or unsubstituted 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 one particular embodiment, R1 is substituted or unsubstituted pyridyl.


In a further embodiment, with respect to compounds of formula I, R1 is substituted or unsubstituted heteroaryl. In one particular embodiment, R1 is substituted or unsubstituted quinoline, substituted or unsubstituted benzodioxole, substituted or unsubstituted benzodioxane, substituted or unsubstituted benzofuran, substituted or unsubstituted benzothiophene, or substituted or unsubstituted benzodioxepine.


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 alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl and halo. In one particular embodiment, R5 is selected from Me, cyclopropyl, Cl, F and CF3.


In another embodiment, with respect to compounds of formula I, the compound is the compound is according to formula II, III or IV:









    • wherein

    • W is CR4; Z is CR4;

    • L1, 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″ is H.


In another embodiment, with respect to compounds of formulae II-IV, R2′ is halo; and R2″ is H.


In another embodiment, with respect to compounds of formulae II-IV, R2′ is Cl or F; and R2″ is H.


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 R1 is substituted or unsubstituted naphthalene.


In another embodiment, with respect to compounds of formulae II-IV, each of R1 is unsubstituted naphthalene.


In another embodiment, with respect to compounds of formulae II-IV, each of R1 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, or substituted or unsubstituted benzodioxepine.


In another embodiment, with respect to compounds of formula I, the compound is 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 described for formula I;

    • 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 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 pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof.





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 Cl or F; and R2″ is H.


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, the compound is the compound is according to formula VIII, IX, or X:







and wherein L1, R2′ R3, W, and Z are as described for formula I; and wherein (R4a)m and R5 are as described for formulae V-VII.


In one embodiment, with respect to compounds of formulae V-X, each of W and Z is independently CR4. In another embodiment each of W and Z is CH.


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 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, Cl, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH═CH—CO2H, SOMe, SO2Me, SO3H, SO3Me, and pyridyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is a C1-C5 alkylene group.


In another embodiment, with respect to compounds of formulae I-X, L1 is a C1-C5 alkylene group unsubstituted or substituted by one or more substituents selected from alkyl, hydroxyl, oxo and hydroxyalkyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is an ethylene group unsubstituted or substituted sby one or more substituents selected from Me, Et, i-Pr, hydroxy, and hydroxymethyl.


In another embodiment, with respect to compounds of formulae L1 is a methylene group unsubstituted or substituted by one or more substituents selected from Me, Et, i-Pr, and hydroxymethyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is CO and R3 is alkyl, or substituted alkyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is a CO and R3 is methyl, ethyl, propyl, iso-propyl, t-butyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxy iso-propyl, or hydroxy t-butyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is a CO and R3 is cyclopropylmethyl, cyclopropylethyl, cyclopropylpropyl, cyclobutylmethyl, cyclobutylethyl, cyclobutylpropyl, cyclopentylmethyl, cyclopentylethyl, cyclopentylpropyl, cyclohexylmethyl, cyclohexylethyl, cyclohexylpropyl, cycloheptylmethyl, cycloheptylethyl, cycloheptylpropyl, phenylmethyl, phenethyl, or phenylpropyl, unsubstituted or substituted with alkyl or hydroxy.


In another embodiment, with respect to compounds of formulae I-X, L1 is a CO and R3 is substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted or unsubstituted bicycloheteroaryl.


In another embodiment, with respect to compounds of formulae I-X, L1 is a CO and R3 is substituted or unsubstituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, phenyl, naphthyl, pyridyl, pyrrolidinyl, benzopyranyl, benzodioxanyl, or benzofuranyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is a CO and R3 is phenyl 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.


In another embodiment, with respect to compounds of formulae I-X, L1 is a CO and R3 is benzyl 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.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is alkyl, or substituted alkyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is methyl, ethyl, propyl, iso-propyl, t-butyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxy iso-propyl, or hydroxy t-butyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is cyclopropylmethyl, cyclopropylethyl, cyclopropylpropyl, cyclobutylmethyl, cyclobutylethyl, cyclobutylpropyl, cyclopentylmethyl, cyclopentylethyl, cyclopentylpropyl, cyclohexylmethyl, cyclohexylethyl, cyclohexylpropyl, cycloheptylmethyl, cycloheptylethyl, cycloheptylpropyl, phenylmethyl, phenethyl, or phenylpropyl, which can be optionally substituted with alkyl or hydroxy.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted or unsubstituted bicycloheteroaryl.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is substituted or unsubstituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, phenyl, naphthyl, pyridyl, pyrrolidinyl, benzopyranyl, benzodioxanyl, or benzofuranyl.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is phenyl 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.


In another embodiment, with respect to compounds of formulae I-X, L1 is SO2 and R3 is benzyl 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.


In another embodiment, with respect to compounds of formulae I-X, the group -L1-R3 is selected from







In a particular embodiment, with respect to compounds of formulae I-X, the group -L1-R3 is







In another embodiment, with respect to compounds of formulae I-X, the group -L1-R3 is selected from







In a particular embodiment, with respect to compounds of formulae I-X, R2′ is Me.


In a particular embodiment, with respect to compounds of formulae I-X, R2′ is H.


In another embodiment, with respect to compounds of formula I, the compound is according to formula XIa, XIb, XIc, XId, XIe, XIf, XIg, XIh, or XIj:










and R5 and (R4a)m are as described for formulae V-VII.


In another embodiment, with respect to compounds of formula I, the compound is according to formula XIIa, XIIb, or XIc:







and wherein R5 and (R4a)m are as described for formulae V-VII; 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.


In another embodiment, with respect to compounds of formula I, the compound is according to formula XIIIa, XIIIb, XIIIc, XIIId, or XIIIe:







and wherein R5 and (R4a)m are as described for formulae V-VII.


In one embodiment, with respect to compounds of formulae XIa-XIIIe, m is 1, 2 or 3.


In another embodiment, with respect to compounds of formulae XIa-XIIIe, m is 1 or 2. In a particular embodiment m is 2.


In another embodiment, with respect to compounds of XIa-XIIIe, each of R4a is independently selected from Me, Et, Ph, Cl, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH═CH—CO2H, SOMe, SO2Me, SO3H, SO3Me, and pyridyl.


In another embodiment, with respect to compounds of V-XIIIe, m is 1 and R″ is CF3.


In another embodiment, with respect to compounds of V-XIIIe, m is 2 and R4a is F and CF3.


In another embodiment, with respect to compounds of V-XIIIe, m is 2 and R4a is F and Cl.


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.


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-XIIIe, R5 is H.


In one embodiment, with respect to compounds of formulae II-XIIIe, R5 is selected from alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl and halo. In one particular embodiment, R5 is selected from Me, cyclopropyl, Cl, F and CF3.


In one embodiment, with respect to compounds of formulae II-XIIIe, R5 is Me.


In one embodiment, with respect to compounds of formulae II-XIIIe, R5 is CF3.


In one embodiment, with respect to compounds of formulae II-XIIIe, R5 is F.


In a further embodiment with respect to compounds of formulae II-XIIIe, R5 is Cl.


In a further embodiment with respect to compounds of formulae II-XIIIe, 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 pharmaceutically 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 C1 to C8 alkyl, C2-C8 alkenyl, aryl, C7-C12 substituted aryl, and C7-C12 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 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 corn 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 paraffinic 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 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, Pa., 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.


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 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 or 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.


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 methods are presented with details as to the preparation of representative bicycloheteroaryls that have been listed hereinabove. 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.


Synthesis of Intermediates
Intermediate 1
5-Amino-6-methoxy-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester











To a stirring solution of p-anisaldehyde (SM) (20.00 g, 146.89 mmol) in 100 mL of toluene equipped with a Dean-Stark apparatus and a reflux condenser was added aminoacetaldehyde-dimethylacetal (16 mL, 146.89 mmol). The resulting solution was refluxed for 16 h and the solvent was evaporated, the residue was diluted with toluene and evaporated to give a oily residue. The oily residue was dissolved in dry THF (100 mL) and cooled to −10° C. To this cooled solution was added ethyl chloroformate (14.04 mL, 146.89 mmol), and the resultant solution was stirred for 10 min at this temperature and allowed to warm to room temperature. To this mixture was added trimethylphosphite (20.79 mL, 176.26 mmol) and the resultant mixture was allowed to stir at room temperature for 16 h. The solvent along with unreacted trimethyphosphite in the reaction mixture was evaporated by adding toluene. The resultant oily residue was dissolved in DCE (100 mL) and titanium tetrachloride (96.64 mL, 881.34 mmol) was added and refluxed at 100° C. for 16 h. The solvent and titanium tetrachloride were distilled under reduced pressure to leave a residue of crude compound. The crude compound residue was treated with ice-cold water, basified and then extracted with DCM. The DCM layer was washed with brine, dried over Na2SO4 and concentrated to afford the title compound in 64.6% yield. 1H NMR (300 MHz, CDCl3): δ 3.95 (s, 3H), 7.06 (s, 1H), 7.21-7.26 (m, 1H), 7.54 (d, J=5.9 Hz, 1H), 7.85 (d, J=8.7 Hz, 1H), 8.44 (d, J=5.5 Hz, 1H), 9.10 (s, 1H); MS: 160 (M++1).


b. 6-Methoxy-5-nitro-isoquinoline (RN036_Int02A)

To an ice-cold solution of RN036_Int01A (15.00 g, 94.33 mmol) in conc. H2SO4 (200 mL) was added KNO3 (11.43 g, 113.20 mmol) in portions. The reaction mixture was allowed to warm to room temperature and stirred for 1 h. After completion of the reaction (TLC), the mixture was cooled to 0° C., diluted with ice-cold water and basified with ice-cold NaOH solution. The resulting mixture was extracted with EtOAc, the combine organic layers were washed with brine, dried over anhy. Na2SO4 and concentrated to leave a residue of title compound as yellow solid in 82.6% yield. 1H NMR (300 MHz, CDCl3): δ 4.10 (s, 3H), 7.47 (d, J=9.1 Hz, 1H), 7.55 (d, J=5.9 Hz, 1H), 8.15 (d, J=9.1 Hz, 1H), 8.60 (d, J=6.1 Hz, 1H), 9.23 (s, 1H); MS: 205 (M++1).


c. 6-Methoxy-5-nitro-1,2,3,4-tetrahydro-isoquinoline (RN036_Int03A)

