Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS WHICH SELECTIVELY MODULATE THE CB2 RECEPTOR
Document Type and Number:
WIPO Patent Application WO/2010/147792
Kind Code:
A2
Abstract:
Compounds of formula (I) are disclosed. Compounds according to the invention bind to and are agonists, antagonists or inverse agonists of the CB2 receptor, and are useful for treating inflammation. Those compounds which are agonists are additionally useful for treating pain.

Inventors:
BARTOLOZZI ALESSANDRA (US)
HICKEY EUGENE RICHARD (US)
RIETHER DORIS (US)
WU LIFEN (US)
ZINDELL RENEE (US)
BLUMIRE NIGEL (GB)
ERMANN MONIKA (GB)
GLENN EDWARD THOMAS (GB)
KHOR SOMEINA (GB)
ZAWADZKI PRZEMYSLAW (PL)
Application Number:
PCT/US2010/037697
Publication Date:
December 23, 2010
Filing Date:
June 08, 2010
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BOEHRINGER INGELHEIM INT (DE)
BARTOLOZZI ALESSANDRA (US)
HICKEY EUGENE RICHARD (US)
RIETHER DORIS (US)
WU LIFEN (US)
ZINDELL RENEE (US)
BLUMIRE NIGEL (GB)
ERMANN MONIKA (GB)
GLENN EDWARD THOMAS (GB)
KHOR SOMEINA (GB)
ZAWADZKI PRZEMYSLAW (PL)
International Classes:
C07D261/14; A61K31/42; A61P29/00; C07D413/12; C07D413/14; C07D417/06; C07D417/12; C07D417/14
Domestic Patent References:
WO2008014199A22008-01-31
WO2008039645A12008-04-03
WO2009105509A12009-08-27
WO1997012687A11997-04-10
WO1994007607A11994-04-14
WO1997012683A11997-04-10
WO1997020590A11997-06-12
Foreign References:
US20090034464W2009-02-19
Other References:
EXPERT OPINION ON INVESTIGATIONAL DRUGS, vol. 14, no. 6, 2005, pages 695 - 703
GOSH, A.K. ET AL., J MED. CHEM., vol. 36, 1993, pages 2300 - 2310
A. GENNARO: "Remington: The Science and Practice of Pharmacy,20th Edition,", 2000, LIPPINCOTT WILLIAMS & WILKINS
"Handbook ofPharmaceutical Additives", 1995, GOWER
A.H. KIBBE: "Handbook of Pharmaceutical Excipients", 2000, AMERICAN PHARMACEUTICAL ASS'N
H.C. ANSEL; N.G. POPOVISH: "Pharmaceutical Dosage Forms and Drug Delivery Systems, 5th ed.,", 1990, LEA AND FEBIGER
Attorney, Agent or Firm:
MORRIS, Michael, P. et al. (Inc.900 Ridgebury Road,P.O. Box 36, Ridgefield CT, US)
Download PDF:
Claims:
Claims

1. A compound of the formula (I)

wherein:

ring A is a 5-membered heteroaryl ring;

R1 is hydrogen, C1-10 alkyl or C3-10 cycloalkyl which is optionally substituted with 1-3 C1- io alkyl, each R1 or it's substituent is optionally halogenated;

R2 is C1-10 alkyl, C3-10 cycloalkyl, arylsulfonyl, arylcarbonyl, C1-10 acyl, C3-10 cycloalkylcarbonyl, heterocyclylcarbonyl, heteroarylcarbonyl, heterocyclyl, benzyl, phenethyl, aryl or heteroaryl each optionally independently substituted with 1 to 3 substituents chosen from Cr6 alkyl, C3-10 cycloalkyl, C1-6 alkoxy, C1-6 alkylthio, C1-6 alkylsulfonyl, C1-6 alkoxycarbonyl, C1-6 alkylamino, C3-6 cycloalkylamino, C1-6 dialkylamino, C1-6 alkylaminocarbonyl, C1-6 acylamino, C1-6 dialkylaminocarbonyl, hydroxyl, halogen, cyano, nitro, oxo, heterocyclyl, aryl and heteroaryl, each substituent on R2 where possible is optionally halogenated or substituted with 1 to 3 C1-6 alkyl, C1-6 acyl, C1-6 alkyl sulfonyl, cyano, aryl, oxo or hydroxyl; or R1 and R2 together with the nitrogen atom to which they are attached form a monocyclic, bicyclic or spirocyclic heterocycle or monocyclic or bicyclic heteroaryl ring each optionally substituted with 1 to 3 C1-6 alkyl, C1-6 alkoxy, C1-6 acyl, C1-6 alkyl sulfonyl, cyano, aryl, oxo, hydroxyl or halogen each ring substituent being further optionally halogenated where possible; R3 and R3 are independently hydrogen or C1-6 alkyl optionally halogenated with the proviso that R3 and R3 cannot simultaneously be hydrogen; or R3 and R3 together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl or heterocyclic ring each optionally halogenated;

R4 is hydrogen or methyl;

R is chosen from

m is 0, 1, 2 or 3;

R6 is hydrogen, C1-4 alkyl or C1-4 alkoxy; R7 and R8 are each independently hydrogen, or C1-4 alkyl with the proviso that both R7 and R8 cannot be hydrogen; and wherein R7 and R8 optionally can cyclize to form a C3-7 cycloalkyl ring;

R ,9 is C1-6 alkyl or aryl; ring B is a 5-6 membered heterocyclic ring; n is 0, 1 or 2;

wherein any carbon atom on the formula (I) or any R substituent listed above is optionally partially or fully halogenated where possible;

or a pharmaceutically acceptable salt thereof.

2. The compound according to claim 1 and wherein ring A is

R is C1-6 alkyl, C3-7 cycloalkyl, 1,1-dioxo-lλ -thiomorpholinylcarbonyl, phenylsulfonyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl or piperidinyl, each optionally independently substituted with 1 to 3 substituents chosen from C1-5 alkyl, C3-7 cycloalkyl, C1-5 alkoxy, C1-5 alkylthio, C1-5 alkylsulfonyl, C1-5 alkoxycarbonyl, C1-5 alkylamino, C3-6 cycloalkylamino, C1-5 dialkylamino, C1-5 alkylaminocarbonyl, C1-5 acylamino, C1-5 dialkylaminocarbonyl, hydroxyl, halogen, cyano, nitro, oxo, phenyl and heterocyclyl chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, 1,1-dioxo-lλ6- thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl and piperazinyl, each R2 substituent where possible is optionally halogenated or substitut alkyl, C1-5 acyl, methyl sulfonyl, cyano, phenyl, oxo or hydroxyl; or R1 and R2 together with the nitrogen atom to which they are attached form a ring chosen from thiomorpholinyl, I,l-dioxo-lλ6-thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, benzimidazolyl, pyrazolyl, imidazolyl, triazinyl, indazolyl, indolyl, indolinyl, isoindolyl, isoindolinyl, and 2-aza-spiro[4.5]dec-2-yl, l-aza-spiro[4.5]dec-1-yl, 1- aza-spiro[4.4]non-1-yl, 2-aza-spiro[4.4]non-2-yl, 2-aza-spiro[5.5]undec-2-yl, 1-aza- spiro[5.5]undec-1-yl, each optionally substituted with 1 to 3 C1-6 alkyl, C1-6 alkoxy, C1-6 acyl, C1-6 alkyl sulfonyl, cyano, phenyl, oxo, hydroxyl and halogen each ring substituent being further optionally halogenated where possible;

R3 and R3 are each methyl or ethyl, each optionally halogenated, or R3 and R4 together with the carbon atom to which they are attached form a cyclopropyl or cyclobutyl ring each optionally halogenated.

3. The compound according to claim 2 and wherein ring A is

R1 is hydrogen or C1-3 alkyl optionally halogenated;

R2 is C1-6 alkyl or C3-7 cycloalkyl optionally independently substituted with 1 to 3 halogen, one C3_7 cycloalkyl, C \-β alkoxy or one heterocyclyl chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, I,l-dioxo-lλ6-thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl and piperazinyl each optionally halogenated or substituted with C1-4 alkyl or methyl sulfonyl; or R1 and R2 together with the nitrogen atom to which they are attached form a ring chosen form thiomorpholinyl, I,l-dioxo-lλ6-thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, azetidinyl, indolyl, indolinyl, isoindolyl, isoindolinyl, and 2-aza-spiro[4.5]dec-2-yl each optionally substituted with 1 to 3 C1-4 alkyl, C1-4 alkoxy and halogen each ring substituent being further optionally halogenated where possible;

R3 and R3 are each methyl or ethyl or R3 and R4 together with the carbon atom to which they are attached form a cyclopropyl or cyclobutyl ring;

R is hydrogen or C1-2 alkyl; R7 and R8 are each C1-2 alkyl.

4. The compound according to claim 3 and wherein ring A is

R2 is methyl optionally substituted with one heterocyclyl chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, I,l-dioxo-lλ6-thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl and piperazinyl each optionally halogenated or substituted with C1-4 alkyl or methyl sulfonyl;

or R1 and R2 together with the nitrogen atom to which they are attached form a ring chosen form thiomorpholinyl, I,l-dioxo-lλ6-thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, azetidinyl, indolyl and 2-aza-spiro[4.5]dec-2-yl each optionally substituted with 1 to 3 C1-3 alkyl, C1-3 alkoxy and halogen each ring substituent being further optionally halogenated where possible.

5. The compound according to claim 1 and wherein

ring A is

R is hydrogen, C1-6 alkyl or C3_7 cycloalkyl optionally substituted with 1-3 C1-6 alkyl, each R1 or it's substituent is optionally halogenated;

R2 is cyclopentyl, cyclohexyl, tetrahydropyranyl, tetrahydrofuranyl, thiomorpholinylcarbonyl, I,l-dioxo-lλ6-thiomorpholinylcarbonyl, morpholinylcarbonyl, phenylsulfonyl, phenylcarbonyl, phenyl, pyridinyl, piperidinyl, pyrimidinyl or thiazolyl, each optionally independently substituted with 1 to 3 substituents chosen from C1-5 alkyl, C3-5 cycloalkyl, C1-5 alkoxy, C1-5 alkylthio, C1-5 alkylsulfonyl, C1-5 alkoxycarbonyl, C1-5 alkylamino, C3-6 cycloalkylamino, C1-5 dialkylamino, C1-5 alkylaminocarbonyl, C1-5 acylamino, C1-5 dialkylaminocarbonyl, hydroxyl, halogen, cyano and nitro, each R2 substituent where possible is optionally halogenated or substituted with 1 to 3 C1-5 alkyl or C1- 5 alkyl sulfonyl, or R1 and R2 together with the nitrogen atom to which they are attached form a ring chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, 1,1-dioxo-lλ6- thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, benzimidazolyl, pyrazolyl, imidazolyl, triazinyl, indazolyl, indolyl, indolinyl, isoindolyl, isoindolinyl, and 2-aza-spiro[4.5]dec-2-yl, l-aza-spiro[4.5]dec-1-yl, l-aza-spiro[4.4]non-1-yl, 2-aza-spiro[4.4]non-2-yl, 2-aza-spiro[5.5]undec-2-yl, l-aza-spiro[5.5]undec-1-yl each optionally substituted with 1 to 3 C1-6 alkyl, C1-6 alkoxy, C1-6 acyl, C1-6 alkyl sulfonyl, cyano, phenyl, oxo, hydroxyl or halogen each ring substituent being further optionally halogenated where possible;

R3 and R3 are each methyl optionally halogenated , or R3 and R3 together with the carbon atom to which they are attached form a cyclopropyl or cyclobutyl ring each optionally halogenated;

R4 is hydrogen;

R is chosen from

and

R6 is hydrogen or C1-3 alkyl;

H J4 wherein R and R are each C1-3 alkyl or C3_6 cycloalkyl.

6. The compound according to claim 3, and wherein ring A is

R3 and R3 are methyl;

R is hydrogen or C1-2 alkyl; wherein R7 and R8 are each C1-2 alkyl.

7. The compound according to claim 6, and wherein ring A is

R1 and R2 together with the nitrogen atom to which they are attached form a ring chosen from thiomorpholinyl, I,l-dioxo-lλ6-thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, azetidinyl, indolyl and 2-aza-spiro[4.5]dec-2-yl each optionally substituted with 1 to 3 C1-3 alkyl, C1-3 alkoxy, hydroxy and halogen each ring substituent being further optionally halogenated where possible.

8. The compound according to claim 7 and wherein

ring A is

9. The compound according to claim 8 and wherein

R1 and R2 together with the nitrogen atom to which they are attached form a ring chosen from morpholinyl, pyrrolidinyl and piperidinyl, each optionally substituted with 1 to 3 C1-3 alkyl.