To an ice-cold solution of RN036_Int02A (16.00 g, 78.40 mmol) in acetic acid (320 mL) was added NaBH4 (14.83 g, 392.15 mmol) in small portions. The resultant reaction mixture was stirred at 0° C. for 15 min after complete additions. After completion of the reaction the reaction mixture was diluted with ice-cold water and basified with aq. NaOH and extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4 and the solvent was evaporated under reduced pressure to afford a dark brown oil of the title compound in 80% yield. 1H NMR (300 MHz, CDCl3): δ 2.67 (t, J=5.9 Hz, 2H), 3.10 (t, J=5.9 Hz, 2H), 3.85 (s, 3H), 3.96 (s, 2H), 6.84 (d, J=8.7 Hz, 1H), 7.08 (d, J=8.7 Hz, 1H); MS: 209 (M++1).


d. 6-Methoxy-5-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (RN036_Int04A)

To a stirring solution of RN036_Int03A (13.10 g, 62.98 mmol) in DCM (60 mL) was added aq. NaOH (12.50 g, 318.05 mmol) and Boc2O (28.48 mL, 125.96 mmol) and the resultant reaction mixture was stirred for overnight. After completion of the reaction the organic layer was separated and the aq. layer was extracted with DCM, the combined organic layers were washed with brine, dried over Na2SO4 and the solvent was evaporated to leave a residue of crude compound. The crude compound was purified by column chromatography employing EtOAc:Hex (10:90) as eluent to afford the title compound in 75% yield. 1H NMR (300 MHz, CDCl3): δ 1.48 (s, 9H), 2.74 (t, J=5.8 Hz, 2H), 3.63 (t, J=6.0 Hz, 2H), 3.88 (s, 3H), 4.54 (s, 2H), 6.91 (d, J=8.7 Hz, 1H), 7.18 (d, J=8.2 Hz, 1H).


e. 5-Amino-6-methoxy-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (RN036_A)

To a cooled solution of RN036_Int04A (14.5 g, 47.20 mmol) in MeOH (500 mL) was added Pd/C (4.00 g) and the resultant reaction mixture was subjected to reduction in the presence of hydrogen pressure (60 psi) in a Parr apparatus for 2 h. After completion of the reaction Pd/C was filtered using a celite pad. The filtrate was concentrated to afford the title compound in a 95% yield. 1H NMR (300 MHz, CDCl3): δ 1.47 (s, 9H), 2.57 (t, J=5.9 Hz, 2H), 3.68 (t, J=5.9 Hz, 2H), 3.75 (br, 2H), 3.84 (s, 3H), 4.50 (s, 2H), 6.52 (d, J=8.1 Hz, 1H), 6.70 (d, J=8.1 Hz, 1H); LCMS: 223 (M++1−tbutyl).


Intermediate 2
5-Amino-6-cyclopropyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester











To a stirring solution of 4-bromobenzaldehyde (SM) (30.00 g, 162.16 mmol) in 150 mL of toluene equipped with a Dean-Stark apparatus and a reflux condenser was added aminoacetaldehyde-dimethylacetal (17.02 g, 162.16 mmol). The resulting solution was refluxed for 16 h and the solvent was evaporated, the residue was diluted with toluene and evaporated to give an oily residue. The oily residue was dissolved in dry THF (100 mL) and cooled to −10° C., to this cooled solution was added ethyl chlroformate (17.59 g, 162.16 mmol) and the resultant solution was stirred for 10 min at this temperature and allowed to warm to room temperature. To this mixture was added trimethylphosphite (24.12 g, 195.59 mmol) and the resultant mixture was allowed to stir at room temperature for 16 h. The solvent along with unreacted trimethyphosphite in the reaction mixture was evaporated by adding toluene. The resultant oily residue was dissolved in DCE (150 mL) and titanium tetrachloride (184.50 g, 972.90 mmol) was added and refluxed at 100° C. for 16 h. The solvent and titanium tetrachloride were distilled under reduced pressure to leave a residue of crude compound. The crude compound residue was treated with ice-cold water, basified and then extracted with DCM. The DCM layer was washed with brine, dried over Na2SO4 and concentrated to leave a residue of crude compound. The crude compound was purified by column chromatography employing EtOAc:Hex (15:85) as eluents to afford the title compound in 24.7% yield. 1H NMR (300 MHz, CDCl3): δ 7.57 (d, J=5.6 Hz, 1H), 7.70 (d, J=9.9 Hz, 1H), 7.85 (d, J=8.7 Hz, 1H), 8.01 (s, 1H), 8.56 (d, J=5.9 Hz, 1H), 9.23 (s, 1H). MS: 208, 210 (M++1).


b. 6-Bromo-5-nitro-isoquinoline (RN036_Int03D)

To an ice-cold solution of RN036_Int01D (14.00 g, 67.63 mmol) in Conc. H2SO4 (50 mL) was added KNO3 (8.19 g, 81.15 mmol) in portions. The reaction mixture was allowed to warm to room temperature and stirred for 1 h. After completion of the reaction (TLC) the reaction mixture was cooled to 0° C. and diluted with ice-cold water, basified with ice-cold NaOH solution. The resulting mixture was extracted with EtOAc, the combined organic layers were washed with brine, dried over anhy. Na2SO4 and concentrated to leave a residue of the title compound as a yellow solid in 87.4% yield. 1H NMR (300 MHz, CDCl3): δ 7.54 (d, J=5.9 Hz, 1H), 7.85 (d, J=8.7 Hz, 1H), 8.02 (d, J=8.7 Hz, 1H), 8.73 (d, J=6.2 Hz, 1H), 9.35 (s, 1H). MS: 253, 255 (M++1).


c. 6-Bromo-5-nitro-1,2,3,4-tetrahydro-isoquinoline (RN036_Int06D)

To an ice-cold solution of RN036_Int03D (14.90 g, 59.12 mmol) in acetic acid (100 mL) was added NaBH4 (8.94 g, 236.48 mmol) in small portions. The resultant reaction mixture was stirred at 0° C. for 15 min after complete additions. After completion of the reaction the reaction mixture was diluted with ice-cold water and basified with aq. NaOH and extracted with DCM. The combined organic layers were washed with brine, dried over Na2SO4 and the solvent was evaporated under reduced pressure to afford a dark brown oil of the compound in 78.6% yield. 1H NMR (300 MHz, CDCl3): δ 2.71 (t, J=5.7 Hz, 2H), 3.14 (t, J=5.8 Hz, 2H), 4.01 (s, 2H), 7.03 (d, J=8.2 Hz, 1H), 7.43 (d, J=8.2 Hz, 1H). MS: 257, 259 (M++1).


d. Tert-butyl 6-bromo-3,4-dihydro-5-nitroisoquinoline-2(1H)-carboxylate (RN036_Int07D)

To a stirring solution of RN036_Int06D (11.9 g, 46.48 mmol) in 1,4-dioxane (100 mL) was added aq. NaOH (13.94 g, 348.6 mmol) and Boc2O (16.01 g, 69.72 mmol) and the resultant reaction mixture was stirred for overnight. After completion of the reaction the reaction mixture was extracted with DCM the organic layer was washed with brine, dried over Na2SO4 and the solvent was evaporated to leave a residue of crude compound. The crude compound was purified by column chromatography employing EtOAc:Hex (10:90) as eluent to afford the title compound in 75.5% yield. 1H NMR (300 MHz, CDCl3): δ 1.52 (s, 9H), 2.79 (t, J=5.9 Hz, 2H), 3.69 (t, J=5.9 Hz, 2H), 4.61 (s, 2H), 7.15 (d, J=8.1 Hz, 1H), 7.53 (d, J=8.1 Hz, 1H). MS: 301, 303 (M+tbutyl+1).


e. 6-Cyclopropyl-5-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (RN036_Int08D)

Cyclopropylboronic acid (3.65 g, 42.48 mmol), K3PO4 (24.22 g, 135.30 mmol), tricyclohexylphosphine (0.92 g, 3.25 mmol), water (0.5 mL), Pd(OAc)2 (0.36 g, 1.62 mmol) were added successively to a stirring solution of RN036_Int07D (12.50 g, 35.11 mmol) in dry toluene (150 mL) under N2 atmosphere. The resultant reaction mixture was heated to 100° C. for 12 h. After completion of the reaction, the reaction was cooled to room temperature, diluted with water and extracted with EtOAc. The organic layer was dried over. Na2SO4 and concentrated to leave a residue of crude product. The crude product was purified by column chromatography employing EtOAc:Hex (5:95) as eluent to afford the desired product in 68% yield. 1H NMR (300 MHz, CDCl3): δ 0.66-0.71 (m, 2H), 0.94-1.00 (m, 2H), 1.49 (s, 9H), 1.87-1.90 (m, 1H), 2.74 (t, J=5.5 Hz, 2H), 3.65 (t, J=5.7 Hz, 2H), 4.58 (s, 2H), 6.94 (d, J=8.2 Hz, 1H), 7.14 (d, J=7.9 Hz, 1H). LCMS: 262 (M+tbutyl+1), 99.01%.


f. 5-Amino-6-cyclopronyl-3,4-dihydro-1H-isocquinoline-2-carboxylic acid tert-butyl ester (RN036-D)

To a Stirring Solution of RN036-Int08D (7.60 g, 23.90 mmol) in EtOH (30 mL) was added 1,4-dioxane (70 mL), Fe (6.67 g, 119.40 mmol) and NH4Cl (5.11 g, 95.60 mmol) in 20 mL of H2O. The resulting reaction mixture was heated to 80° C. for 1 h. After completion of the reaction, the reaction mixture was cooled to room temperature and basified by aq. NaOH. The resultant reaction mixture was concentrated to ¼th of the total volume and then extracted with DCM. The combine organic layers were washed with brine, dried over anhy. Na2SO4 and concentrated to afford the title compound in 69% yield. 1H NMR (300 MHz, CDCl3): δ 0.56-0.59 (m, 2H), 0.88-0.92 (m, 2H), 1.47 (s, 9H), 1.63-1.68 (m, 1H), 2.57 (t, J=5.7 Hz, 2H), 3.71 (t, J=5.9 Hz, 2H), 3.97 (br s, 2H), 4.52 (s, 2H), 6.50 (d, 7.8 Hz, 1H), 6.93 (d, J=7.8 Hz, 1H). LCMS: 232 (M+tbutyl+1), 98.71%.