10. The compound according to claim 2, and wherein R2 is cyclopropyl, cyclohexyl, tetrahydropyranyl, pyrrolidinyl or piperidinyl, each optionally independently substituted with 1 to 2 substituents chosen from Cr3 alkyl and Cr2 alkylsulfonyl.

11. The compound according to claim 2, and wherein R2 is CF3-CH2-CH2-CH2-, CH3-CH2-CH2(CH3)-, cyclopropyl-CH2-, hydroxylcyclohexyl, tetrahydrofuranyl-CH2- ; ring A is

Rs is wherein wherein R and R optionally can cyclize to form a C3_7 cycloalkyl ring.

12. The compound according to claim 2, and wherein RR22 hhyyddrrooxxyyllccyycclloohheexxyyll,, tetrahydropyranyl, hydroxylpyrrolidinyl or methylsulfonylpyrrolidinyl;

ring A is

R5 is wherein wherein R7 and R8 optionally can cyclize to form a C3-7 cycloalkyl ring;

R1 and R2 optionally can cyclize to form piperidinyl, methyl sulfonylpiperidinyl.

13. A compound of the formula (I) wherein the defintions: ring A, R1 , R2 , R3 , R3' , R4 , R5 , R6 , R7 , R8 , R9, m, ring B, and n are as defined in claim 1 and wherein R7 and R6 can cyclize to form a 4-6 membered heterocyclic ring.

14. A compound of the formula (II)

wherein

of the formula (II) is chosen from column Al - A42 in the table, and of the formula (II) is chosen from column Bl - B22 in the table, with the proviso that when column B is B3, B 13 or B22, then A must be A20, A23, A38, or A42

or a pharmaceutically acceptable salt thereof.

15. A compound chosen from

or a pharmaceutically acceptable salt thereof.

16. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to claims 1 or 15 and one or more pharmaceutically acceptable carriers and/or adjuvants.

17. A method of treating pain comprising administering a therapeutically effective amount of a compound according to claims 1 or 15.

18. A method of treating a disease or condition chosen from a lung disease, a rheumatic disease, an autoimmune disease, a musculoskeletal disease, an allergic disease, an allergic reaction, a vascular disease, a dermatological disease, a renal disease, a hepatic disease, a gastrointestinal disease, neurodegeneration eye disease, diseases of the ear, nose, and throat, neurological disease blood disease, tumors, endocrine diseases, organ and tissue transplantations and graft-versus-host diseases, severe states of shock, acute pain, visceral pain, spasm of the gastrointestinal tract or uterus, colics, neuropathic pain, inflammatory and nociceptive pain, cancer pain, headache, restenosis, atherosclerosis, reperfusion injury, congestive heart failure, myocardial infarction, thermal injury, multiple organ injury secondary to trauma, necrotizing enterocolitis and syndromes associated with hemodialysis, leukopheresis, and granulocyte transfusion, sarcoidosis, gingivitis and pyrexia comprising administering a therapeutically effective amount of a compound according to claims 1 or 15 or 15.

Description:
Compounds Which Selectively Modulate The CB2 Receptor

Application Data

This application claims benefit to US provisional application serial no. 61/186920 filed June 15, 2009.

BACKGROUND OF THE INVENTION

1. TECHNICAL FIELD

The present invention relates to novel compounds which modulate the CB2 receptor and their use as medicaments.

2. BACKGROUND INFORMATION

WO2008014199, WO2008039645 discuss the CB2 receptor, and the therapeutic uses of the CB2 receptor agonist compounds disclosed therein. It is believed that the highly selective activation of the CB2 receptor with an agonist may offer avenues of harnessing the beneficial effects while avoiding the adverse effects seen with dual CB1/CB2 cannabinoid receptor agonists (see e.g. Expert Opinion on Investigational Drugs (2005), 14(6), 695-703). It is desirable therefore to provide agonists of CB2 with minimized CB 1 activity.

PCT Application Number PCT/US 09/34464 discloses compounds having CB2 agonist activity. The compounds of the present invention differ structurally from the above disclosed compounds, for example the present R5 in the formula (I) disclosed hereinbelow. Additionally, the compounds of the present invention have lower CB 1 activity than the compounds disclosed in the cited art. BRIEF SUMMARY OF THE INVENTION

The present invention provides novel compounds which bind to and modulate the CB2 receptor and have lower CBl receptor activity. The invention also provides methods and pharmaceutical compositions for treating inflammation by way of the administration of therapeutic amounts of the compounds of the invention. Lastly, the invention provides a method and pharmaceutical compositions for treating pain by way of the administration of therapeutic amounts of the compounds of the invention.

BRIEF DESCRIPTION OF THE FIGURES

Fig. 1: Flow rate graph time vs %organic (B) using evaporative light scattering detectors. Fig. 2: Flow rate graph time vs %organic (B) using HPLC-MS equipment.

DETAILED DESCRIPTION OF THE INVENTION

In the broadest generic embodiment 1, the invention provides compounds of the formula

wherein:

ring A is a 5-membered heteroaryl ring;

R 1 is hydrogen, C 1-10 alkyl or C 3-10 cycloalkyl which is optionally substituted with 1-3 C 1 . io alkyl, each R 1 or it's substituent is optionally halogenated; R 2 is C 1-10 alkyl, C 3-10 cycloalkyl, arylsulfonyl, arylcarbonyl, C 1-10 acyl, C 3-10 cycloalkylcarbonyl, heterocyclylcarbonyl, heteroarylcarbonyl, heterocyclyl, benzyl, phenethyl, aryl or heteroaryl each optionally independently substituted with 1 to 3 substituents chosen from C 1-6 alkyl, C 3-10 cycloalkyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1-6 alkylsulfonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylamino, C 3-6 cycloalkylamino, C 1-6 dialkylamino, C 1-6 alkylaminocarbonyl, C 1-6 acylamino, C 1-6 dialkylaminocarbonyl, hydroxyl, halogen, cyano, nitro, oxo, heterocyclyl, aryl and heteroaryl, each substituent on R 2 where possible is optionally halogenated or substituted with 1 to 3 C 1-6 alkyl, C 1-6 acyl, C 1-6 alkyl sulfonyl, cyano, aryl, oxo or hydroxyl; or R 1 and R 2 together with the nitrogen atom to which they are attached form a monocyclic, bicyclic or spirocyclic heterocycle or monocyclic or bicyclic heteroaryl ring each optionally substituted with 1 to 3 C 1-6 alkyl, C 1-6 alkoxy, C 1-6 acyl, C 1-6 alkyl sulfonyl, cyano, aryl, oxo, hydroxyl or halogen each ring substituent being further optionally halogenated where possible;

R 3 and R 3 are independently hydrogen or C 1-6 alkyl optionally halogenated with the proviso that R 3 and R 3 cannot simultaneously be hydrogen; or R 3 and R 3 together with the carbon atom to which they are attached form a 3- to 6-membered cycloalkyl or heterocyclic ring each optionally halogenated;

R 4 is hydrogen or methyl;

R 5 is chosen from

m is 0, 1, 2 or 3

R 6 is hydrogen, C 1-4 alkyl or C 1-4 alkoxy

R 7 and R 8 are each independently hydrogen, or C 1-4 alkyl with the proviso that both R 7 and R 8 cannot be hydrogen; and wherein R 7 and R 8 optionally can cyclize to form a C 3 - 7 cycloalkyl ring;

R ,9 is C 1-6 alkyl or aryl; ring B is a 5-6 membered heterocyclic ring;

n is O, 1 or 2;

wherein any carbon atom on the formula (I) or any R substituent listed above is optionally partially or fully halogenated where possible;

or a pharmaceutically acceptable salt thereof. In another embodiment 2, the invention provides compounds of the formula (I) according to the embodiment immediately above, and wherein ring A is

R 2 is C 1-6 alkyl, C 3 _ 7 cycloalkyl, I,l-dioxo-lλ 6 -thiomorpholinylcarbonyl, phenylsulfonyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl or piperidinyl, each optionally independently substituted with 1 to 3 substituents chosen from C 1 - 5 alkyl, C 3 - 7 cycloalkyl, C 1 - 5 alkoxy, C 1 - 5 alkylthio, C 1 - 5 alkylsulfonyl, C 1 - 5 alkoxycarbonyl, C 1 - 5 alkylamino, C 3 - 6 cycloalkylamino, C 1 - 5 dialkylamino, C 1 - 5 alkylaminocarbonyl, C 1 - 5 acylamino, C 1 - 5 dialkylaminocarbonyl, hydroxyl, halogen, cyano, nitro, oxo, phenyl and heterocyclyl chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, 1,1-dioxo-lλ 6 - thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl and piperazinyl, each R 2 substituent where possible is optionally halogenated or substituted with 1 to 3 C 1 - 5 alkyl, C 1 - 5 acyl, methyl sulfonyl, cyano, phenyl, oxo or hydroxyl;

or R 1 and R 2 together with the nitrogen atom to which they are attached form a ring chosen from thiomorpholinyl, I,l-dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, benzimidazolyl, pyrazolyl, imidazolyl, triazinyl, indazolyl, indolyl, indolinyl, isoindolyl, isoindolinyl, and 2-aza-spiro[4.5]dec-2-yl, l-aza-spiro[4.5]dec-1-yl, 1- aza-spiro[4.4]non-1-yl, 2-aza-spiro[4.4]non-2-yl, 2-aza-spiro[5.5]undec-2-yl, 1-aza- spiro[5.5]undec-1-yl each optionally substituted with 1 to 3 C 1-6 alkyl, C 1-6 alkoxy, C 1-6 acyl, C 1-6 alkyl sulfonyl, cyano, phenyl, oxo, hydroxyl and halogen each ring substituent being further optionally halogenated where possible;

R 3 and R 3 are each methyl or ethyl, each optionally halogenated, or R 3 and R 4 together with the carbon atom to which they are attached form a cyclopropyl or cyclobutyl ring each optionally halogenated.

In another embodiment 3, the invention provides compounds of the formula (I) according to the embodiment described immediately above, and wherein ring A is

.

R 1 is hydrogen or C 1-3 alkyl optionally halogenated;

R 2 is C 1-6 alkyl or C 3 - 7 cycloalkyl optionally independently substituted with 1 to 3 halogen, one C 3 - 7 cycloalkyl, C 1-6 alkoxy or one heterocyclyl chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, I,l-dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl and piperazinyl each optionally halogenated or substituted with C 1-4 alkyl or methyl sulfonyl;

or R 1 and R 2 together with the nitrogen atom to which they are attached form a ring chosen form thiomorpholinyl, I,l-dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, azetidinyl, indolyl, indolinyl, isoindolyl, isoindolinyl, and 2-aza-spiro[4.5]dec-2-yl each optionally substituted with 1 to 3 C 1-4 alkyl, C 1-4 alkoxy and halogen each ring substituent being further optionally halogenated where possible; R 3 and R 3 are each methyl or ethyl or R 3 and R 4 together with the carbon atom to which they are attached form a cyclopropyl or cyclobutyl ring;

R 6 is hydrogen or C 1-2 alkyl; R and R are each C 1-2 alkyl.

In another embodiment 4, the invention provides compounds of the formula (I) according to the embodiment immediately above, and wherein ring A is

R 2 is methyl optionally substituted with one heterocyclyl chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, I,l-dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl and piperazinyl each optionally halogenated or substituted with C 1-4 alkyl or methyl sulfonyl;

or R 1 and R 2 together with the nitrogen atom to which they are attached form a ring chosen form thiomorpholinyl, I,l-dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, azetidinyl, indolyl and 2-aza-spiro[4.5]dec-2-yl each optionally substituted with 1 to 3 C 1-3 alkyl, C 1-3 alkoxy and halogen each ring substituent being further optionally halogenated where possible.