Intermediate 3
5-Amino-3,4-dihydro-6-methyl-1H-isoquinoline-2-carboxylic acid tert-butyl ester











To a stirring solution of p-tolualdehyde (SM; 20.00 g, 166.6 mmol) in toluene (200 mL) equipped with a Dean-Stark apparatus and a reflux condenser was added aminoacetaldehyde-dimethylacetal (18.06 mL, 166.6 mmol) and the resulting mixture was heated to reflux. After 16 h the solvent was evaporated, the residue was diluted with toluene and evaporated to give an oily residue. The oily residue was dissolved in dry THF (200 mL) and cooled to −10° C. To this cooled solution was added ethyl chloroformate (15.78 mL, 166.6 mmol) and the resultant solution was stirred for 10 min at this temperature and allowed to warm to room temperature. At room temperature, trimethylphosphite (23.5 mL, 199.9 mmol) was added and the resultant mixture was stirred further. After 16 h, the mixture was concentrated and traces of solvent and unreacted trimethyphosphite were removed by co-evaporation with toluene. The resultant oily residue was dissolved in DCE (170 mL) and titanium tetrachloride (126.9 mL, 1166.6 mmol) was added and refluxed at 100° C. for 16 h. The solvent and titanium tetrachloride were distilled under reduced pressure to leave a residue of crude compound. The crude compound residue was treated with ice-cold water, basified and then extracted with DCM. The combined organic layer was washed with brine, dried over Na2SO4 and concentrated to afford the title compound in 67% yield. 1H NMR (300 MHz, CDCl3): δ 2.54 (s, 3H), 7.43 (d, J=8.5 Hz, 1H), 7.54-7.58 (m, 2H), 7.86 (d, J=8.5 Hz, 1H), 8.47 (d, J=5.5 Hz, 1H), 9.18 (s, 1H); MS: 144 [M+H]+.


b. 6-Methyl-5-nitro-isoquinoline (RN009_Int02)

To an ice-cold solution of RN009_Int01 (17.5 g, 122.37 mmol) in conc. H2SO4 (225 mL) was added KNO3 (14.8 g, 146.85 mmol) in portions. The reaction mixture was allowed to warm to room temperature and stirred for 1 h. After completion of the reaction (TLC), the mixture was cooled to 0° C., diluted with ice-cold water and basified with ice-cold NaOH solution. The resulting mixture was extracted with EtOAc, the combined organic layer was washed with brine, dried over anhy. Na2SO4 and concentrated to afford the title compound in 78% yield. 1H NMR (300 MHz, CDCl3): δ 2.58 (s, 3H), 7.52 (d, J=8.5 Hz, 1H), 7.59 (d, J=5.9 Hz, 1H), 8.04 (d, J=8.1 Hz, 1H), 8.65 (d, J=5.9 Hz, 1H), 9.29 (s, 1H); MS: 189 [M+H]+.


c. 6-Methyl-5-nitro-1,2,3,4-tetrahydro-isoquinoline (RN009_Int03)

To an ice-cold solution of RN009_Int02 (20 g, 106.38 mmol) in acetic acid (180 mL) was added NaBH4 (8.08 g, 212.76 mmol) in small portions. The resultant reaction mixture was stirred further at 0° C. After completion of the reaction (TLC), the mixture was diluted with ice-cold water, basified with NH4OH and extracted with EtOAc. The combined organic layer was washed with brine, dried over Na2SO4 and concentrated under reduced pressure to afford the title compound. The compound was used as such in the next step without further purification. 1H NMR (300 MHz, CDCl3): δ 2.01 (s, 1H), 2.28 (s, 3H), 2.72 (t, J=5.9 Hz, 2H), 3.15 (t, J=5.9 Hz, 2H), 4.04 (s, 2H), 7.06-7.07 (m, 2H); MS: 193 [M+H]+


d. 6-Methyl-5-nitro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (RN009_Int04)

To a stirring solution of RN009_Int03 (22 g, 114.58 mmol) in THF (180 mL) was added aq. NaOH (18.2 g, 458 mmol) and Boc2O (37.4 g, 171 mmol) and the resultant reaction mixture was stirred further. After 16 h, the mixture was concentrated and diluted with EtOAc and water. The layers were separated and the aq. layer was extracted with EtOAc. The combined organic layer was washed with brine, dried over Na2SO4 and the solvent was evaporated to leave a residue of crude compound. The crude compound was purified by column chromatography (EtOAc/Hex 10:90) to afford the title compound in 66% yield (over two steps). 1H NMR (300 MHz, CDCl3): δ 1.48 (s, 9H), 2.30 (s, 3H), 2.74 (t, J=5.8 Hz, 2H), 3.63 (t, J=6.0 Hz, 2H), 4.58 (s, 2H), 7.13-7.16 (m, 2H); MS: 193 [M−Boc+H]+; LCMS Purity: 99.97%.


e. 5-Amino-6-methyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (RN009_FP)

To a cooled solution of RN009_Int04 (21 g, 71.9 mmol) in THF (210 mL) was added Pd/C (3.00 g) and the resultant reaction mixture was subjected to reduction in the presence of hydrogen pressure (60 psi) in a Parr apparatus for 2 h. After completion of the reaction Pd/C was filtered using a celite pad. The filtrate was concentrated to afford the title compound as white solid in 74% yield. 1H NMR (300 MHz, CDCl3): 1.47 (s, 9H), 2.16 (s, 3H), 2.56 (t, J=5.9 Hz, 2H), 3.60 (br s, 2H), 330 (t, J=5.9 Hz, 2H), 4.52 (s, 2H), 6.50 (d, J=7.6 Hz, 1H), 6.92 (d, J=7.2 Hz, 1H); LCMS Purity: 99.36%.


Intermediate 4
5-Amino-6-chloro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester











To a stirring solution of 4-chlorobenzaldehyde (SM; 20.00 g, 142.8 mmol) in 200 mL of toluene equipped with a Dean-Stark apparatus and a reflux condenser was added aminoacetaldehyde-dimethylacetal (14.99 g, 142.8 mmol) and the resulting mixture was heated to reflux. After 16 h the solvent was evaporated, the residue was diluted with toluene and evaporated to give an oily residue. The oily residue was dissolved in dry THF (200 mL) and cooled to −10° C. To this cooled solution was added ethyl chloroformate (15.42 g, 142.85 mmol) and the resultant solution was stirred for 10 min at this temperature and allowed to warm to room temperature. At room temperature, trimethylphosphite (21.25 g, 171.42 mol) was added and the resultant mixture stirred further. After 16 h, the mixture was concentrated and traces of solvent and unreacted trimethyphosphite were removed by co-evaporation with toluene. The resultant oily residue was dissolved in DCE (150 mL) and titanium tetrachloride (93.6 g, 189 mmol) was added and refluxed at 100° C. for 16 h. The solvent and titanium tetrachloride were distilled under reduced pressure to leave a residue of crude compound. The crude compound residue was treated with ice-cold water, basified and then extracted with DCM. The DCM layer was washed with brine, dried over Na2SO4 and concentrated to leave a residue of crude compound. The crude compound was purified by column chromatography (EtOAc/Hex 15:85) to afford the title compound in 52% yield. 1H-NMR (300 MHz, CDCl3): δ 7.52-7.57 (m, 2H), 7.79 (s, 1H), 7.90 (d, J=8.7 Hz, 1H), 8.54 (d, J=5.9 Hz, 1H), 9.22 (s, 1H); MS: 164 [M+H]+.


b. 6-Chloro-5-nitro-isoquinoline (RN010_Int02A)

To an ice-cold solution of RN010_Int01A (13.80 g, 84.66 mmol) in Conc. H2SO4 (150 mL) was added KNO3 (10.27 g, 101.68 mmol) in portions. The reaction mixture was allowed to warm to room temperature and stirred for 1 h. After completion of the reaction (TLC) the reaction mixture was cooled to 0° C., diluted with ice-cold water and basified with ice-cold NaOH solution. The resulting mixture was extracted with EtOAc, the combined organic layer was washed with brine, dried over anhy. Na2SO4 and concentrated to afford title compound in 98% yield. 1H NMR (300 MHz, CDCl3): 7.55 (d, J=5.9 Hz, 1H), 7.69 (d, J=8.7 Hz, 1H), 8.10 (d, J=8.7 Hz, 1H), 8.72 (d, J=6.3 Hz, 1H), 9.35 (s, 1H); MS: 209 [M+H]+.


c. 6-Chloro-5-nitro-1,2,3,4-tetrahydro-isoquinoline (RN010_Int03A)

To an ice-cold solution of RN010_Int02A (17.7 g, 85.09 mmol) in acetic acid (200 mL) was added NaBH4 (9.7 g, 255.2 mmol) in small portions. The resultant reaction mixture was stirred further at 0° C. After completion of the reaction (TLC) the mixture was diluted with ice-cold water, basified with NH4OH and extracted with DCM. The combined organic layer was washed with brine, dried over Na2SO4 and the solvent was evaporated under reduced pressure to afford the title compound. Crude RN010_Int03A was used as such in the without further purification.


d. Tert-butyl 6-Chloro-3,4-dihydro-5-nitroisoquinolinc-2(1H)-carboxylate (RN010_Int04a)

To a stirring solution of crude RN010_Int03A (˜17 g, 80.1 mmol) in THF (150 mL) and in water (300 mL) was added. NaOH (12.8 g, 320 mmol) and Boc2O (34.9 g, 160 mmol) and the resultant reaction mixture was stirred further. After 16 h, the mixture was concentrated and diluted with EtOAc and water. The layers were separated and the aq. layer was extracted with EtOAc. The combined organic layer was washed with brine, dried over Na2SO4 and the solvent was evaporated to leave a residue of crude compound. The crude compound was purified by column chromatography (EtOAc/Hex 10:90) to afford the title compound in 75% yield over two steps. 1H NMR (300 MHz, CDCl3): δ 1.48 (s, 9H), 2.75 (t, J=5.8 Hz, 2H), 3.65 (t, J=5.9 Hz, 2H), 4.59 (s, 2H), 7.18 (d, J=8.3 Hz, 1H), 7.33 (d, J=8.4 Hz, 1H).


e. 5-Amino-6-chloro-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester (RN010-FP)

To a Stirring Solution of RN010-Int04a (20 g, 64.1 mmol) in THF (150 mL) was added Pd/C (2.00 g) and the resultant reaction mixture was subjected to reduction in the presence of hydrogen pressure (60 psi) in a Parr apparatus for 2 h. After completion of the reaction Pd/C was filtered using a celite pad. The filtrate was concentrated to afford the title compound as a white solid in 66% yield (12.0 g). 1H NMR (300 MHz, CDCl3): δ 1.48 (s, 9 H), 2.55 (t, J=5.9 Hz, 2H), 3.69 (t, J=5.9 Hz, 2H), 4.02 (br, 2H), 4.50 (s, 2H), 6.48 (d, J=8.3 Hz, 1H), 7.10 (d, J=8.3 Hz, 1H); MS: 183 [M−Boc+H]+; LCMS Purity: 99.76%.


Representative Synthetic Methods
Method A
Compound 3001
N-[6-Chloro-2-(2-chloro-benzenesulfonyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-2-(4-trifluoromethyl-phenyl)-acetamide






a. tert-Butyl 6-chloro-5-(2-(4-(trifluoromethyl)phenyl)acetamido)-3,4-dihydroisoquinoline-2(1H)-carboxylate and N-(6-Chloro-1,2,3,4-tetrahydro-isoquinolin-5-yl)-2-(4-trifluoromethyl-phenyl)-acetamide

A round bottom flask (100 mL) was charged with 2-(4-(trifluoromethyl)phenyl)acetic acid (1.733 g, 0.008488 mol) and methylene chloride (20 mL, 0.3 mol) and oxalyl chloride (0.7182 mL, 0.008488 mol) was added slowly at 0° C. followed by few drops of DMF. The reaction mixture was stirred at that temperature for one hour. The solvent was removed and dissolved in DCM (10 mL) and added to a solution of tert-butyl 5-amino-6-chloro-3,4-dihydroisoquinoline-2(1H)-carboxylate (2.00 g, 0.00707 mol) in methylene chloride (20 mL, 0.3 mol) and the reaction stirred at 0° C. for 1 hour and then at room temperature over night. The reaction mixture provided both the boc deprotected compound as well as the product. The solvent was removed and the residue purified by flash chromatography to obtain the product and the deprotected compound.


b. N-(6-Chloro-1,2,3,4-tetrahydroisoquinolin-5-yl)-2-(4-(trifluoromethyl)phenyl)acetamide hydrochloride

A round bottom flask (100 mL) was charged with tert-butyl 6-chloro-5-(2-(4-(trifluoromethyl)phenyl)acetamido)-3,4-dihydroisoquinoline-2(1H)-carboxylate (1.8 g, 0.0038 mol) and methylene chloride (20 mL, 0.3 mol) and hydrogen chloride in diethyl ether (2M, 20 mL) was added and the reacction stirred at room temperature for 4 h. The product precipitated out, which was filtered and dried. The mother liquor was evaporated and the residue purified by flash chromatography to obtain more product as a white solid.


c. N-(6-Chloro-2-(2-chlorophenylsulfonyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)-2-(4-(trifluoromethyl)phenyl)acetamide

A vial (5 mL) was charged with N-(6-chloro-1,2,3,4-tetrahydroisoquinolin-5-yl)-2-(4-(trifluoromethyl)phenyl)acetamide hydrochloride (20.0 mg, 0.0000494 mol), 2-chlorobenzenesulfonyl chloride (16 mg, 0.000074 mol), chloroform (1 mL, 0.01 mol) and pyridine (0.15 mL, 0.0018 mol) and the reaction stirred at room temperature for 1 h. N,N-Dimethylethylenediamine (0.1 mL) was added and the solvent was removed and the residue was purified by mass triggered HPLC to obtain the product as a white solid. MS m/z=543.2 (M+H).



1H NMR (400 MHz, DMSO): δ 9.84 (s, 1H), 8.04 (d, J=7.90 Hz, 1H), 7.72-7.65 (m, 4H), 7.60-7.55 (m, 3H), 7.36 (d, J=8.23 Hz, 1H), 7.17 (d, J=8.23 Hz, 1H), 4.46 (s, 2H), 3.79 (s, 2H), 3.51 (t, J=6.40 Hz, 2H), 2.62 (t, J=6.40 Hz, 2H).


Method B
Compound 3053
N-(2-Cyano-1,2,3,4-tetrahydro-isoquinolin-5-yl)-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide






a. N-(2-cyano-1,2,3,4-tetrahydroisoquinolin-5-yl)-2-(3-fluoro-4-(trifluoromethyl)phenyl)acetamide

A solution of cyanogen bromide (30 mg, 0.00028 mol) in methanol (1 mL, 0.02 mol) was added drop wise to a ice-cooled mixture of 2-(3-fluoro-4-(trifluoromethyl)phenyl)-N-(1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide hydrochloride (100 mg, 0.0002 mol), sodium acetate (59 mg, 0.00072 mol) and methanol (5 mL, 0.1 mol). The reaction was stirred at 5° C. for 2.5 hours. MeOH was removed over rotovapor, the residue was dissolved in EtOAC (100 mL), washed with 1N HCl (30 mL×2) and brine (30 mL), dried (Na2SO4), filtered, concentrated and purified via flash chromatography (12 g of silica gel, EtOAc/Hexane 0-50%) to give a white solid.



1H NMR δ (CDCl3) δ: 7.64 (br, 1H), 7.52-7.49 (m, 1H), 7.26-7.12 (m, 2H), 6.98-6.89 (m, 2H), 4.40 (br, 2H), 3.80-3.70 (m, 2H), 3.46-3.40 (m, 2H), 2.71-2.61 (m, 2H).


Method C
Compound 3056
N-[2-((S)-2-Amino-3-phenyl-propionyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide






a. N-[2-((S)-2-Amino-3-phenyl-propionyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide

(S)-tert-Butyl 1-(5-(2-(3-fluoro-4-(trifluoromethyl)phenyl)acetamido)-3,4-dihydroisoquinolin-2(1H)-yl)-1-oxo-3-phenylpropan-2-ylcarbamate (25 mg, 0.000042 mol) and hydrogen chloride (4 mL, 0.008 mol) ether solution were stirred at room temperature overnight. The mixture was concentrated, and purified via prep-HPLC to give the desired product as a white solid. 1H NMR δ (DMSO, d-6) δ: 9.61 (s, 1H), 7.78-7.74 (m, 1H), 7.48 (d, J=11.2 Hz, 1H), 7.38 (d, J=8.0 Hz, 1H), 7.25-6.94 (m, 8H), 4.67 (t, J=16.2 Hz, 1H), 4.43 (dd, J=17.0, 55.2 Hz, 1H), 3.96-3.32 (m, 5H), 2.85-2.45 (m, 4H), 1.69 (br, 2H).


Method D
Compound 3057
N-[2-(3-Chloro-2-hydroxy-propyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide






a. N-(2-(3-Chloro-2-hydroxypropyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)-2-(3-fluoro-4-(trifluoromethyl)phenyl)-acetamide

2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide hydrochloride (70 mg, 0.0002 mol), (+/−)-Epichlorohydrin (30 mg, 0.0004 mol), potassium fluoride (5 mg, 0.00009 mol), potassium carbonate (70 mg, 0.0005 mol) and acetone (4 mL, 0.05 mol) were refluxed for 6 hours. The cooled mixture was concentrated and purified via flash chromatography (12 g of silica gel, 0-50% EtOAc/Hexane) to give a white solid.



1H NMR δ (CDCl3) δ: 7.83 (br, 1H), 7.59 (t, J=8.0 Hz, 1H), 7.26 (t, J=9.2 Hz, 1H), 7.04 (t, J=7.8 Hz, 1H), 6.84 (d, J=7.9 Hz, 1H), 3.94-3.91 (m, 1H), 3.71-3.46 (m, 6H), 2.76-2.50 (m, 6H).


Method E
Compound 3058
2-(3-Fluoro-4-trifluoromethyl-phenyl)-N-[2-(2-hydroxy-butyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(2-hydroxybutyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide hydrochloride (70 mg, 0.0002 mol), ethyl oxirane, (1 g, 0.01 mol), potassium fluoride (5 mg, 0.00009 mol), potassium carbonate (70 mg, 0.0005 mol) and acetone (4 mL, 0.05 mol) were refluxed overnight. The cooled mixture was purified via flash chromatography (12 g of silica gel, 0-100% Acetonitrile/hexane) to give a white solid.



1H NMR δ (CDCl3) δ: 7.84 (br, 1H), 7.59 (t, J=7.8 Hz, 1H), 7.26 (t, J=9.2 Hz, 2H), 7.04 (t, J=7.7 Hz, 1H), 6.84 (d, J=7.5 Hz, 1H), 3.71-3.46 (m, 5H), 3.11 (br, 1H), 2.79-2.72 (m, 1H), 2.65-2.54 (m, 2H), 2.38-2.29 (m, 2H), 2.07 (br, 1H), 1.87-1.85 (m, 1H), 1.43-1.23 (m, 2H), 0.86 (t, J=7.4 Hz, 3H).


Method F
Compound 3059
N-{2-[1-(4-Fluoro-phenyl)-ethyl]-1,2,3,4-tetrahydro-isoquinolin-5-yl}-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(1-(4-fluorophenyl)ethyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide hydrochloride (60 mg, 0.0002 mol), 1-(1-bromoethyl)-4-fluorobenzene (60 mg, 0.0003 mol), potassium fluoride (4 mg, 0.00008 mol), potassium carbonate (60 mg, 0.0005 mol) and acetone (4 mL, 0.05 mol) were refluxed overnight. The cooled mixture was purified via flash chromatography (12 g of silica gel, 0-50% EtOAc/Hexane) to give a white solid. 1H NMR δ (CDCl3) δ: 8.10 (br, 1H), 7.91 (t, J=8.1 Hz, 1H), 7.59-7.55 (m, 3H), 7.52-7.48 (m, 2H), 7.34 (t, J=7.8 Hz, 1H), 7.27-7.23 (m, 2H), 7.15 (d, J=7.7 Hz, 1H), 4.02 (s, 2H), 3.86 (s, 2H), 3.08 (t, J=7.1 Hz, 2H), 2.97-2.85 (m, 3H), 2.18-2.16 (m, 3H).