In another embodiment 5, the invention provides compounds of the formula (I) according to the embodiment 1, and wherein

ring A is

R 1 is hydrogen, C 1-6 alkyl or C 3 - 7 cycloalkyl optionally substituted with 1-3 C 1-6 alkyl, each R 1 or it's substituent is optionally halogenated;

R 2 is cyclopentyl, cyclohexyl, tetrahydropyranyl, tetrahydrofuranyl, thiomorpholinylcarbonyl, I,l-dioxo-lλ 6 -thiomorpholinylcarbonyl, morpholinylcarbonyl, phenylsulfonyl, phenylcarbonyl, phenyl, pyridinyl, piperidinyl, pyrimidinyl or thiazolyl, each optionally independently substituted with 1 to 3 substituents chosen from C 1 - 5 alkyl, C 3 - 5 cycloalkyl, C 1 - 5 alkoxy, C 1 - 5 alkylthio, C 1 - 5 alkylsulfonyl, C 1 - 5 alkoxycarbonyl, C 1 - 5 alkylamino, C 3 - 6 cycloalkylamino, C 1 - 5 dialkylamino, C 1 - 5 alkylaminocarbonyl, C 1 - 5 acylamino, C 1 - 5 dialkylaminocarbonyl, hydroxyl, halogen, cyano and nitro, each R 2 substituent where possible is optionally halogenated or substituted with 1 to 3 C 1 - 5 alkyl or C 1 - 5 alkyl sulfonyl, or R 1 and R 2 together with the nitrogen atom to which they are attached form a ring chosen from tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, thiomorpholinyl, 1,1-dioxo-lλ 6 - thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, piperazinyl, azetidinyl, benzimidazolyl, pyrazolyl, imidazolyl, triazinyl, indazolyl, indolyl, indolinyl, isoindolyl, isoindolinyl, and 2-aza-spiro[4.5]dec-2-yl, l-aza-spiro[4.5]dec-1-yl, l-aza-spiro[4.4]non-1-yl, 2-aza-spiro[4.4]non-2-yl, 2-aza-spiro[5.5]undec-2-yl, l-aza-spiro[5.5]undec-1-yl each optionally substituted with 1 to 3 C 1-6 alkyl, C 1-6 alkoxy, C 1-6 acyl, C 1-6 alkyl sulfonyl, cyano, phenyl, oxo, hydroxyl or halogen each ring substituent being further optionally halogenated where possible; R 3 and R 3 are each methyl optionally halogenated , or R 3 and R 3 together with the carbon atom to which they are attached form a cyclopropyl or cyclobutyl ring each optionally halogenated;

R is hydrogen;

R 5 is chosen from

R is hydrogen or C 1-3 alkyl; wherein R 7 and R 8 are each C 1-3 alkyl or C 3 _ 6 cycloalkyl.

In another embodiment 6, the invention provides compounds of the formula (I) according to the embodiment 3, and wherein ring A is

R 3 and R 3 are methyl;

R is hydrogen or C 1-2 alkyl; wherein R and R are each C 1-2 alkyl.

In another embodiment 7, the invention provides compounds of the formula (I) according to the embodiment 6, and wherein ring A is

R 1 and R 2 together with the nitrogen atom to which they are attached form a ring chosen from thiomorpholinyl, I,l-dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyrrolidinyl, piperidinyl, azetidinyl, indolyl and 2-aza-spiro[4.5]dec-2-yl each optionally substituted with 1 to 3 C 1-3 alkyl, C 1-3 alkoxy, hydroxy and halogen each ring substituent being further optionally halogenated where possible.

In another embodiment 8, the invention provides compounds of the formula (I) according to the embodiment described immediately above, and wherein

ring A is

In another embodiment 9, the invention provides compounds of the formula (I) according to the embodiment described immediately above, and wherein

R 1 and R 2 together with the nitrogen atom to which they are attached form a ring chosen from morpholinyl, pyrrolidinyl and piperidinyl, each optionally substituted with 1 to 3 C 1-3 alkyl.

In another embodiment 10, the invention provides compounds of the formula (I) according to embodiment 2, and wherein R 2 is cyclopropyl, cyclohexyl, tetrahydropyranyl, pyrrolidinyl or piperidinyl, each optionally independently substituted with 1 to 2 substituents chosen from Cr 3 alkyl and Cr 2 alkylsulfonyl.

In another embodiment 11, the invention provides compounds of the formula (I) according to embodiment 2, and wherein

R 2 is CF 3 -CH 2 -CH 2 -CH 2 -, CH 3 -CH 2 -CH 2 (CH 3 )-, cyclopropyl-CH 2 -, hydroxylcyclohexyl, tetrahydrofuranyl-CH 2 - ; ring A is

R 5 is wherein wherein R and R optionally can cyclize to form a C 3 _ 7 cycloalkyl ring.

In another embodiment 12, the invention provides compounds of the formula (I) according to embodiment 2, and wherein

R 2 hydroxylcyclohexyl, tetrahydropyranyl, hydroxylpyrrolidinyl or methylsulfonylpyrrolidinyl;

ring A is R 5 is

in wherein R 7 and R 8 optionally can cyclize to form a C 3 - 7 cycloalkyl ring;

R 1 and R 2 optionally can cyclize to form piperidinyl, methyl sulfonylpiperidinyl.

In another aspect of the invention there is provided a second set of generic embodiments, wherein for each of the above embodiments 1-10 of the formula (I), R 7 and R 6 can cyclize to form a 4-6 membered heterocyclic ring.

In another embodiment there is provided a compound of the formula (II)

wherein

of the formula (I) is chosen from column Al - A42 in Table I, and of the formula (I) is chosen from column Bl - B22 in Table I, with the proviso that when column B is B3, B 13 or B22, then A must be A20, A23, A38, or A42

Table I

In another embodiment, the invention provides made compounds in Table II which can be made in view of the general schemes, examples and methods known in the art.

or a pharmaceutically acceptable salt thereof.

Of the above compounds, the following are preferred CB2 agonists: Table III

In all the compounds disclosed hereinabove in this application, in the event the nomenclature is in conflict with the structure, it shall be understood that the compound is defined by the structure.

The invention also relates to pharmaceutical preparations, containing as active substance one or more compounds of formula (I), or the pharmaceutically acceptable derivatives thereof, optionally combined with conventional excipients and/or carriers.

Compounds of the invention also include their isotopically-labelled forms. An isotopically- labelled form of an active agent of a combination of the present invention is identical to said active agent but for the fact that one or more atoms of said active agent have been replaced by an atom or atoms having an atomic mass or mass number different from the atomic mass or mass number of said atom which is usually found in nature. Examples of isotopes which are readily available commercially and which can be incorporated into an active agent of a combination of the present invention in accordance with well established procedures, include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, e.g., 2 H, 3H, 13 C, 14 C, 15 N, 18 O, 17 0, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively. An active agent of a combination of the present invention, a prodrug thereof, or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other isotopes of other atoms is contemplated to be within the scope of the present invention. The invention includes the use of any compounds of described above containing one or more asymmetric carbon atoms may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Isomers shall be defined as being enantiomers and diastereomers. All such isomeric forms of these compounds are expressly included in the present invention. Each stereogenic carbon may be in the R or S configuration, or a combination of configurations.

Some of the compounds of formula (I) can exist in more than one tautomeric form. The invention includes methods using all such tautomers.

All terms as used herein in this specification, unless otherwise stated, shall be understood in their ordinary meaning as known in the art. For example, "C 1 _ 4 alkoxy" is a C 1-4 alkyl with a terminal oxygen, such as methoxy, ethoxy, propoxy, butoxy. All alkyl, alkenyl and alkynyl groups shall be understood as being branched or unbranched where structurally possible and unless otherwise specified. Other more specific definitions are as follows:

Carbocycles include hydrocarbon rings containing from three to twelve carbon atoms. These carbocycles may be either aromatic either aromatic or non-aromatic ring systems. The non- aromatic ring systems may be mono- or polyunsaturated. Preferred carbocycles include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptanyl, cycloheptenyl, phenyl, indanyl, indenyl, benzocyclobutanyl, dihydronaphthyl, tetrahydronaphthyl, naphthyl, decahydronaphthyl, benzocycloheptanyl and benzocycloheptenyl. Certain terms for cycloalkyl such as cyclobutanyl and cyclobutyl shall be used interchangeably.

The term "heterocycle" refers to a stable nonaromatic 4-8 membered (but preferably, 5 or 6 membered) monocyclic or nonaromatic 8-11 membered bicyclic heterocycle radical which may be either saturated or unsaturated. Each heterocycle consists of carbon atoms and one or more, preferably from 1 to 4 heteroatoms chosen from nitrogen, oxygen and sulfur. The heterocycle may be attached by any atom of the cycle, which results in the creation of a stable structure.

The term "heteroaryl" shall be understood to mean an aromatic 5-8 membered monocyclic or 8-11 membered bicyclic ring containing 1-4 heteroatoms such as N,0 and S.

Unless otherwise stated, heterocycles and heteroaryl include but are not limited to, for example benzoxazolyl, benzothiazolyl, benzimidazolyl, tetrahydropyranyl, dioxanyl, tetrahydrofuranyl, oxazolyl, isoxazolyl, thiazolyl, pyrazolyl, pyrrolyl, imidazolyl, thienyl, thiadiazolyl, triazolyl, thiomorpholinyl, l,l-Dioxo-lλ 6 -thiomorpholinyl, morpholinyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, pyrrolidinyl, piperidinyl, piperazinyl, purinyl, quinolinyl, Dihydro-2H-quinolinyl, isoquinolinyl, quinazolinyl, indazolyl, thieno[2,3-d]pyrimidinyl, indolyl, isoindolyl, benzofuranyl, benzopyranyl and benzodioxolyl.

The term "heteroatom" as used herein shall be understood to mean atoms other than carbon such as O, N, S and P.

In all alkyl groups or carbon chains one or more carbon atoms can be optionally replaced by heteroatoms: O, S or N, it shall be understood that if N is not substituted then it is NH, it shall also be understood that the heteroatoms may replace either terminal carbon atoms or internal carbon atoms within a branched or unbranched carbon chain. Such groups can be substituted as herein above described by groups such as oxo to result in defintions such as but not limited to: alkoxycarbonyl, acyl, amido and thioxo.

The term "aryl" as used herein shall be understood to mean aromatic carbocycle or heteroaryl as defined herein. Each aryl or heteroaryl unless otherwise specified includes it's partially or fully hydrogenated derivative. For example, quinolinyl may include decahydroquinolinyl and tetrahydroquinolinyl, naphthyl may include its hydrogenated derivatives such as tetrahydronaphthyl. Other partially or fully hydrogenated derivatives of the aryl and heteroaryl compounds described herein will be apparent to one of ordinary skill in the art.

As used herein, "nitrogen" and "sulfur" include any oxidized form of nitrogen and sulfur and the quaternized form of any basic nitrogen. . For example, for an -S-C 1-6 alkyl radical, unless otherwise specified, this shall be understood to include -S(O)-C 1-6 alkyl and -S(O) 2 -C 1-6 alkyl.

The term "halogen" as used in the present specification shall be understood to mean bromine, chlorine, fluorine or iodine, preferably fluorine. The definitions "partially or fully halogenated"; partially or fully fluorinated; "substituted by one or more halogen atoms", includes for example, mono, di or tri halo derivatives on one or more carbon atoms. For alkyl, a nonlimiting example would be -CH 2 CHF 2 , -CF 3 etc.

The compounds of the invention are only those which are contemplated to be 'chemically stable' as will be appreciated by those skilled in the art. For example, a compound which would have a 'dangling valency', or a 'carbanion' are not compounds contemplated by the inventive methods disclosed herein.

The invention includes pharmaceutically acceptable derivatives of compounds of formula (I). A "pharmaceutically acceptable derivative" refers to any pharmaceutically acceptable salt or ester, or any other compound which, upon administration to a patient, is capable of providing (directly or indirectly) a compound useful for the invention, or a pharmacologically active metabolite or pharmacologically active residue thereof. A pharmacologically active metabolite shall be understood to mean any compound of the invention capable of being metabolized enzymatically or chemically. This includes, for example, hydroxylated or oxidized derivative compounds of the formula (I). Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfuric, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfuric and benzenesulfonic acids. Other acids, such as oxalic acid, while not themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds and their pharmaceutically acceptable acid addition salts. Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N-(C j -C4 alkyl)4 + salts.

In addition, within the scope of the invention is use of prodrugs of compounds of the formula (I). Prodrugs include those compounds that, upon simple chemical transformation, are modified to produce compounds of the invention. Simple chemical transformations include hydrolysis, oxidation and reduction. Specifically, when a prodrug is administered to a patient, the prodrug may be transformed into a compound disclosed hereinabove, thereby imparting the desired pharmacological effect.

The compounds of formula I may be made using the general synthetic methods described below, which also constitute part of the invention.

GENERAL SYNTHETIC METHODS

The invention also provides processes for making compounds of Formula (I) or Formula (II). In all Schemes, unless specified otherwise, R 1 R 2 R 3 R 3 R 4 , R 5 and A in the Formulas below shall have the meaning of R 4 , R 5 and A in Formula (I) or Formula (II) of the invention described herein above.

Optimum reaction conditions and reaction times may vary depending on the particular reactants used. Unless otherwise specified, solvents, temperatures, pressures, and other reaction conditions may be readily selected by one of ordinary skill in the art. Specific procedures are provided in the Synthetic Examples section. Typically, reaction progress may be monitored by thin layer chromatography (TLC), if desired, and intermediates and products may be purified by chromatography on silica gel and/or by recrystallization. The examples which follow are illustrative and, as recognized by one skilled in the art, particular reagents or conditions could be modified as needed for individual compounds without undue experimentation. Starting materials and intermediates used, in the Schemes below, are either commercially available or easily prepared from commercially available materials by those skilled in the art.

The compounds of Formula (I) or Formula (II) may be synthesized according to Scheme 1:

Scheme 1

As illustrated in scheme 1, reaction of an acid of formula (III) with reagents such as thionyl chloride or oxalyl chloride provides the corresponding acid chloride of formula (IV). Reaction of this acid chloride (IV) with an amine of formula (V), in a suitable solvent, in the presence of a suitable base such as N,N-diisopropylethylamine, provides an amide of formula (VI). Alternatively, the acid of formula (III) above may also be coupled with the corresponding amine of formula (V), under standard coupling conditions, to provide an amide of formula (VI). Standard peptide coupling reactions known in the art (see for example M. Bodanszky, 1984, The Practice of Peptide Synthesis, Springer- Verlag) may be employed in these syntheses. An example of suitable coupling conditions is treatment of a solution of the carboxylic acid in a suitable solvent such as DMF with EDC, HOBT, and a base such as diisopropylethylamine, followed by the desired amine.

Reaction of the intermediate of formula (VI) with an amine of formula (VII), in a suitable solvent, in the presence of a suitable base such as cesium carbonate, provides a compound of Formula (I) or Formula (II) .

The compounds of Formula (I) or Formula (II) may be prepared according to Scheme 2:

Scheme 2

As outlined in scheme 2, reaction of an amide of formula (VI) with an amine of formula (VIII), in a suitable solvent, in the presence of a suitable base such as cesium carbonate, provides an amine of formula (IX). Reaction of the intermediate amine of formula (IX) with an acid chloride of formula (Xa) or a sulfonyl chloride of formula (Xb), in a suitable solvent, in the presence of a suitable base, provides a compound of Formula (I) or Formula (II) wherein R 2 = R-C(O)- or R-SO 2 -

The compounds of Formula (I) or Formula (II) may be synthesized as outlined in Scheme 3:

Scheme 3

As illustrated in scheme 3, reaction of an amine of formula (IX) with a compound of formula (XI), in a suitable solvent, in the presence of a suitable base such as cesium carbonate, provides a compound of Formula (I) or Formula (II)

Alternatively, reaction of an amine of formula (IX) with a compound of formula (XII) wherein

Hal = bromo or iodo, in a suitable solvent, in the presence of a suitable base such as sodium hydride, provides a compound of Formula (I) or Formula (II) .

The compounds of Formula (I) or Formula (II) may be synthesized as shown in Scheme 4:

Scheme 4 As shown in scheme 4, reaction of an amino acid of formula (XIII) with an aldehyde of formula (XIV), under standard reductive amination conditions, provides an amine of formula (XV). Further reaction of amine of formula (XV) with an aldehyde of formula (XVI) provides the corresponding amine of formula (XVII). Coupling the intermediate of formula (XVII) with an amine of formula (V), as in scheme 1, provides a compound of Formula (I) or Formula (II) wherein R 1 = R a - CH 2 - and R 2 = R b -CH 2 -

The compounds of Formula (I) or Formula (II) may be synthesized according to Scheme 5:

Scheme 5

As illustrated above in scheme 5, reaction of a bromo ester of formula (XVIII) with an amine of formula (VIII), in a suitable solvent, provides the corresponding N-substituted product of formula (XIX). Reaction of the compound (XIX) with a halide of formula (XII), in a suitable solvent, in the presence of a suitable base, provides a tertiary amine of formula (XX). Ester hydrolysis under standard conditions provides an acid of formula (XXI). Coupling the acid formula (XXI) with an amine of formula (V), as in scheme 1, provides a compound of Formula (I) or Formula (II). Further modification of the initial product of Formula (I) or Formula (II), by methods known in the art and illustrated in the Examples below, may be used to prepare additional compounds of this invention.

EXAMPLES:

Method A

Synthesis of Example 1

Step 1: Synthesis of compound 2

To a flask containing 2-bromo-2-methyl-propionic acid (1) (0.6 g, 3.5 mmol) under nitrogen is added thionyl chloride (2 mL). The reaction mixture is heated to 70 °C where it is maintained for 2 h. After this time, the reaction is cooled to room temperature and concentrated under reduced pressure. The crude acid chloride is used without further purification.

The crude acid chloride is dissolved in dichloromethane (DCM) (5 mL) and N,N- diisopropylethylamine (1 mL, 5.7 mmol) is added followed by 3-(2-methoxy-l,l-dimethyl- ethyl)-isoxazol-5-ylamine (0.6 g, 3.5 mmol). The reaction is stirred for 16 h at room temperature. After this time, additional 2-bromo-2- methyl-propionic acid (0.3 g, 1.7 mmol) is activated as its acid chloride, as described above) and added to the reaction mixture. After further 1.5 h stirring, the mixture is washed with saturated aqueous NaHCO 3 solution (x 2), brine and the organic layer is separated, dried (MgSO 4 ) and concentrated under reduced pressure. The crude product is purified by chromatography (silica, eluent: heptanes, 20% ethyl acetate) to provide compound 2 (0.6 g,

51%);

According to the above procedure the following intermediates are synthesised: Table 1:

Step 2: Synthesis of example 1

To a solution of 4-aminotetrahydropyran (0.1 g, 0.94 mmol) in THF (2 mL) are added Cs 2 CO 3 (0.6 g, 1.84 mmol) and compound 2 (0.3 g, 0.94 mmol). The reaction is heated to 50 °C for 3 h. The solvent is then removed under reduced pressure and the residue is partitioned between DCM and IM aqueous HCl solution. The acidic aqueous layer is basified with 5N aqueous NaOH solution and extracted with DCM (3 x 10 mL). The combined organic extracts are washed with brine, dried (MgSO 4 ), filtered and concentrated under reduced pressure. The crude product is purified by column chromatography (silica, eluent: heptanes, 50% ethyl acetate) to yield example 1 (0.15 g, 41%). LC-MS (LC Method a): retention time: 2.40 min, m/z 340 [M+H]

Compounds listed in Table IV under method A are made following this procedure. Method B

Synthesis of Example 2

Example 2 Step 1: Synthesis of compound 3

To a flask containing compound 1 (3 g, 18 mmol) under nitrogen is added thionyl chloride (6.6 rnL). The reaction mixture is heated to 70 °C where it is maintained for 3 h. After this time, the reaction is cooled to room temperature and concentrated under reduced pressure. The crude acid chloride is used without further purification.

The crude acid chloride is dissolved in THF (11 mL) and N,N-diisopropylethylamine (3.1 mL , 18 mmol) is added, followed by 3-[l,l-dimethyl-2-(tetrahydro-pyran-2-yloxy)-ethyl]- isoxazol-5-ylamine (4.3 g, 18 mmol). The reaction is stirred for 16 h at 40 °C. The mixture is washed with saturated aqueous NaHCO 3 solution (x 2), brine and the organic layer is separated, dried (MgSO 4 ) and concentrated under reduced pressure. Purification of the residue by chromatography (silica, eluent: heptanes, 20% ethyl acetate) allows separation and isolation of compound 3 (1 g, 18%), m/z 305/307 [M+H + ], from compound 4 (0.9 g, 13%), LC-MS (LC Method b): retention time: 2.35 min, m/z 411/413 [M+Na+H]. Step 2: Synthesis of example 2

To a solution of 4-aminotetrahydropyran (0.05 g, 0.5 mmol) in THF (3 niL) are added Cs 2 CO 3 (0.3 g, 1 mmol) and compound 3 (0.15 g, 0.5 mmol). The reaction is heated to 50 °C for 18 h. The solvent is then removed under reduced pressure and the residue is partitioned between DCM and IM aqueous HCl solution. The acidic aqueous layer is basified with 5N aqueous NaOH solution and extracted with DCM (3 x 10 mL). The combined organic extracts are washed with brine, dried (MgSO 4 ), filtered and concentrated under reduced pressure. The crude product is purified twice by column chromatography (silica, eluent: heptanes, 20% EtOAc) followed by trituration with IM HCl in dioxane to yield the hydrochloride salt of example 2 (0.04 g, 22%). LC-MS (LC Method a): retention time: 2.16 min, m/z 326

[M f c ebase +H]

Compounds listed in Table IV under method B are made following this procedure.

Method C

Synthesis of Example 3

Step 1: Synthesis of compound 4

To a flask containing compound 1 (20.85 g, 125 mmol) in DCM (150 mL) under nitrogen is added oxalyl chloride (16.2 mL, 187.3 mmol), followed by DMF (5 drops). The reaction mixture is stirred at room temperature for 16 h and then concentrated under reduced pressure. The crude acid chloride, used without further purification, is dissolved in toluene (50 mL) and added dropwise over 10 min to a solution of N,N- diisopropylethylamine (32.6 mL , 187 mmol) and 3-[l,l-dimethyl-2-(tetrahydro-pyran-2- yloxy)-ethyl]-isoxazol-5-ylamine (15 g, 62.4 mmol) in toluene (300 mL) at 50 °C. The reaction is stirred for 4 h at 60 °C. The reaction mixture is concentrated under reduced pressure and the residue is partitioned between DCM (300 mL) and water (250 mL). The organic layer is separated and dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude orange oil is dissolved in diethyl ether and precipitated by addition of n- heptanes. The precipitate is isolated by filtration and washed sparingly with diethyl ether/heptanes to yield the first crop (9 g) of compound 4 as off-white solid. The mother liquor is concentrated and a second crop (3.3 g) is isolated by crystallisation/precipitation from diethyl ether/heptanes. The mother liquor is concentrated under reduced pressure and the residual orange oil filtered through silica (eluent: heptanes, 20% ethyl acetate), the filtrate is concentrated. The residue is dissolved in diethyl ether and the third crop (3.1 g) is isolated by diethyl ether/ heptanes treatment as described above. The combined yield of crop 1-3 is 15.4 g. Yield 63%; LC-MS (LC Method b): retention time: 2.35 min, m/z 411/413 [M+Na+H].

Step 2: Synthesis of compound 5

A solution of N-methyl-(tetrahydro-pyran-4-yl)-amine (0.25 g, 2.17 mmol), Cs 2 CO 3 (1.41 g, 4.34 mmol) and compound 4 (0.85 g, 2.17 mmol) in THF (20 mL) is stirred at room temperature for 72 h. The reaction mixture is filtered through Celite ®, the solids are washed with ethyl acetate and DCM and the filtrate is concentrated under reduced pressure. The residue is purified by column chromatography (silica, eluent: heptanes, 50-100% ethyl acetate) to yield 374 mg of compound 5. Yield 41%;. LC-MS (LC Method b): retention time: 1.51 min, m/z 424 [M+H] The following amines are synthesised by adaptation of the above procedure with the following modification to be noted:

For intermediate II and III: excess of the corresponding amine (2.5 equ) and Cs 2 CO 3 (2 equ.) are used, the work-up procedure includes a wash of the organic layer with 10 % aqueous citric acid solution followed by brine and the product is isolated by column chromatography (silica, eluent heptanes, 30% ethyl acetate).

For intermediate XI the reaction is stirred for 48 h at ambient temperature.