Method G
Compound 3060
N-[6-Methyl-2-((R)-pyrrolidine-2-carbonyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-2-(4-trifluoromethyl-phenyl)-acetamide






a. (R)—N-(6-Methyl-2-(pyrrolidine-2-carbonyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)-2-(4-(trifluoromethyl)phenyl)acetamide

(R)-tert-Butyl 2-(6-methyl-5-(2-(4-(trifluoromethyl)phenyl)acetamido)-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)pyrrolidine-1-carboxylate (46 mg, 0.000084 mol) and hydrogen chloride (4 mL, 0.008 mol) ether solution were stirred at room temperature overnight. The mixture was concentrated and purified via prep-HPLC to give the desired product as a white solid.


NMR δ (CDCl3) δ: 7.67-7.64 (m, 2H), 7.54-7.51 (m, 2H), 7.22 (br, 1H), 7.11-7.03 (m, 1H), 6.92 (d, J=7.8 Hz, 1H), 4.62 (d, J=2.0 Hz, 1H), 4.48 (s, 1H), 3.89-3.80 (m, 3H), 3.72 (t, J=5.9 Hz, 1H), 3.59-3.56 (m, 1H), 3.18-3.11 (m, 1H), 2.84-2.77 (m, 1H), 2.61-2.56 (m, 1H), 2.25-2.04 (m, 6H), 1.83-1.55 (m, 3H).


Method H
Compound 3061
2-(3-Fluoro-4-trifluoromethyl-phenyl)-N-[6-methyl-2-((R)-pyrrolidine-2-carbonyl)-1,2,3,4-tetrahydro-isoquinolin-5-yl]-acetamide






a. (R)-tert-Butyl 2-(5-(2-(3-fluoro-4-(trifluoromethyl)phenyl)acetamido)-6-methyl-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)pyrrolidine-1-carboxylate

2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(6-methyl-1,2,3,4-tetrahydro isoquinolin-5-yl)acetamide hydrochloride (180 mg, 0.00042 mol), N-(tert-butoxycarbonyl)-D-proline (200 mg, 0.0009 mol), N,N,N′,N′-tetramethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (300 mg, 0.0008 mol), N,N-diisopropylethylamine (240 mg, 0.0018 mol) and N,N-dimethylformamide (6 mL, 0.08 mol) were stirred at room temperature overnight. The mixture was purified via flash chromatography (40 g of silica gel, 0-50%-100%) EtOAc/Hexane) gave the desired product as a white solid.


b. (R)-2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(6-methyl-2-(pyrrolidine-2-carbonyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

(R)-tert-Butyl 2-(5-(2-(3-fluoro-4-(trifluoromethyl)phenyl)acetamido)-6-methyl-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)pyrrolidine-1-carboxylate (43 mg, 0.000076 mol) and hydrogen chloride (4 mL, 0.008 mol) ether solution were stirred at room temperature overnight, concentrated, purified via prep-HPLC gave the desired product as a white solid.


NMR δ (CDCl3) δ: 7.64-7.60 (m, 1H), 7.51 (br, 1H), 7.29-7.26 (m, 2H), 7.09-7.06 (m, 1H), 6.91 (t, J=7.9 Hz, 1H), 4.67-4.47 (m, 2H), 3.89-3.55 (m, 5H), 3.17-3.13 (m, 1H), 2.84-2.76 (m, 1H), 2.62-2.50 (m, 2H), 2.21-2.00 (m, 5H), 1.86-1.52 (m, 3H).


Method J
Compound 3065
N-{2-[2-(4-Fluoro-phenyl)-2-hydroxy-ethyl]-1,2,3,4-tetrahydro-isoquinolin-5-yl}-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(2-(4-fluorophenyl)-2-hydroxyethyl)-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide hydrochloride (70 mg, 0.0002 mol), 2-(4-fluorophenyl)-oxirane (100 mg, 0.0009 mol), potassium fluoride (5 mg, 0.00009 mol), potassium carbonate (70 mg, 0.0005 mol) and acetone (4 mL, 0.05 mol) were refluxed overnight. The cooled mixture was purified via flash chromatography (12 g of silica gel, 0-100% Acetonitrile/hexane) to give a white solid. 1H NMR δ (CDCl3) δ: 7.69-7.62 (m, 2H), 7.36 (dd, J=5.4, 8.5 Hz, 2H), 7.27-7.13 (m, 3H), 7.04 (t, J=8.7 Hz, 2H), 6.91 (d, J=7.7 Hz, 1H), 6.83 (s, 1H), 4.83 (dd, J=3.5, 10.0 Hz, 1H), 3.92 (d, J=15.1 Hz, 1H), 3.81 (s, 2H), 3.68 (d, J=15.1 Hz, 1H), 3.04-3.01 (m, 1H), 2.80-2.54 (m, 5H), 1.75-1.55 (m, 1H).


Method K
Compound 3474
N-[2-(2,2-Dimethyl-propionyl)-6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl]-2-(3-fluoro-4-trifluoromethyl-phenyl)-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(6-methoxy-2-pivaloyl-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

Into a 20 ml reaction vial was combined 2-(3-fluoro-4-(trifluoromethyl)phenyl)-N-(6-methoxy-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide hydrochloride (55 mg, 0.00013 mol), N,N-diisopropylethylamine (200 μL, 0.0009 mol) and methylene chloride (8 mL, 0.1 mol). 2,2-dimethylpropanoyl chloride (32.3 μL, 0.000263 mol) was added dropwise and the mixture was stirred at room temperature for an hour. The mixture was purified using flash chromatography with Ethyl acetate and Hexanes. The combined pure fractions were collected to produce a white solid.



1H-NMR (400 MHz, DMSO-d6) [δ] 9.52 (s, 1H), 7.77 (t, 1H, J=7.94 Hz), 7.49 (d, 1H, J=12.07 Hz), 7.40 (d, 1H, J=8.15 Hz), 7.05 (s, 2H), 4.64 (s, 2H), 3.81 (s, 2H), 3.70 (t, 2H, J=5.80 Hz), 2.56 (br′s′, 2H), 2.06 (s, 3H), 1.20 (s, 9H).


Method L
Compound 3476
2-(3-Fluoro-4-trifluoromethyl-phenyl)-N-[2-(1-hydroxy-cyclopropanecarbonyl)-6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl]-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(1-hydroxycyclopropanecarbonyl)-6-methoxy-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

In a 20-ml vial was combined 2-(3-fluoro-4-trifluoromethyl-phenyl)-N-(6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl)-acetamide (100.0 mg, 0.0002615 mol), 1-hydroxycyclopropanecarboxylic acid (0.040 g, 0.00039 mol), and N,N-diisopropylethylamine (230 μL, 0.0013 mol) in methylene chloride (1000 μL, 0.02 mol). The reaction was stirred for one hour at 50° C. The reaction was then purified by HPLC to afford the title compound as a white powder.



1H NMR (400 MHz, DMSO-d6) [δ] 9.48 (s, 1H), 7.77 (t, 1H, J=7.93 Hz), 7.50 (d, 1H, J=12.12 Hz), 7.39 (d, 1H, J=8.08 Hz), 7.10 (d, 1H, J=8.55 Hz), 6.92 (d, 1H, J=8.55 Hz), 6.35 (s, 1H), 4.87-4.53 (m, 2H), 3.88-3.55 (m, 7H), 2.58 (br′s′, 2H), 0.90 (br′s′, 2H), 0.79-0.76 (m, 2H).


Method M
Compound 3477
2-(3-Fluoro-4-trifluoromethyl-phenyl)-N-[2-(1-hydroxy-cyclohexanecarbonyl)-6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl]-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(1-hydroxycyclohexanecarbonyl)-6-methoxy-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

In a 20-ml vial was combined 2-(3-fluoro-4-trifluoromethyl-phenyl)-N-(6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl)-acetamide (100.0 mg, 0.0002615 mol), 1-hydroxy-cyclohexanecarboxylic acid (0.056 g, 0.00039 mol), and N,N-diisopropylethylamine (230 μL, 0.0013 mol) in methylene chloride (1000 μL, 0.02 mol). The reaction was stirred for one hour at 50° C. The reaction mixture was then purified by HPLC to afford the title compound.



1H-NMR (400 MHz, DMSO-d6) [δ] 9.45 (s, 1H), 7.77 (t, 1H, J=7.89 Hz), 7.49 (d, 1H, J=12.22 Hz), 7.39 (d, 1H, J=8.08 Hz), 7.07 (d, 1H, J=8.67 Hz), 6.90 (d, 1H, J=8.67 Hz), 5.28 (s, 1H), 5.04 (br′s′, 1H), 4.55 (br′s′, 1H), 4.03 (br′s′, 1H), 3.79 (s, 2H), 3.73 (s, 3H), 3.56 (br′, s′, 1H), 2.53 (br′s′, 1H), 1.67-1.49 (m, 10H), 1.21-1.13 (m, 1H).


Method N
Compound 3478)
2-(3-Fluoro-4-trifluoromethyl-phenyl)-N-{2-[2-(1-hydroxy-3,3-dimethyl-cyclohexyl)-acetyl]-6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl}-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(2-(1-hydroxy-3,3-dimethylcyclohexyl)acetyl)-6-methoxy-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

In a 20-ml vial was combined 2-(3-fluoro-4-trifluoromethyl-phenyl)-N-(6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl)-acetamide (100.0 mg, 0.0002615 mol), 2-(1-hydroxy-3,3-dimethylcyclohexyl)acetic acid (0.073 g, 0.00039 mol), and N,N-diisopropylethylamine (230 μL, 0.0013 mol) in methylene chloride (1000 μL, 0.02 mol). The reaction was stirred for one hour at 50° C. The reaction was then purified by HPLC to afford the title compound. 1H-NMR (400 MHz, DMSO-d6) [δ] 9.51 (d, 1H, J=4.56 Hz), 7.77 (t, 1H, J=8.02 Hz), 7.49 (d, 1H, J=12.17 Hz), 7.39 (d, 1H, J=8.02 Hz), 7.10 (t, 1H, J=8.57 Hz), 6.93 (dd, 1H, J=8.57 Hz), 4.78 (d, 1H, J=11.06 Hz), 4.63 (s, 1H), 4.56 (s, 1H), 3.80 (s, 2H), 3.74 (s, 3H), 3.62-3.55 (m, 2H), 2.56 (t, 1H, J=5.81 Hz), 2.48-2.41 (m, 2H), 1.74-1.65 (m, 1H), 1.59 (d, 1H, J=13.51 Hz), 1.44 (dd, 1H, J=13.51 Hz), 1.34-1.29 (m, 2H), 1.24-1.13 (m, 2H), 1.03 (d, 3H, J=1.66 Hz), 0.80 (d, 3H, J=8.39 Hz).