For intermediate XII and X: the reaction mixture is filtered through celite and washed with

DCM. The solvent is removed under reduced pressure and the product purified by column chromatography (silica, eluent; heptanes, 0 - 50% ethyl acetatec)

According to the above procedure the following intermediates are synthesised: Table 2:

a) n. . nterme ate s carr e t roug to t e next step w t out urt er pur cat ona) Interme ate VI: 1 H NMR (400 MHz, chloroform-d) δ ppm 1.26 (6H, s), 1.34 (3H, s), 1.37 (3H, s), 1.47-1.83 (HH, m), 2.18 (2H, d, 7.2 Hz), 2.22 (3H, s), 3.36-3.50 (3H, m), 3.77-3.82 (3H, m), 3.99 (2H, m), 4.57 (1H, m), 6.32 & 6.38 (1H, 2s), 9.4-9.8 (1H, br) b) Intermediate VII: 1 H NMR (400 MHz, chloroform-d) δ ppm 1.07 (6H, d, J=6.4 Hz), 1.32-1.37 (12H, m), 1.46-1.84 (6H, m), 2.19 (3H, s), 2.98 (1H, q), 3.37 (1H, d, J=9.6 Hz), 3.45-3.50 (1H, m), 3.76-3.81 (2H, m), 4.57 (1H, t, J=6.8 Hz), 6.32 (1H, s), 10.10 (1H, br) c) Intermediate VIII: 1 H NMR (400 MHz, chloroform-d) δ ppm 1.25 (12H, s), 1.31-1.76 (1OH, m), 2.70 (2H, t, 11.2 Hz), 2.80 (3H, s), 3.31-3.42 (6H, m), 3.62-3.66 (2H, m), 4.53 (1H, s), 6.25 (1H, s), 6.80 (1H, br) Step 3: Synthesis of example 3

To a solution of compound 5 (806 mg, 1.9 mmol) in DCM/ethanol (1/1, 16 niL) is added polymer- supported tosic acid (MP-TsOH (65), loading 3.3 mmol/g, 1.74 g). The mixture is shaken on an orbital- shaker at room temperature for 18 h. The resin is isolated by filtration and washed with DCM and methanol. The product is then released form the resin with 7M solution of ammonia in methanol. The resin is further rinsed with methanol and DCM. Concentration of the basic filtrate gives a clear oil which is triturated with diethyl ether/heptanes to afford 549 mg of example 3 as a white solid. Yield 85%, LC-MS (LC Method a): retention time: 2.17 min m/z 410 [M+H];, mp 129-131 °C.

Compounds listed in Table IV under method C are made following this procedure, with the following modification for examples 7-9, and 20: Pyridinium p-toluenesulfonate (1.2 equ) is used instead of MP-TsOH and the reaction is performed under refluxing conditions (4 h) in EtOH instead of DCM/ethanol.

Example 86-87 are separated by chiral preparative LC.

Example 4 and 5 are converted into the oxalate salt by stirring in the presence of oxalic acid (1 equ) in ethanol at 50 °C, followed by concentration under reduced pressure.

Examples 42, 43, 45, 47, 48, 49, 51, 53, 56, 57, 60, 63, 64, 68, 86, 87 are converted converted into the hydrochloride salt by stirring in the presence of IM HCl solution in diethyl ether (2 eq) at room temperature, followed by concentration under reduced pressure.

Method D

Synthesis of Example 6

A solution of 2-methylpiperidine (40 mg, 0.33 mmol), Cs 2 CO 3 (218 mg, 0.66 mmol) and compound 4 (130 mg, 0.33 mmol) in THF (10 mL) is heated to 55 °C for 3 h in a pressure tube. After cooling to room temperature, the reaction mixture is diluted with ethyl acetate (40 mL) and washed with brine (2 x 15 mL), dried (MgSO 4 ), filtered and concentrated under reduced pressure. The residue is purified by column chromatography (silica, eluent: heptanes, 10% ethyl acetate) and then using a Flash SCX-2 cartridge (Isolute®) (eluent: DCM, methanol then 5% NH 3 /MeOH) which removes the tetrahydropyranyl protecting group. Final purification using silica, eluent: heptanes, 10% ethyl acetate) yield 23 mg of example 6. Yield 22%. LC-MS (LC Method a): retention time: 2.35 min, m/z 324 [M+H]

A portion of example 6 is converted into its hydrochloride salt by stirring in the presence of IM HCl solution in diethyl ether (2 equ.) at room temperature, followed by concentration under reduced pressure.

Compounds listed in Table IV under method D are made following this procedure

Method E

Synthesis of Example 10

Example 12 (80 mg, 0.24 mmol, prepared according to Method A) and 600 mg (2.35 mmol) of aluminium tribromide are dissolved in ethanethiol (4 mL, CAUTION: stench) and stirred at room temperature for 3 h. The reaction is quenched by addition of water (8 mL) and acidified by careful addition of 1 M aqueous HCl solution to pH 4-5. The reaction mixture is extracted with ethyl acetate (3 x 10 mL) and the combined organic layers are washed with brine, dried over Na 2 SO 4 and filtered. The filtrate is concentrated under reduced pressure and the residue purified by column chromatography (silica, eluent: DCM, 50% ethyl acetate) to afford 50 mg of example 10. Yield 65%, LC-MS (LC Method a): retention time: 2.14 min, m/z 326 [M+H]

Compounds listed in Table IV under method E are made following this procedure

Method F

Synthesis of Example 46

To a solution of example 42 (0.34g, 1.01 mmol) and NaH (0.12 g, 3.04 mmol, 60% dispersion in mineral oil,) in THF (6 mL) is added methyl iodide (95 μL, 1.52 mmol). The reaction mixture is stirred at room temperature for 18 h and filtered through celite. The crude product is purified by preparative HPLC (neutral method) to yield 53 mg of example 46 as a colourless oil. Yield: 17%, LC-MS (LC Method a): retention time: 2.60 min, m/z 368 [M+H]. example 46 is converted to the hydrochloride salt by stirring in the presence of 2M solution of hydrogen chloride in diethyl ether (1 mL) at room temperature, followed by concentration under reduced pressure.

Compounds listed in Table IV under method F are made following this procedure

Method G Synthesis of Example 50

Step 1: Synthesis of compound 6

A solution of methylamine (2.89 niL, 23.1 mmol), Cs 2 CO 3 (0.75 g, 2.3 mmol) and compound 4 (0.45 g, 1.2 mmol) in THF (10 mL) is heated to 65 °C in a sealed tube for 3 h. The reaction mixture is cooled and diluted with ethyl acetate (20 mL). The mixture is washed with brine (8 mL). The organic layer is dried (MgSO 4 ), filtered and the filtrate is concentrated under reduced pressure. The crude product is purified by column chromatography (silica, eluent: heptanes, 0-50% ethyl acetate) to yield 0.29 g of compound 6. Yield: 74%; LC-MS (LC Method b): retention time: 1.24 min, m/z 340 [M+H].

Step 2: Synthesis of compound 7

To a solution of compound 6 (0.2 g, 0.59 mmol) and N,N-diisopropylethylamine (0.2 mL, 1.18 mmol) in THF (8 mL) is added (tetrahydro-pyran-4-yl)-acetyl chloride (0.11 g, 0.59 mmol). The reaction is heated to 60 °C for 18 h and the solvent is removed under reduced pressure. The residue is dissolved in ethyl acetate (20 mL) and washed with water (2 x 5 mL) and brine (2 x 5 mL). The organic layer is dried (MgSO 4 ), filtered and the filtrate is concentrated under reduced pressure. The residue is purified by column chromatography

(silica, eluent: heptanes, 0-50% ethyl acetate) to yield 0.25 g of compound 7; yield: 94%, LC-

MS (LC Method b): retention time: 1.90 min, m/z 464 [M-H].

According to the above method the following intermediates are synthesised with the following modifications to be noted: For intermediate XXX and XXXIII, the corresponding acid is converted into its acidchloride by treatment with oxalylchloride (4 equ.) and DMF (cat) in DCM at room temperature for 4 h.

The reaction mixture is concentrated under reduced pressure and the crude acid chloride is used in the above procedure without further purification.

Intermediates XXXI and XXXII are synthesised using standard amide coupling conditions: 1 equ of compound 6 is coupled with the corresponding acid (1 equ) using EDCHCl (3 equ) and

DMAP (1 equ) in acetonitrile. The reaction mixture is stirred at room temperature for 18 h and the concentrated under reduced pressure. The residue is purified by column chromatography

(silica, eluent:n-heptanes, 30% ethyl actetate) to afford the desired intermediate.

According to the above procedure the following intermediates are synthesised: Table 3:

Step 3: Synthesis of example 50

Compound 7 is deprotected by shaking in the presence of polymer-supported tosic acid (MP- TsOH (65), loading 3.3 mmol/g, 0.6 g) on an orbital shaker for 18 h. The resin is removed by filtration and washed with DCM. The product is released from the resin by treatment with 7M ammonia in methanol and the filtrate is concentrated under reduced pressure and the residue is purified by preparative HPLC (neutral method) to afford 40 mg of example 50. Yield 18%, LC-MS (LC Method a): retention time: 3.06 min, m/z 382 [M+H].

Compounds listed in Table IV under method G are made following this procedure

Method H

Synthesis of Example 52

Step 1: Synthesis of compound 9

To a solution of compound 6 (0.2 g, 0.59 mmol, prepared according to Method G, step 1) and Cs 2 CO 3 (0.38 g, 1.18 mmol) in THF (3 mL) is added compound 8 (115 mg, 1.0 mmol, prepared as described in Gosh, A.K. et al. J. Med. Chem. 1993, 36, 2300-2310). The reaction is heated to 65 °C for 3 h in a sealed tube and diluted in ethyl acetate (12 mL). The organic layer is washed with brine (8 mL) and dried (MgSO 4 ). The solvent is removed under reduced pressure and purified by column chromatography (silica, eluent: heptanes, 0-50% ethyl acetate) to afford 0.2 g of compound 9. Yield: 12%; LC-MS (LC Method b): retention time: 1.37 min, m/z 438 [M+H] .

Step 2: Synthesis of Example 52

To a solution of compound 9 in DCM/methanol (1/1, 4 mL) is added polymer-supported tosic acid (MP-TsOH (65), loading 3.3 mmol/g, 0.28 g) and the reaction mixture is shaken on an orbital shaker for 18 h. The resin is removed by filtration and washed with DCM. The product is released from the resin by treatment with 7M ammonia solution in methanol and the resin is further washed with methanol and DCM. The filtrate is concentrated under reduced pressure. The residue is purified by preparative LC (neutral method) to afford 69 mg of example 52 as a clear oil, yield 36%, LC-MS (LC Method a): retention time: 2.19 min, m/z 354 [M+H], which is converted in its hydrochloride salt by treatment with 2M HCl in diethyl ether (0.12 mL) and concentration under reduced pressure. The crude material is recrystallised from ethyl acetate/heptanes to afford the hydrochloride salt of example 52 as a white crystalline solid.

Compounds listed in Table IV under method H are made following this procedure Method I

Synthesis of Example 54

Step 1: Synthesis of compound 10

A solution of (S)-sec-butylamine (0.3 g, 4.1 mmol), Cs 2 CO 3 (2.67 g, 8.2 mmol) and compound 4 (1.59 g, 4.1 mmol) in THF (10 mL) is stirred at room temperature for 18 h. The reaction mixture is heated to 50 °C for 1 h and the reaction is filtered through celite. The solids are washed with DCM and ethyl acetate and the filtrate is concentrated under reduced pressure to afford a yellow oil. This residue is purified by column chromatography (silica, eluent: heptanes, 50% ethyl acetate) to yield 0.65 g of compound 10. Yield: 42%, LC-MS (LC Method b): retention time: 1.38 min, m/z 382 [M+H]. The following intermediates are made according to the above procedure. According to the above procedure the following intermediates are synthesised: Table 4:

Step 2: Synthesis of compound 11

To a solution of the compound 10 (392 mg, 1.02 mmol) in THF (3 niL) is added sodium hydride (41 mg, 1.02 mmol, 60% dispersion in mineral oil) and methyl iodide (64 μl, 1.02 mmol). The reaction mixture is heated to 70 °C for 1 h in a microwave. The reaction mixture is concentrated under reduced pressure and the residue is dissolved in methanol and filtered. The filtrate is concentrated under reduced pressure and the crude material is purified by preparative HPLC (neutral method) to afford 137 mg of compound 11 as a clear oil. Yield: 34%, LC-MS (LC Method b): retention time: 1.25 min, m/z 396 [M+H].

The following intermediates are made according to the above procedure. Table 5:

Step 3: Synthesis of Example 54

To a solution of compound 11 (137 mg, 0.34 mmol) in DCM/methanol (1/1, 10 niL) is added polymer- supported tosic acid (MP-TsOH (65), loading 3.3 mmol/g, 0.31 g). The mixture is shaken on an orbital- shaker at room temperature for 18 h. The resin is isolated by filtration and washed with DCM. The product is then released from the resin with 2M solution of ammonia in methanol. Concentration of the basic filtrate affords 88 mg of example 54. Yield: 63%, LC-MS (LC Method a): retention time: 2.35 min, m/z 312 [M+H].

Compounds listed in Table IV under method I are made following this procedure with the following modifications to be noted:

Example 54, 55, 84, 85 are converted into the hydrochloride salt by stirring in the presence of hydrogen chloride in diethyl ether (2M, 1 mL) at room temperature, followed by concentration under reduced pressure.

Method J

Synthesis of Example 27

Step 1: Synthesis of compound 13 A high pressure vessel on the HEL 100 system containing compound 12 (2.5 g; 24.244 mmol) and tetrahydro-pyran-4-carbaldehyde (5.05 mL; 48.487 mmol) is added palladium hydroxide, 20% on carbon, wet (3.405 g; 10 mol%; 2.424 mmol) and 50 mL of ethanol. The reaction is subjected to 400 psi H 2 at 50 °C for 40 h. After this time, the reaction is cooled to room temperature and filtered through Celite®. The CeIi te® is rinsed with methanol and water.

The combined filtrates are concentrated and slurried in dichloromethane, then filtered to afford compound 13 as a white solid (3.95g; 81%), LC-MS (LC Method h): retention time 0.32 min, m/z 202 [M+H] +

Step 2: Synthesis of compound 14

A high pressure vessel on the HEL 100 system containing compound 13 (Ig; 4.969mmol) and formaldehyde, 37% in water (0.74mL; 9.937mmol) is added palladium hydroxide, 20% on carbon, wet (0.698g; 10mol%; 0.497mmol) and 3OmL of ethanol. The reaction is subjected to 400 psi H 2 at 100 °C for 27.5 h. After this time, the reaction is cooled to room temperature and filtered through Celite®. The Celite® is rinsed with methanol and water. The combined filtrates are concentrated to afford compound 14 as a colorless oil (1.364g; quantitative), LC- MS (LC Method h): retention time 0.32 min, m/z 216 [M+H] +

Step 3: Synthesis of Example 27

To a vial containing compound 14 (0.2g; 0.929 mmol) is added O-(7-azabenzotriazol-1-yl)- ΛWΛ^N'-tetramethyluronium hexafluorophosphate (0.53Og; 1.394 mmol) and N,N- diisopropylethylamine (0.579mL; 3.252mmol). The solution is stirred at room temperature for 30 minutes. To a vial containing 3-[2-(2-methoxy-ethoxy)-l,l-dimethyl-ethyl]-isoxazol-5- ylamine (0.199g; 0.929mmol) is added N,N-dimethylformamide (2mL) and sodium hydride (0.093g; 2.323mmol) at 0 °C. To this solution is added the activated ester. Once addition is complete, the cold bath is removed and the reaction warmed to room temperature and stirred for Ih. The reaction is quenched with methanol/water and concentrated under reduced pressure. Purification is done by reverse phase HPLC to afford example 27 (0.107g; 28%), LC-MS (LC Method c): retention time 0.97 min, m/z 412 [M+H] + .

Compounds listed in Table IV under method J are made following this procedure with the following modifications to be noted: for example 26, 28, and 32 TBTU (O-(benzotriazol-1- yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate) is used as the coupling reagent instead of HATU as described in step 3.

Compounds listed in Table IV under method J are made following this procedure The following intermediates are made according to the above procedure. Table 6:

Method .Tl

Same as Method C step 3.

Compounds listed in Table IV under Method Jl are made following this procedure.

Method J2

Synthesis of Example 30

Intermediate XLIX (0.127 g; 0.209 mmol) is diluted with 3 mL methanol and HF pyridine complex (0.130 ml; 1.045 mmol; 70%) is added. The reaction is heated at 45°C in a sealed tube for 7 days adding aliquots of HF pyridine complex on a daily basis. Concentrated and purified by prepartive HPLC. Pooled product fractions and concentrated. Diluted in methanol and passed through a cartridge containing MP-Carbonate resin to neutralize molecule from trifluoroacetic acid contained in HPLC mobile phase to afford example 30 (0.024 g; 31.1%); LC-MS (LC method c): retention time 0.40 min, m/z 371[M+H + ] Compounds listed in Table IV under Method J2 are made following this procedure.

Method K:

Synthesis of Example 38

Step 1: Synthesis of compound 16

Compound 15 (6.4mL, 49.4mmol) is dissolved in 10OmL of acetonitrile under nitrogen and potassium iodide (739mg, 4.5mmol) is added followed by the addition of potassium carbonate (13.7g, 98.9mmol) and 4-aminotetrahydropyran (5g, 49.4mmol). The reaction mixture is heated in 90°C oil bath for 16 hours, After this time, the reaction mixture is cooled to room temperature and methyl 2-bromoisobutyrate (1.6mL, 12.4mmol) and potassium iodode (184.5mg, l.l lmmol) are added and the reaction mixture is heated in 90°C oil bath for 18 hours. After this time, the reaction mixture is cooled to room temperature and filtered through a glass funnel and the solid is washed with acetonitrile. The filtrate is concentrated under reduced pressure. Purification of the residue by flash chromatography on silica gel using ethyl acetate/heptane provides compound 16 (36%), LC-MS (LC method d): retention time: 0.27 min, m/z 202 [M+H + ]. The following intermediates aree made following the above procedure: Table 7:

Step 2: Synthesis of compound 17

Methyl iodide (2.5mL, 39.9mmol) is added dropwise to a mixture of compound 16 (4.38g, 17.7mmol) and potassium carbonate (10.94g, 79.1mmol) in 74mL of DMF. The reaction mixture is stirred at room temperature for 18 hours. After this time, methyl iodide (ImL, lβmmol) is added and the reaction mixture is stirred at room temperature for 65 hours. After this time, the reaction mixture is filtered and the solid is washed with ethyl acetate. The filtrate is concentrated under reduced pressure. Purification of the residue by flash chromatography on silica gel using ethyl acetate/heptane provides compound 17 (78%), LC-MS (Method d): retention time: 0.27 min, m/z 216 [M+H + ].

The following intermediates are made following the above procedure: Table 8:

Step 3: Synthesis of compound 18

Compound 17 (3.24g, 13.8mmol) is dissolved in 14mL of methanol in a pressure tube and sodium hydroxide aqueous solution (4N, 13.8mL, 55.3mmol) is added and the reaction mixture is heated in 80°C oil bath for 3 hours. After this time, the reaction mixture is concentrated under reduced pressure and the residue is neutralized to pH ~ 7 by adding IN HCl aqueous solution. The resulting solution is diluted with acetonitrile and dried in lyophilizer for 18 hours. After this time, the solid is suspended in acetone and filtered. The filtrate is concentrated under reduced pressure to afford compound 18 (100%), LC-MS (LC Method d): retention time: 0.29 min, m/z 202 [M+H + ].

The following intermediates are made following the above procedure: Table 9:

Step 4: Synthesis of Example 38

To a vial containing compound 18 (142mg, 0.707mmol) in ImL of DMF is added O- (benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (272mg, 0.848mmol) and diisopropylethylamine (0.27mL, 1.55mmol). The mixture is stirred at room temperature for 1 hour. To a vial contaning compound 25 (149mg, 0.707mmol) in 1.2mL of DMF is added sodium hydride in mineral oil (60%, 62mg, 1.55mmol). The mixture is stirred at room temperature for 15 minutes. To this solution is added drop wise the activated ester. The reaction mixture is stirred at room temperature for 1.5 hours. After this time, the reaction mixture is concentrated under reduce pressure to remove the solvent. The residue is quenched with saturated sodium bicarbonate aqueous solution and extracted with ethyl acetate twice. The organcis are combined and washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification of the residue by flash chromatography on silica gel using methanol/methylene chloride provides example 38 (17%), LC-MS (LC method d): retention time 1.03 min, m/z 394 [M+H + ].

Compounds listed in Table IV under Method K are made following this procedure. Also, the following interemediates are made following this procedure. Table 10:

Method Kl

Same as Method C step 3.

Compounds listed in Table IV under Method Kl are made following this procedure.

Method L

Synthesis of Example 36

Step 1: Synthesis of compound 19

To a pressure vessel containing compound 1 (1.25g, 7.49mmol) under nitrogen is added thionyl chloride (2.5mL, 34.5mmol) and catalytic amount of DMF, The pressure vessel is sealed and heated to 70°C where it is maintained for 2 hours. After this time, the reaction mixture is cooled to room temperature and diluted with dichloromethane and the acid chloride solution is concentrated in vacuo until ~ ImL left and this process is repeated twice. The crude acid chloride is used without further purification. The crude acid chloride is diluted with 2mL of tolune and added to the vial containing 3-[l-(2-Methoxy-ethoxy)-1-methyl-ethyl]-isoxazol- 5-ylamine (Ig, 5.0mmol) and N,N-diisopropylethylamine (4.35mL, 24.98mmol) in 1OmL of toluene and the reaction mixture is heated at 55°C for 18 hours. After this time, the reaction mixture is quenched with saturated sodium bicarbonate aqueous solution and extracted with ethyl acetate twice. The organics are combined and washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification of the residue by flash chromatography on silica gel using ethyl acetate/heptane provides compound 19 (52%), m/z 350 [M+H + ]

Compounds listed in intermediate table below are made following this procedure with the following modifications to be noted: for LXII reaction with thionyl chloride is heated at 80°C, for LXIV-LXV reaction with thionyl chloride is heated at 60 °C. Table 11:

Step 2: Synthesis of Example 36

Sodium hydride in mineral oil (60%, 61.8mg, 1.55mmol) is suspended in 2mL of THF and the solution of 4,4,4-Trifluoro-butylamine (81.9mg, 0.64mmol) in ImL of THF is added and the mixture is stirred at room temperature for 5 minutes under nitrogen before dropwise adding the solution of compound 19 (180mg, 0.52mmol) in 1.5mL of THF. The reaction mixture is stirred at room temperature for 18 hours. After this time, the reaction mixture is quenched with saturated sodium bicarbonate aqueous solution and extracted with ethyl acetate twice. The organics are combined and washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification of the residue by flash chromatography on silica gel using ethyl acetate/heptane provides example 36 (47%), LC-MS (LC Method d): retention time 1.05 min, m/z 396 [M+H + ].

Compounds listed in Table IV under Method L are made following this procedure, with the exception of example 14, 31, and 76 for which the last step is run according to Method B Step 2, and the crude is purified by prep HPLC or silica gel chromatography. Further for example 15 in Table IV NaH (4 equivalents) in mineral oil is used.

Also, the following intermediates are made following this procedure, with the exeption that for intermediates LXVII-LXXI NaH (4 equivalents) in mineral oil is used. Table 12:

Method Ll

Same as Method C step 3.

Compounds listed in Table IV under Method Ll are made following this procedure.

Method M

Synthesis of Example 22

Aluminium chloride (188mg, 1.4mmol) is added to a cooled (0°C) suspension of example 91 (33mg, 0.083mmol) in ethanethiol (ImL, 13.5mmol). The reaction mixture is stirred at room temperature for 65 hours. Aluminum chloride (lOOmg, 0.74mmol) is added and the reaction mixture is stirred at room temperature for 24 hours. After this time, the reaction mixture is quenched with 2OmL of water and 5 drops of concentrated hydrochloric acid aqueous solution and the mixture is stirred at room temperature for 1 hour. The mixture is extracted with ethyl acetate and the layers are separated. The aquous layer is basicified to pH -14 by adding 4N sodium hydroxide aqueous solution and the resulting mixture is extracted with ethyl acetate twice. The organics are combined and washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. Purification of the residue by flash chromatography on silica gel using methanol/methylene chloride provides example 22 (88%), m/z 384 [M+H + ].

Compounds listed in Table IV under Method M are made following this procedure.

Method N

Synthesis of Example 90

Step 1: Synthesis of Compound 20

To a flask containing compound 1 (11.8 g, 71 mmol) in MeCN (500 mL) is added EDCHCl (37.7 g, 212 mmol) and DMAP (8.6 mg, 71 mmol) at room temperature. 2-tert-butyl-2H- tetrazol-5-ylamine (10 g, 71 mmol) is added and the reaction mixture is stirred at room temperature for 24 h. Water (50 mL) is added and extracted with ethyl acetate (3 x 250 mL). The combined organic layers are dried (Na 2 SO 4 ) and the solvent is concentrated under reduced pressure. The crude material is purified by column chromatography (silica, eluent: heptanes, 0 - 50% ethyl acetate) to afford 9.4 g of compound 20. Yield: 45%, LC-MS (LC Method b): retention time: 1.71 min, m/z 290/292 [M+H].

Step 2: Synthesis of Example 90

N-(cis-4-Aminocyclohexyl)methanesulfonamide (0.22 g, 1.14 mmol) and compound 20 (0.32 g, 1.14 mmol) are charged into a pressure tube. The reaction mixture is heated to 150 °C for 20 min until the solids are melted. After cooling to room temperature, the residue is dissolved in ethyl acetate (10 mL) and washed with saturated NaHCO 3 solution (10 mL). The aqueous layer is extracted with ethyl acetate (2 x 20 mL) and the combined organic layers are dried over Na 2 SO 4 and filtered. The filtrate is concentrated under reduced pressure and the residue is purified by preparative LC (neutral method) followed by column chromatography (silica, eluent: heptanes, 0-60% ethyl acetate) to afford 6 mg of example 90. Yield: 1.3 %, LC-MS (LC Method a): retention time: 2.36 min, m/z 402 [M+H].