Method O
Compound 3479
2-(3-Fluoro-4-trifluoromethyl-phenyl)-N-[2-(3-hydroxy-2,2-dimethyl-propionyl)-6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl]-acetamide






a. 2-(3-Fluoro-4-(trifluoromethyl)phenyl)-N-(2-(3-hydroxy-2,2-dimethylpropanoyl)-6-methoxy-1,2,3,4-tetrahydroisoquinolin-5-yl)acetamide

In a 20-ml vial was combined 2-(3-fluoro-4-trifluoromethyl-phenyl)-N-(6-methoxy-1,2,3,4-tetrahydro-isoquinolin-5-yl)-acetamide (100.0 mg, 0.0002615 mol), 3-hydroxy-2,2-dimethyl-propionic acid (0.046 g, 0.00039 mol), and N,N-diisopropylethylamine (230 μL, 0.0013 mol) in methylene chloride (1000 μL, 0.02 mol). The reaction was stirred for one hour at 50° C. The reaction was then purified by HPLC to afford the title compound.



1H-NMR (400 MHz, DMSO-d6) [δ] 9.47 (s, 1H), 7.77 (t, 1H, J=7.96 Hz), 7.49 (d, 1H, J=12.05 Hz), 7.39 (d, 1H, J=7.96 Hz), 7.12 (d, 1H, J=8.61 Hz), 6.92 (d, 1H, J=8.61 Hz), 4.61 (s, 2H), 4.55 (t, 1H, J=5.89 Hz), 3.78 (s, 2H), 3.73 (s, 3H), 3.66 (t, 2H, J=5.73 Hz), 3.43 (d, 2H, J=5.95 Hz), 2.52 (t, 2H, J=5.95 Hz), 1.15 (s, 6H).


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′-(O)-(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 (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-PRO1 fluorescence; (iii) ethidium bromide fluorescence; and (iv) IL-1β 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), 2 mM glutamine, 5 mM HEPES, and 100 μg/ml streptomycin. Cells are allowed to adhere to culture flasks for 1-2 h, after which nonadherent cells are washed away. Adherent cells are cultured for 7-14d in this medium plus interferon-γ (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 12-24 h 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 KCl, 2 mM CaCl2, 1 mM MgCl2, 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 μM) or BzATP (30 μM) (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 Ercv by first computing a (═PNo/PK′ where P is permeability) for internal (i) and external (o) concentrations [Na]1=20 mM, [Na]o=145 mM, [K]o=0 mM, and [K]1=140 mM from α=([145/exp(ErevF/RT)]−20)/140 (where F is the Faraday, R is the gas constant, and T is the absolute temperature). Other Px/PNa values, when [X]o=145 mM, [Na]1=20 mM, [K]1=140 mM, and [Na]o=[K]o=[X]1=0 mM, are computed from Px/PNa=[(exP)ErevF/RT)] (20+140α)/145. In order of size, X is 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 HCl, histidine, or Hepes as required, and the osmolarity of all solutions is 295 to 315.


Example 3
YO-PRO1 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 40× Fluor objective. YO-PRO-1 (10 μM; Molecular Probes, Eugene, Oreg.) is added to the superfusion 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-PROI fluorescence is measured using 491/509 nm excitation/emission wavelengths. Images are obtained at 5-20s intervals during continuous superfusion (2 ml/min) with YO-PRO1 and varying concentrations of control ATP, BzATP or compound to be tested. For each experiment, the time course of YO-PRO1 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 μl of test solution comprising 200 μl of a suspension of P2X7-expressing cells (e.g. THP-1 cells, J774 cells, etc.)(2.5×106 cells/ml) containing 104M ethidium bromide, 25 μl of a high potassium buffer solution containing 10−5M BzATP, and 25 μl 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 mu. 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 pIC50 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-1β Release

This Example demonstrates the testing of the compounds of this invention for efficacy as inhibitors of P2X7-mediated release of IL-1β from human macrophages activated by the Alzheimer's beta amyloid peptide 1-42.


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 800×g 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 800×g for 5 minutes. Cells are then washed by sequential resuspension of the cell pellet in wash buffer and centrifugation at 600×g 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 serum-free RPMI 1640 in 96-well plates and incubated for 1 hour at 37° C. in a 5% CO2/95% humidified tissue culture incubator. After 1 hour, the medium is replaced with 100 μl 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 μl fresh complete culture medium in the absence or presence of human beta amyloid 1-42 peptide (5 μM) and incubated at 37° C. in a 5% CO2/95% 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 μl of HBSS/CaCl2-inhibiting compound of the present invention (10× 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 μl of HBSS/CaCl2 or 10 μl of benzoyl ATP (BzATP; 3 mM stock in HBSS/CaCl2 for a 300 μM 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-18 content was quantitated by ELISA (from R&D Systems). The cells are washed once with HBSS/CaCl2 followed by lysing the cells with 100 μl 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-113 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.


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.100 mg).


Pertussis Toxin


Bordetella Pertussis, (Lyophilized powder containing PBS and lactose) is obtained from List Biological Laboratorie, 1-408-866-6363 (Product #180, 50 ug).


Induction of EAE in Mice

PLP139-151 peptide is dissolved in H2O:PBS (1:1) solution to a concentration 7.5 mg/10 ml (for 75 μg 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.


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% O2, 70% N2O 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 carotid 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 with 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 <300 gm, 19 mm for rats weighing 0.300 gm) effectively blocking blood flow to the middle cerebral artery.


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.


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 45×23×15 cm. Animals are housed in APEC® cages (Allentown Caging, Allentown, N.J. 08501, USA) in a positive pressure isolator (NuAire®, Mode: Nu-605, airflow velocity 50±5 ft/min, 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).


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.1″ 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)/BW×100%. 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% NaCl 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.


Equipment

Glass syringe (1 ml and 2 ml Mitsuba, Japan), Hypodermic needle 24Gx1″ (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 H 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 H 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 μg 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% NaCl, 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:





inhibition (%): [1−(Tn−To)/(Cn−Co)]×100


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 gm (Charles River Laboratories, San Diego, Calif.) 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 nerve 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 ⅓-½ 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, Calif.).


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, Calif.) 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.


Example 8
Pore Formation

THP-1 cells (ATCC Cat # 285-IF-100) 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-γ 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 in M d-glucose, 118 mM NMDG, 5 mM KCl, 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′-O-(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-1β Release Assay

THP-1 cells (ATCC Cat # 285-IF-100) 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-γ 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′-O-(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-1β levels determined via ELISA (R&D systems Cat. # HSLB50) according to manufacturer's recommendations using the Tecan Safire plate reader. Background adjusted IL-1β 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, which can be determined using the IL-1β assay method described in Example 9, is expressed as follows:


“+” compound exhibited 0-25% inhibition at 0.3 μM concentration


“++” compound exhibited 26-50% inhibition at 0.3 μM concentration


“+++” compound exhibited 51-75% inhibition at 0.3 μM concentration


“++++” compound exhibited 76% or greater inhibition at 0.3 μM concentration


“*” compound exhibited 0-25% inhibition at 0.1 μM concentration


“**” compound exhibited 26-50% inhibition at 0.1 μM concentration


“***” compound exhibited 51-75% inhibition at 0.1 μM concentration


“****” compound exhibited 76% or greater inhibition at 0.1 μM concentration


Compounds with a percent inhibition represented by “++++” or “****” are of particular interest.









TABLE 1







IL-1β% Inhibition of Exemplary Compounds














MS
IL-1β% Inhib.


ID
Structure
MW
(obs)
@ 0.3 or 0.1 μM














3001





543.39
543.20
++++





3002





526.94
527.40
*





3003





526.94
527.50
*





3004





526.94
527.40
*





3005





544.93
345.10
*





3006





544.93
545.20
*





3007





544.93
545.20
*





3008





544.93
545.20
*





3009





544.93
545.10
*





3010





540.96
541.20
*





3011





540.96
541.00
*





3012





543.39
543.20
*





3013





543.39
543.20
*





3014





577.84
577.10
*





3015





577.84
577.20
*





3016





561.38
561.10
*





3017





561.38
561.20
*





3018





561.38
561.10
*





3019





56138
560.80
*





3020





611.39
611.00
*





3021





611.39
611.40
*





3022





611.39
611.30
*





3023





522.97
523.40
*





3024





522.97
523.40
*





3025





537.00
537.20
++++





3026





537.00
537.20
*





3027





533.96
534.20
*





3028





533.96
533.90
*





3029





533.96
534.30
*





3030





538.97
539.10
*





3031





576.94
577.30
*





3032





576.94
577.20
*





3033





576.94
577.30
*





3034





592.94
593.00
*





3035





451.46
452.00
*





3036





514.47
515.2
+





3037





514.47
515.2
++++





3038





464.46
465.20
+





3039





450.43
451.10
++





3040





512.50
513.30
++++





3041





420.40
420.60
+





3042





476.51
476.60
++++





3043





490.54
490.60
++++





3044





471.45
471.70
+





3045





599.62
600.40
++





3046





549.56
550.40
+





3047





593.61
594.20
+++





3048





523.52
524.40
+





3049





549.56
550.40
+





3050





446.88
447.20
++





3051





449.45
449.90
++++





3052





493.50
493.70
+





3053





377.34
378.00
+





3054





423.41
423.80
+





3055





449.45
451.00
+++





3056





499.51
499.90
++++





3057





444.85
447.00
++





3058





424.44
424.60
++





3059





474.47
474.50
+++





3060





445.48
445.80
++++





3061





463.47
463.60
++++





3062





463.47
463.60
++++





3063





438.46
438.70
++





3064





428.40
430.10
+





3065





490.47
490.70
+++





3066





506.92
506.80
+++





3067





570.50
572.30
++++





3068





559.77
558.80
+





3069





428.81
429.10
+





3070





490.88
491.10






3071





442.84
443.10






3072





504.91
505.10






3073





525.33
525.10
+





3074





504.91
505.10
++





3075





510.96
511.10
+





3076





520.91
521.10
+





3077





566.98
567.20
+





3078





544.85
545.10
+





3079





491.87
491.90
+





3080





482.90
483.10
+++





3081





564.96
565.20
+





3082





458.84
459.10






3083





522.90
523.10
+





3084





532.96
533.10
+





3085





496.93
497.50






3086





559.77
558.80
+





3087





550.93
551.10
+





3088





456.86
457.10
+





3089





518.94
519.10
+





3090





520.91
521.10
+





3091





508.87
509.10
+





3092





470.89
471.10
++++





3093





525.33
525.10
+





3094





532.96
533.10






3095





470.89
471.50






3096





533.95
533.90
++





3097





534.93
535.10
+





3098





515.89
515.90
+





3099





530.95
531.10
+





3100





559.77
558.80
+





3101





468.88
469.10
+





3102





454.85
455.10
+





3103





496.93
497.50
+++





3104





555.35
555.20
+





3105





539.35
539.10
++





3106





558.88
559.20
++





3107





557.34
556.80
+





3108





571.96
572.00
+





3109





550.93
551.10






3110





548.92
549.10
+





3111





526.86
527.10






3112





504.91
505.10
+





3113





526.86
527.10
+





3114





508.87
509.10
+





3115





559.77
558.80
+





3116





495.86
496.30
+





3117





508.90
509.10






3118





516.92
517.10
++





3119





504.91
505.10
++





3120





520.91
521.10
+





3121





540.94
541.10
+





3122





539.95
540.30
+





3123





456.86
45710
+





3124





546.99
547.10
+





3125





541.93
541.90
++





3126





491.87
491.90
+





3127





532.92
533.10






3128





547.93
547.90
+





3129





550.93
551.10
+





3130





524.98
525.10
+++





3131





559.77
558.80
+





3132





559.87
560.00
+





3133





583.97
584.00
+





3134





497.90
498.30
++





3135





534.93
535.10
++





3136





428.81
429.10
+





3137





490.88
491.10
+





3138





442.84
443.10
+





3139





525.33
525.10
+





3140





504.91
505.10
+





3141





520.91
521.10
+





3142





566.98
567.20
+





3143





544.85
545.10
+





3144





482.90
483.10
+





3145





458.84
459.10
+++





3146





532.96
533.10
++





3147





496.93
497.50
+





3148





559.77
558.80
+





3149





550.93
551.10
++





3150





456.86
457.10
+





3151





518.94
519.10
++





3152





520.91
521.10
+





3153





520.91
521.10
+





3154





508.87
509.10
+





3155





470.89
471.50
+





3156





525.33
525.10
+





3157





532.96
533.10
+





3158





470.89
471.50
+





3159





533.95
533.90
+





3160





534.93
535.10
+





3161





515.89
516.30
+





3162





530.95
531.10
++





3163





559.77
559.20
++





3164





468.88
469.10
+





3165





454.85
455.10
+





3166





496.93
497.50
+





3167





555.35
555.20
++





3168





539.35
539.10
+





3169





576.87
577.20
+





3170





558.88
559.20
+





3171





557.34
557.20
+





3172





571.96
572.00
+





3173





550.93
551.10






3174





548.92
549.10
+





3175





526.86
527.10
+





3176





504.91
505.10
+





3177





526.86
527.10
+





3178





508.87
509.10
+





3179





559.77
558.80
++





3180





495.86
496.30
+





3181





508.90
509.10
+





3182





516.92
517.10
+





3183





504.91
505.10
+





3184





520.91
521.10
+





3185





540.94
541.10
+





3186





539.95
540.30






3187





456.86
457.10
+





3188





546.99
547.10
++++





3189





541.93
541.90
+





3190





491.87
492.30
+





3191





532.92
533.10






3192





547.93
548.30






3193





550.93
551.10
+





3194





524.98
525.10
++





3195





559.77
558.80
+





3196





559.87
560.00
+





3197





583.97
584.00
+





3198





497.90
498.30
+





3199





534.93
535.10
+





3200





539.35
539.10
++





3201





408.39
409.10
++++





3202





470.46
471.10
+





3203





422.42
423.10
++++





3204





484.49
485.10
+





3205





504.91
505.10
+





3206





484.49
485.10
+





3207





490.54
491.10
+++





3208





462.48
463.10
++++





3209





544.54
545.10
++





3210





438.42
439.10
+





3211





502.48
503.10
+++





3212





512.54
513.10






3213





539.35
539.10
+





3214





530.52
531.10
+





3215





436.45
437.10
++++





3216





498.52
499.10
+





3217





500.49
501.10
++





3218





500.49
501.10






3219





488.45
489.10
+





3220





450.47 450.90
++++






3221





512.54
513.50
+





3222





450.47
451.10
+





3223





513.53
514.30
+





3224





514.52
515.10
++





3225





495.47
496.30
+





3226





510.53
511.50
++





3227





539.35
539.10
+





3228





476.51
477.10
+





3229





534.93
535.10






3230





518.94
519.10
++





3231





536.93
537.10
+





3232





551.54
552.30
+





3233





530.52
531.10






3234





506.44
507.10
+++





3235





484.49
485.10
+





3236





506.44
507.10
+





3237





488.45
489.10
++





3238





539.35
539.10
+





3239





475.44
475.90
+





3240





488.48
489.10
++





3241





496.50
497.10
++





3242





484.49
485.10
++++





3243





500.49
501.10
+





3244





520.52
521.10






3245





519.54
520.30
+





3246





526.57
527.10
+





3247





521.51
522.30
++





3248





471.45
472.30
+





3249





527.52
527.90
+





3250





530.52
531.10
+





3251





504.56
505.10
++++





3252





539.35
539.10
+





3253





539.45
540.30
+





3254





563.55
564.40
+





3255





477.48
477.90
++++





3256





514.52
515.10
+++





3257





486.51
486.50
++





3258





722.46
722.00
+





3259





561.38
560.80
+





3260





556.96
557.20
+





3261





506.95
507.10
++





3262





544.93
545.10
++





3263





649.06
649.20
++





3264





540.96
541.10
++





3265





544.93
545.10
++





3266





492.92
493.10
+





3267





532.86
532.70
+





3268





586.99
587.20
+





3269





540.96
541.10
+





3270





687.92
687.20
+





3271





583.02
582.80
+





3272





478.89
479.10
++++





3273





562.92
563.20
++++





3274





619.03
619.20
++





3275





556.96
556.80
++





3276





586.99
587.20
+





3277





619.03
619.20
+





3278





540.96
541.10
++





3279





586.99
587.20
++





3280





490.90
491.10
++++





3281





594.93
595.20
+





3282





610.93
611.20
+





3283





540.96
541.10
+++





3284





610.93
611.20
+++





3285





561.38
560.80
++





3286





569.02
569.20
+





3287





561.38
560.80
++++





3288





619.03
619.20
+





3289





687.92
687.20
+





3290





687.03
687.20
+





3291





492.92
493.10
++





3292





586.99
587.20
++





3293





633.06
633.20
+





3294





583.02
582.80
+





3295





575.41
575.20
++





3296





592.94
593.20
++





3297





603.03
603.20
+





3298





595.83
594.80
++





3299





545.94
545.90
+++





3300





544.93
545.10
+





3301





687.92
686.80
++





3302





620.02
620.40
++





3303





544.93
545.10
++





3304





492.92
493.10
++++





3305





561.38
560.80
+





3306





562.92
563.20
++





3307





558.95
558.80
+





3308





722.46
722.00
++





3309





608.96
609.20
+++





3310





569.02
569.20
+





3311





556.96
557.20
++





3312





561.38
560.80
++





3313





490.90
491.10
++++





3314





603.03
603.20
++





3315





577.00
577.20
++





3316





619.03
619.20
+





3317





594.93
595.20
++





3318





577.00
577.20
++





3319





561.38
560.80
++





3320





556.96
557.20
++





3321





562.92
563.20
++++





3322





478.89
479.10
+





3323





617.06
617.20
+





3324





575.41
575.20
+





3325





579.37
579.20
+





3326





544.93
545.10
+





3327





506.95
507.10
++++





3328





617.06
617.20
+





3329





610.93
610.80
+





3330





687.92
687.20
+





3331





633.06
633.20
+





3332





594.93
595.20
+





3333





492.92
493.10
++++





3334





570.95
571.20
+





3335





540.96
541.10
++





3336





620.02
620.40
++





3337





579.37
579.20
++





3338





544.93
545.10
+





3339





608.96
609.20
+





3340





526.94
527.10
+





3341





558.95
559.20
++





3342





532.86
532.70
++





3343





575.41
575.20
++++





3344





595.83
595.20
+





3345





610.93
611.20
+





3346





594.93
595.20
+++





3347





629.38
629.20
+





3348





551.95
551.90
++++





3349





595.83
594.80
+





3350





608.96
609.20
+





3351





676.96
677.20
+





3352





594.93
595.20
++





3353





584.97
585.20
++





3354





585.02
585.20
+





3355





577.00
577.20
+





3356





579.37