Synthesis of compound 25

Stepl: Synthesis of Compound 22 Paratoluenesulfonyl chloride (300.0 g, 1.58 mol) is added to a solution of compound 21 (100.0 g, 0.98 mol) in 800.0 mL of pyridine at 0°C. After stirring the reaction mixture at room temperature for 14 hours, it is pored into an ice solution of 6N HCl. Some precipitate is formed that is filtered and triturated with hexanes. After drying under vacuum the crude compound 22 is used without further purification in the next step. 1 H NMR (400 MHz, chloroform-d) δ ppm 1.70-1.89 (4H, m), 2.45 (3H, s), 3.44-3.50 (2H, m), 3.86 (2H, p), 4.70 (1H, m), 7.33 (2H, d, 8.4 Hz), 7.79 (2H, d, 8.0 Hz).

Step2: Stynthesis of compound 23

n-BuLi (1.6M solution in hexane, 122.5 mL) is added slowly to a stirred solution of DIPA in dry THF (150 mL) at -78 °C. After stirring the solution for 1 h at -78 °C, Isobutyric acid methyl ester (10. Og, 98.0 mmol) is added slowly and stirred for 3 h at -78 °C. Compound 22 (25.1g, 98.0 mmol) is dissolved in THF and added to the reaction mixture that is stirred for 1 h at -78 °C and 14 h at room temperature. The reaction mixture is quenched with water and extracted with ethyl acetate. The organic layer is washed with water and brine, dried over anhy. Na 2 SO 4 , filtered and concentrated under vacuum. The crude is purified by silica flash column chromatography (30% Ethyl actetate in heptane), to afford 7.0 g of the compound 23 as a pale yellow oil. Yield: 38%. 1H NMR (400 MHz, chloroform-d) δ ppm 1.13 (6H, s), 1.38-1.45 (4H, m), 1.78-1.85 (1H, m), 3.33-3.40 (2H, m), 3.66 (3H, s), 3.98 (2H, dd, 2.8 Hz).

Step 3: Synthesis of compound 24

A solution of compound 23 (6.Og, 32.2mmol) in CH 3 CN (2.2mL) is added to a stirred suspension of NaH (2.1g, 48.3mmol) in toluene (60.OmL) at 80 °C and the reaction mixture is further maintained at reflux temperature for 14 h. The reaction mixture is cooled to room temperature; water is added and extracted with EtOAc (3 times). The aqueous layer is acidified with 2N aq. HCl solution and extracted with EtOAc. The combined EtOAc layers are washed with brine, dried over anhy. Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the compound 24 (4.2g) that is used in the next step without further purification.

Step 4: Synthesis of compound 25

Hydroxylamine sulphate (2.95 g, 17.9 mmol) and solid NaOH (2.16g, 53.77mmol) are added to a stirring solution of compound 24 (3.5g, 17.9 mmol) in water (35.0 mL), and the resultant reaction mixture is stirred at reflux for 14 h. The reaction mixture is cooled to room temperature and then extracted with EtOAc (3 times). The combined EtOAc layers are washed with water, brine, dried over anhy. Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude is purified by silica gel column chromatography (50% ethyla cetate in heptane) to afford 1.4 g of compound 25. Yield: 37%, LC-MS (Method d): retention time: 1.09 min, m/z 211 [M+H].

Synthesis of compound 28

A solution of compound 26 and compound 27 in cone. HCl isheated at 100 °C for 3 h. The reaction mixture is quenched with aq NH 3 solution and extracted with ethyl acetate (twice). The combined EtOAc fraction is washed with brine solution, dried over anh. Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude is purified by silica gel column chromatography (5% methanol in dichloromethane) to afford 0.85 g of compound 28. Yield: 30%, LC-MS (Method d): retention time: 0.74 min, m/z 186 [M+H].

Method O: Synthesis of Example 58:

A microwave reaction vessel is charged with a solution of compound 29 (120 mg, 0.37 mmol, prepared according to WO2009105509, Boehringer Ingelheim) in THF (2 mL) and NaH (60% dispersion in mineral oil, 15mg, 0.37 mmol) is added. The mixture is stirred at room temperature for 5 min and methanesulfonylchoride (28 μL, 0.37 mmol) is added. The reaction mixture is heated in a microwave to 70 °C for 1 h. The mixture is diluted with ethyl acetate (20 mL) and washed with IN aqueous HCl solution (2 x 3 mL) and brine (3 mL). The organic layer is dried (Na 2 SO 4 ), filtered and concentrated under reduced pressure. The residue is dissolved in ethyl acetate and heptanes are added. The resulting white precipitate is isolated by filtration and dried under reduced pressure to afford 18 mg of example 58. Yield: 12%; LC- MS (Method a): retention time: 3.92 min, m/z 402 [M+H].

Compounds listed in Table IV under Method O are made following this procedure.

Table IV: Examples

Analytical Methods

LC Method a:

HPLC-MS equipment

HPLC pumps: Agilent G1312A Autoinjectors: CTC PAL HTC Detectors: MS: Waters ZQ UV: Waters 2996 photodiode array

Ancillary: Waters 2420 evaporative light scattering detectors (ELS) Hi her s ecification method desi ned for medicinal chemistr sam le screenin

LC Method b:

HPLC-MS equipment:

Shimadzu LCMS-2010EV system: (MS, pump, PDA) Autoinjectors CTC PAL HTS autosampler

Standard method for routine high throughput analysis

LC Method c: Column: Agilent SB-C18 1.8μm 3x50mm, 1.5mL/min flow rate

LC Method d: Column: Agilent SB-AQ 1.8μm 3x50mm, 1.5mL/min flow rate

LC Method e: Column: BEH C18, 1.7μm 2.1x50mm, 0.8mL/min flow rate

LC Method f: Column: Agilent Zorbax C18 SB 3.5μm 4.6x30mm, 2.5mL/min flow rate

LC Method g: Column: Agilent Zorbax Eclipse XDB-C8 5.0μm 4.6xl50mm, 1.5mL/min flow rate

LC Method h: Column: Zorbax XDB-C8 4.6x50 mm 3.5 μm, 2 niL/min flow rate

Assessment of Biological Properties The biological properties of the compounds of the formula I are assessed using the assays described below.

A. Human CBl and CB2 Receptor Binding: Experimental Method: CB2 membranes are purchased and made from HEK293 EBNA cells stably transfected with human CB2 receptor cDNA (Perkin Elmer Life and Analytical Sciences). CBl membranes are isolated from HEK cells stably co-transfected with human CBl receptor and Gαl6 cDNA's. The membrane preparation is bound to scintillation beads (Ysi-Poly-L-lysine SPA beads, GE Healthcare) for 4 hours at room temperature in assay buffer containing 5OmM Tris, pH 7.5, 2.5mM EDTA, 5mM MgCl 2 , 0.8% fatty acid free Bovine Serum Albumin. Unbound membrane is removed by washing in assay buffer. Membrane-bead mixture is added to 96- well assay plates in the amounts of 15ug membrane per well (CB2) or 2.5ug per well (CBl) and lmg SPA bead per well. Compounds are added to the membrane-bead mixture in dose- response concentrations ranging from Ix 10 -5 M to IxIO -10 M with 0.25% DMSO, final. The competition reaction is initiated with the addition of 3 H-CP55940 (Perkin Elmer Life and

Analytical Sciences) at a final concentration of 1.5nM (CB2) or 2.5nM (CBl). The reaction is incubated at room temperature for 18hours and read on TopCount NXT plate reader. Total and non-specific binding is determined in the absence and presence of 1.25uM Win 55212 (Sigma). IC50 values for each compound are calculated as the concentration of compound that inhibits the specific binding of the radioactively labeled ligand to the receptor by 50% using the XLFit 4.1 four parameter logistic model. IC50 values are converted to inhibition constant (Ki) values using Cheng-Prusoff equation.

B. CB 2R mediated modulation of c AMP synthesis: Compounds of the invention are evaluated for their CB2 agonist or inverse agonistic activity in accordance with the following experimental method. Compounds which are shown to bind to CB2 by the binding assay described above but which are not shown to exhibit CB2R- mediated modulation of cAMP synthesis by this assay are presumed to be CB2 antagonists.

Experimental Method:

CHO cells expressing human CB2R (Euroscreen) are plated at a density of 5000 cells per well in 384 well plates and incubated overnight at 37°C. After removing the media, the cells are treated with test compounds diluted in stimulation buffer containing ImM IBMX, 0.25% BSA and lOuM Forskolin. The assay is incubated for 30 minutes at 37°C. Cells are lysed and the cAMP concentration is measured using DiscoverX -XS cAMP kit, following the manufacturer's protocol. In this setting, agonists will decrease forskolin induced production of cAMP while inverse agonists will further increase forskolin induced production of cAMP. EC50 of agonists are calculated as follows. The maximal amount of cAMP produced by forskolin compared to the level of cAMP inhibited by IuM CP55940 is defined as 100%. The EC50 value of each test compound is determined as the concentration at which 50% of the forskolin- stimulated cAMP synthesis is inhibited. Data is analyzed using a four-parameter logistic model. (Model 205 of XLfit 4.0).

C. CBlR mediated modulation of cAMP synthesis:

Compounds of the invention are evaluated for their CB 1 agonist or inverse agonistic activity in accordance with the following experimental method. Compounds which are shown to bind to CBl by the binding assay described above but which are not shown to exhibit CBlR- mediated modulation of cAMP synthesis by this assay are presumed to be CBl antagonists.

Experimental Method:

CHO cells expressing human CBlR (Euroscreen) are plated at a density of 5000 cells per well in 384 well plates and incubated overnight at 37°C. After removing the media, the cells are treated with test compounds diluted in stimulation buffer containing ImM IBMX, 0.25% BSA and lOuM Forskolin. The assay is incubated for 30 minutes at 37°C. Cells are lysed and the cAMP concentration is measured using DiscoverX -XS cAMP kit, following the manufacturer's protocol. In this setting, agonists will decrease forskolin induced production of cAMP while inverse agonists will further increase forskolin induced production of cAMP. EC50 of agonists are calculated as follows. The maximal amount of cAMP produced by forskolin compared to the level of cAMP inhibited by IuM CP55940 is defined as 100%. The EC50 value of each test compound is determined as the concentration at which 50% of the forskolin- stimulated cAMP synthesis is inhibited. Data is analyzed using a four-parameter logistic model. (Model 205 of XLfit 4.0).

Compounds Having Agonist Activity

Through the use of the above described assays compounds are found to exhibit agonistic activity and thus to be particularly well suited for the treatment of pain as well as for the treatment of inflammation. Preferred compounds of the invention will have an activity range of CB2 (<500nM) and CBl (>20000).

Therapeutic Use

As can be demonstrated by the assays described above, the compounds of the invention are useful in modulating the CB2 receptor function. By virtue of this fact, these compounds have therapeutic use in treating disease- states and conditions mediated by the CB2 receptor function or that would benefit from modulation of the CB2 receptor function.

As the compounds of the invention modulate the CB2 receptor function, they have very useful anti-inflammatory and immune-suppressive activity and they can be used in patients as drugs, particularly in the form of pharmaceutical compositions as set forth below, for the treatment of disease-states and conditions. As noted before, those compounds which are CB2 agonists can also be employed for the treatment of pain.