579.20
+





3357





598.01
598.00
+





3358





575.41
575.20
+++





3359





579.37
579.20
+





3360





608.96
609.20
+





3361





609.85
609.20
+





3362





579.37
579.20
++





3363





575.41
575.20
+





3364





551.95
551.90
++





3365





540.96
541.10
+





3366





687.03
687.20
+





3367





681.05
681.20
+





3368





629.38
629.20
+





3369





584.97
585.20
++





3370





526.94
527.10
+





3371





570.95
571.20
+





3372





551.95
551.90
++++





3373





681.05
681.20
+





3374





575.41
575.20
+





3375





598.01
598.00
+





3376





554.99
555.20
+





3377





605.03
605.20
+





3378





592.94
593.20
+





3379





668.01
668.00
+





3380





668.01
668.00
+





3381





609.85
608.80
+





3382





676.96
677.20
+





3383





584.01
584.00
++





3384





584.01
584.00
+





3385





562.01
562.00
+





3386





554.99
555.20
+





3387





562.01
562.00
+





3388





530.93
531.10
++





3389





544.95
545.10
+





3390





609.85
609.20
+





3391





530.93
531.10
++





3392





687.92
687.20
+





3393





544.95
545.10
+





3394





586.39
586.00
++





3395





524.54
525.10
+





3396





656.54
657.60
+





3397





589.43
589.20
+





3398





667.50
667.20
+





3399





588.54
589.20
+





3400





589.43
589.20
+





3401





520.54
521.10
++





3402





563.57
564.40
++





3403





541.59
542.30
++





3404





538.53
539.50
+





3405





588.54
589.20
+





3406





512.44
513.10
++++





3407





534.57
535.10
+





3408





667.50
667.20
+





3409





574.51
575.20
+





3410





584.61
585.20
++





3411





525.52
526.30
+





3412





577.60
578.40
+





3413





510.51
511.50
+





3414





472.50
473.10
++++





3415





564.55
565.20
++++





3416





575.41
575.20
+





3417





628.64
629.60
+





3418





562.61
563.20
+





3419





590.51
591.20
+





3420





558.95
559.20
+





3421





472.50
473.10
+++





3422





575.41
575.20
+





3423





540.96
541.10
+





3424





524.51
525.10
++





3425





542.50
543.10
++++





3426





520.54
521.10
+





3427





566.57
567.20
+





3428





558.95
559.20
+





3429





582.62
583.20
+





3430





660.63
661.20
+





3431





612.64
613.20
+





3432





565.97
566.40
++++





3433





458.47
459.10
++++





3434





506.52
507.10
++





3435





666.61
667.20
+





3436





550.53
551.10
+





3437





647.59
648.40
+





3438





608.96
609.20
+





3439





598.61
599.20
++





3440





540.96
541.10
+





3441





536.54
537.10
+++





3442





596.64
597.20
+





3443





556.58
557.20
+





3444





524.51
525.10
+





3445





536.54
537.10
+





3446





574.51
575.20
++





3447





590.51
591.20
+





3448





486.53
487.50
++++





3449





590.51
591.20
+





3450





520.54
521.10
++





3451





572.52
573.20
+





3452





566.57
567.20
++++





3453





542.50
543.10
++++





3454





574.51
575.20
+





3455





540.96
541.10
++++





3456





599.60
600.40
++





3457





554.99
555.20
++





3458





702.04
702.40
+








3459





598.61
599.20
+











3460





524.51
525.10
++++





3461





548.60
549.50
+





3462





556.58
557.20
+





3463





531.53
531.90
++





3464





564.60
565.60
++





3465





563.59
564.40
+++





3466





554.99
555.20
++





3467





531.53
532.30
++++





3468





450.43
450.90
++++





3469





434.43
435.10
++++





3470





452.45
452.30
++++





3471





448.46
448.90
+++





3472





466.47
467.30
+++





3473





448.46
449.20
++++





3474





466.47
466.70
++++





3475





450.43
450.80
+++





3476





466.43
467.00
++





3477





508.51
509.20
+++





3478





550.59
551.40
+++





3479





482.47
483.10
+++





3480





449.45
450.30
++++





3481





468.44
469.00
+





3482





476.51
477.10
++++









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-1β 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)/(1+10̂((Log EC50−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 IC50s of Exemplary Compounds











IL-1β IC50



ID
(nM)














3001
49.28



3025
84.02



3035
966.7



3036
>1000



3037
145.3



3038
430.2



3039
443.2



3040
64.17



3041
494.8



3042
153.2



3043
82.45



3044
>1000



3045
384.6



3046
417.6



3047
199



3048
473.2



3049
772.8



3050
492



3051
73.02



3052
890.5



3053
>1000



3054
684.4



3055
182.2



3056
56.33



3057
427.5



3058
337.6



3059
256



3060
64.38



3061
35.1



3062
76.66



3063
673.2



3064
>1000



3065
205.5



3066
218.8



3067
150.3



3103
172.2



3201
175.1



3203
85.83



3207
203.7



3208
60.23



3215
48.65



3220
25.49



3251
61.93



3257
424.6



3272
51.41



3287
60.71



3304
71.11



3313
34.8



3327
47.24



3406
107.1



3414
83



3425
78.5



3433
25.24



3448
65.42



3452
27.91



3460
41.31



3468
69.8



3469
52.49



3470
32.16



3471
260.8



3472
202.9



3473
23.77



3474
200.9



3475
242.8



3476
325.2



3477
280.4



3478
192.4



3479
171.3



3480
74.88



3481
925.9



3482
164.8










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 mM time point, where 0 mM 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 mM 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/NIS 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=ln 2/k


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 animals' 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 mUkg.


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 as the area under the concentration-time curve extrapolated to time infinite (AUCinf). The AUCinf is averaged and the oral bioavailability (% F) for individual animal is calculated as:






AUC
inf(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.

Claims
  • 1. A bicycloheteroaryl compound having a formula:
  • 2. A compound according to claim 1 wherein A is CH2.
  • 3. (canceled)
  • 4. A compound according to claim 1 wherein each of B and Y is CH2; and the dotted bond is a single bond.
  • 5. (canceled)
  • 6. (canceled)
  • 7. (canceled)
  • 8. (canceled)
  • 9. (canceled)
  • 10. (canceled)
  • 11. (canceled)
  • 12. A compound according to claim 1 wherein R1 is selected from a 5-13 membered aryl and heteroaryl, unsubstituted or substituted with one or more substituents independently selected from halo, hydroxyl, amino, cyano, sulfo, sulfanyl, sulfinyl, amido, carboxy, carbalkoxy, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, and sulfonamide.
  • 13. (canceled)
  • 14. (canceled)
  • 15. (canceled)
  • 16. A compound according to claim 1 wherein R1 is substituted or unsubstituted phenyl, 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. (canceled)
  • 18. (canceled)
  • 19. (canceled)
  • 20. (canceled)
  • 21. (canceled)
  • 22. (canceled)
  • 23. A compound according to claim 1 wherein the compound is according to formula V, VI or VII
  • 24. (canceled)
  • 25. (canceled)
  • 26. (canceled)
  • 27. (canceled)
  • 28. (canceled)
  • 29. A compound according to claim 1 wherein the compound is according to formula VIII, IX or X:
  • 30. (canceled)
  • 31. (canceled)
  • 32. (canceled)
  • 33. (canceled)
  • 34. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is alkyl, or substituted alkyl.
  • 35. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is methyl, ethyl, propyl, iso-propyl, t-butyl, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxy iso-propyl, or hydroxy t-butyl.
  • 36. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is cyclopropylmethyl, cyclopropylethyl, cyclopropylpropyl, cyclobutylmethyl, cyclobutylethyl, cyclobutylpropyl, cyclopentylmethyl, cyclopentylethyl, cyclopentylpropyl, cyclohexylmethyl, cyclohexylethyl, cyclohexylpropyl, cycloheptylmethyl, cycloheptylethyl, cycloheptylpropyl, phenylmethyl, phenethyl, or phenylpropyl, which can be optionally substituted with alkyl or hydroxy.
  • 37. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted bicycloaryl, and substituted or unsubstituted bicycloheteroaryl.
  • 38. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is substituted or unsubstituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, phenyl, naphthyl, pyridyl, pyrrolidinyl, benzopyranyl, benzodioxanyl, or benzofuranyl.
  • 39. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is phenyl 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.
  • 40. A compound according to claim 1 wherein L1 is a CO or SO2 and R3 is benzyl 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.
  • 41. A compound according to claim 1 wherein the group -L1-R3 is selected from
  • 42. A compound according to claim 1 wherein the group -L1-R3 is selected from
  • 43. A compound according to claim 1 wherein the group -L1-R3 is selected from
  • 44. (canceled)
  • 45. (canceled)
  • 46. A compound according to claim 1 wherein the compound is according to formula XIa, XIb, XIc, XId, XIe, XIf, XIg, XIh, or XIj:
  • 47. A compound according to claim 1 wherein the compound is according to formula XIIa, XIIb, or XIc:
  • 48. A compound according to claim 47 wherein R2d is methyl, i-Pr or hydroxymethyl.
  • 49. (canceled)
  • 50. A compound according to claim 1 wherein the compound is according to formula XIIIa, XIIIb, XIIIc, XIIId, or XIIIe:
  • 51. (canceled)
  • 52. (canceled)
  • 53. (canceled)
  • 54. A compound according to any one of claims 29, 46, 47 and 50, wherein m is 1 or 2; and each R4a is independently selected from Me, Et, Ph, Cl, F, Br, CN, OH, OMe, OEt, OPh, COPh, CF3, CHF2, OCF3, i-Pr, i-Bu, t-Bu, SMe, CH═CH—CO2H, SOMe, SO2Me, SO3H, SO3Me, and pyridyl.
  • 55. (canceled)
  • 56. (canceled)
  • 57. (canceled)
  • 58. (canceled)
  • 59. A compound according to any one of claims 1, 23 and 29, wherein each of W and Z is independently CH.
  • 60. (canceled)
  • 61. (canceled)
  • 62. (canceled)
  • 63. A compound according to any one of claims 29, 46, 47 and 50, wherein R5 is H, Me, cyclopropyl, Cl, F, or CF3.
  • 64. A compound according to claim 1 wherein the compound is selected from compounds listed in Table 1.
  • 65. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound of claim 1.
  • 66. The pharmaceutical composition of claim 65, wherein the carrier is a parenteral, oral or topical carrier.
  • 67. (canceled)
  • 68. (canceled)
  • 69. 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 claim 1, or a pharmaceutical composition according to claim 65.
  • 70. (canceled)
  • 71. (canceled)
  • 72. (canceled)
  • 73. (canceled)
  • 74. 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 claim 1, or a pharmaceutical composition of claim 65.
  • 75. (canceled)
  • 76. (canceled)
  • 77. (canceled)
  • 78. (canceled)
  • 79. (canceled)
  • 80. (canceled)
  • 81. (canceled)
  • 82. (canceled)
  • 83. (canceled)
  • 84. (canceled)
  • 85. (canceled)
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US07/06721 3/16/2007 WO 00 7/30/2010
Provisional Applications (4)
Number Date Country
60783304 Mar 2006 US
60782973 Mar 2006 US
60782782 Mar 2006 US
60918124 Mar 2007 US