The agonist, antagonist and inverse agonist compounds according to the invention can be used in patients as drugs for the treatment of the following disease- states or indications that are accompanied by inflammatory processes:

(i) Lung diseases: e.g. asthma, bronchitis, allergic rhinitis, emphysema, adult respiratory distress syndrome (ARDS), pigeon fancier's disease, farmer's lung, chronic obstructive pulmonary disease (COPD), asthma including allergic asthma (atopic or non-atopic) as well as exercise-induced bronchoconstriction, occupational asthma, viral- or bacterial exacerbation of asthma, other non-allergic asthmas and "wheezy- infant syndrome", pneumoconiosis, including aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis; (ii) Rheumatic diseases or autoimmune diseases or musculoskeletal diseases: all forms of rheumatic diseases, especially rheumatoid arthritis, acute rheumatic fever, and polymyalgia rheumatica; reactive arthritis; rheumatic soft tissue diseases; inflammatory soft tissue diseases of other genesis; arthritic symptoms in degenerative joint diseases (arthroses); tendinitis, bursitis, osteoarthritis, traumatic arthritis; collagenoses of any genesis, e.g., systemic lupus erythematosus, scleroderma, polymyositis, dermatomyositis, Sjogren syndrome, Still disease, Felty syndrome; and osteoporosis and other bone resorption diseases;

(iii) Allergic diseases: all forms of allergic reactions, e.g., angioneurotic edema, hay fever, insect bites, allergic reactions to drugs, blood derivatives, contrast agents, etc., anaphylactic shock (anaphylaxis), urticaria, angioneurotic edema, and contact dermatitis; (iv) Vascular diseases: panarteritis nodosa, polyarteritis nodosa, periarteritis nodosa, arteritis temporalis, Wegner granulomatosis, giant cell arthritis, atherosclerosis, reperfusion injury and erythema nodosum;

(v) Dermatological diseases: e.g. dermatitis, psoriasis; sunburn, burns, eczema;

(vi) Renal diseases: e.g. nephrotic syndrome; and all types of nephritis, e.g., glomerulonephritis; pancreatits;

(vii) Hepatic diseases: e.g. acute liver cell disintegration; acute hepatitis of various genesis, e.g., viral, toxic, drug-induced; and chronically aggressive and/or chronically intermittent hepatitis;

(viii) Gastrointestinal diseases: e.g. inflammatory bowel diseases, irritable bowel syndrome, regional enteritis (Crohns disease), colitis ulcerosa; gastritis; aphthous ulcer, celiac disease, regional ileitis, gastroesophageal reflux disease;

(ix) Neuroprotection: e.g. in the treatment of neurodegeneration following stroke; cardiac arrest; pulmonary bypass; traumatic brain injury; spinal cord injury or the like;

(x) Eye diseases: allergic keratitis, uveitis, or iritis; conjunctivitis; blepharitis; neuritis nervi optici; choroiditis; glaucoma and sympathetic ophthalmia;

(xi) Diseases of the ear, nose, and throat (ENT) area: e.g. tinnitus; allergic rhinitis or hay fever; otitis externa; caused by contact eczema, infection, etc.; and otitis media;

(xii) Neurological diseases: e.g. brain edema, particularly tumor-related brain edema; multiple sclerosis; acute encephalomyelitis; meningitis; acute spinal cord injury; trauma; dementia, particularly degenerative dementia (including senile dementia, Alzheimer's disease; Parkinson's disease and Creutzfeldt-Jacob disease;

Huntington's chorea, Pick's disease; motor neuron disease), vascular dementia

(including multi-infarct dementia) as well as dementia associated with intracranial space occupying lesions; infections and related conditions (including HIV infection); Guillain-Barre syndrome; myasthenia gravis, stroke; and various forms of seizures, e.g., nodding spasms; (xiϋ) Blood diseases: acquired hemolytic anemia; aplastic anemia, and idiopathic thrombocytopenia;

(xiv) Tumor diseases: acute lymphatic leukemia; Hodgkin's disease, malignant lymphoma; lymphogranulomatoses; lymphosarcoma; solid malignant tumors; extensive metastases,; (xv) Endocrine diseases: endocrine ophthalmopathy; endocrine orbitopathia; thyrotoxic crisis; Thyroiditis de Quervain; Hashimoto thyroiditis; Morbus Basedow; granulomatous thyroiditis; struma lymphomatosa; and Graves disease; type I diabetes

(insulin-dependent diabetes);

(xvi) Organ and tissue transplantations and graft-versus-host diseases; (xvii) Severe states of shock, e.g., septic shock, anaphylactic shock, and systemic inflammatory response syndrome (SIRS);

(xviii) Acute pain such as dental pain, perioperative, post-operative pain, traumatic pain, muscle pain, pain in burned skin, sun burn, trigeminal neuralgia, sun burn; spasm of the gastrointestinal tract or uterus, colics; (xix) Visceral pain such as pain associated with chronic pelvic pain, pancreatitis, peptic ulcer, interstitial cystitis, renal colic, angina, dysmenorrhoea, menstruation, gynaecological pain, irritable bowel syndrome (IBS), non-ulcer dyspepsia, non-cardiac chest pain, myocardial ischemia;

(xx) Neuropathic pain such as low back pain, non-herpetic neuralgia, post herpetic neuralgia, diabetic neuropathy, nerve injury, acquired immune deficiency syndrome

(AIDS) related neuropathic pain, head trauma, painful traumatic mononeuropathy, toxin and chemotherapy induced pain, phantom limb pain, painful polyneuropathy, thalamic pain syndrome, post-stroke pain, central nervous system injury, post surgical pain, stump pain, repetitive motion pain, pain induced by post mastectomy syndrome, multiple sclerosis, root avulsions, postthoracotomy syndrome, neuropathic pain associated hyperalgesia and allodynia. (xxi) Inflammatory/nociceptive pain induced by or associated with disorders such as osteoarthritis, rheumatoid arthritis, rheumatic disease, teno-synovitis, gout, vulvodynia, myofascial pain (muscular injury, fibromyalgia), tendonitis, osteoarthritis, juvenile arthritis, spondylitis, gouty arthritis, psoriatic arthritis, muscoskeletal pain, fibromyalgia, sprains and strains, sympathetically maintained pain, myositis, pain associated with migraine, toothache, influenza and other viral infections such as the common cold, rheumatic fever, systemic lupus erythematosus;

(xxii) Cancer pain induced by or associated with tumors such as lymphatic leukemia; Hodgkin's disease, malignant lymphoma; lymphogranulomatoses; lymphosarcoma; solid malignant tumors; extensive metastases; (xxiii) Headache such as cluster headache, migraine with and without aura, tension type headache, headache with different origins, headache disorders including prophylactic and acute use;

(xxiv) various other disease-states or conditions including, restenosis following percutaneous transluminal coronary angioplasty, acute and chronic pain, atherosclerosis, reperfusion injury, congestive heart failure, myocardial infarction, thermal injury, multiple organ injury secondary to trauma, necrotizing enterocolitis and syndromes associated with hemodialysis, leukopheresis, and granulocyte transfusion, sarcoidosis, gingivitis, pyrexia, edema resulting from trauma associated with bums, sprains or fracture, cerebral oedema and angioedema, Diabetes such as diabetic vasculopathy, diabetic neuropathy, diabetic retinopathy, post capillary resistance or diabetic symptoms associated with insulitis (e.g. hyperglycemia, diuresis, proteinuria and increased nitrite and kallikrein urinary excretion).

Other indications include: epilepsy, septic shock e.g. as antihypovolemic and/or antihypotensive agents, cancer, sepsis, osteoporosis, benign prostatic hyperplasia and hyperactive bladder, pruritis, vitiligo, general gastrointestinal disorders, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, tissue damage and postoperative fever, syndromes associated with Itching.

Besides being useful for human treatment, these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like.

For treatment of the above-described diseases and conditions, a therapeutically effective dose will generally be in the range from about 0.01 mg to about 100 mg/kg of body weight per dosage of a compound of the invention; preferably, from about 0.1 mg to about 20 mg/kg of body weight per dosage. For example, for administration to a 70 kg person, the dosage range would be from about 0.7 mg to about 7000 mg per dosage of a compound of the invention, preferably from about 7.0 mg to about 1400 mg per dosage. Some degree of routine dose optimization may be required to determine an optimal dosing level and pattern. The active ingredient may be administered from 1 to 6 times a day.

General Administration and Pharmaceutical Compositions

When used as pharmaceuticals, the compounds of the invention are typically administered in the form of a pharmaceutical composition. Such compositions can be prepared using procedures well known in the pharmaceutical art and comprise at least one compound of the invention. The compounds of the invention may also be administered alone or in combination with adjuvants that enhance stability of the compounds of the invention, facilitate administration of pharmaceutical compositions containing them in certain embodiments, provide increased dissolution or dispersion, increased inhibitory activity, provide adjunct therapy, and the like. The compounds according to the invention may be used on their own or in conjunction with other active substances according to the invention, optionally also in conjunction with other pharmacologically active substances. In general, the compounds of this invention are administered in a therapeutically or pharmaceutically effective amount, but may be administered in lower amounts for diagnostic or other purposes.

Administration of the compounds of the invention, in pure form or in an appropriate pharmaceutical composition, can be carried out using any of the accepted modes of administration of pharmaceutical compositions. Thus, administration can be, for example, orally, buccally (e.g., sublingually), nasally, parenterally, topically, transdermally, vaginally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as, for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages. The pharmaceutical compositions will generally include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, adjuvants, diluents, vehicles, or combinations thereof. Such pharmaceutically acceptable excipients, carriers, or additives as well as methods of making pharmaceutical compositions for various modes or administration are well-known to those of skill in the art. The state of the art is evidenced, e.g., by Remington: The Science and Practice of Pharmacy, 20th Edition, A. Gennaro (ed.), Lippincott Williams & Wilkins, 2000; Handbook of Pharmaceutical Additives, Michael & Irene Ash (eds.), Gower, 1995; Handbook of Pharmaceutical Excipients, A. H. Kibbe (ed.), American Pharmaceutical Ass'n, 2000; H. C. Ansel and N. G. Popovish, Pharmaceutical Dosage Forms and Drug Delivery Systems, 5th ed., Lea and Febiger, 1990; each of which is incorporated herein by reference in their entireties to better describe the state of the art.

As one of skill in the art would expect, the forms of the compounds of the invention utilized in a particular pharmaceutical formulation will be selected (e.g., salts) that possess suitable physical characteristics (e.g., water solubility) that is required for the formulation to be efficacious. Pharmaceutical compositions suitable for buccal (sub-lingual) administration include lozenges comprising a compound of the present invention in a flavored base, usually sucrose, and acacia or tragacanth, and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.

Pharmaceutical compositions suitable for parenteral administration comprise sterile aqueous preparations of a compound of the present invention. These preparations are preferably administered intravenously, although administration can also be effected by means of subcutaneous, intramuscular, or intradermal injection. Injectable pharmaceutical formulations are commonly based upon injectable sterile saline, phosphate-buffered saline, oleaginous suspensions, or other injectable carriers known in the art and are generally rendered sterile and isotonic with the blood. The injectable pharmaceutical formulations may therefore be provided as a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, including 1,3-butanediol, water, Ringer's solution, isotonic sodium chloride solution, fixed oils such as synthetic mono- or diglycerides, fatty acids such as oleic acid, and the like. Such injectable pharmaceutical formulations are formulated according to the known art using suitable dispersing or setting agents and suspending agents. Injectable compositions will generally contain from 0.1 to 5% w/w of a compound of the invention.

Solid dosage forms for oral administration of the compounds include capsules, tablets, pills, powders, and granules. For such oral administration, a pharmaceutically acceptable composition containing a compound(s) of the invention is formed by the incorporation of any of the normally employed excipients, such as, for example, pharmaceutical grades of mannitol, lactose, starch, pregelatinized starch, magnesium stearate, sodium saccharine, talcum, cellulose ether derivatives, glucose, gelatin, sucrose, citrate, propyl gallate, and the like. Such solid pharmaceutical formulations may include formulations, as are well-known in the art, to provide prolonged or sustained delivery of the drug to the gastrointestinal tract by any number of mechanisms, which include, but are not limited to, pH sensitive release from the dosage form based on the changing pH of the small intestine, slow erosion of a tablet or capsule, retention in the stomach based on the physical properties of the formulation, bioadhesion of the dosage form to the mucosal lining of the intestinal tract, or enzymatic release of the active drug from the dosage form.

Liquid dosage forms for oral administration of the compounds include emulsions, microemulsions, solutions, suspensions, syrups, and elixirs, optionally containing pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like. These compositions can also contain additional adjuvants such as wetting, emulsifying, suspending, sweetening, flavoring, and perfuming agents.

Topical dosage forms of the compounds include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants, eye ointments, eye or ear drops, impregnated dressings and aerosols, and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams. Topical application may be once or more than once per day depending upon the usual medical considerations. Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles. The formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation, more usually they will form up to about 80% of the formulation.

Transdermal administration is also possible. Pharmaceutical compositions suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. Such patches suitably contain a compound of the invention in an optionally buffered, aqueous solution, dissolved and/or dispersed in an adhesive, or dispersed in a polymer. A suitable concentration of the active compound is about 1% to 35%, preferably about 3% to 15%.

For administration by inhalation, the compounds of the invention are conveniently delivered in the form of an aerosol spray from a pump spray device not requiring a propellant gas or from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, heptafluoropropane, carbon dioxide, or other suitable gas. In any case, the aerosol spray dosage unit may be determined by providing a valve to deliver a metered amount so that the resulting metered dose inhaler (MDI) is used to administer the compounds of the invention in a reproducible and controlled way. Such inhaler, nebulizer, or atomizer devices are known in the prior art, for example, in PCT International Publication Nos. WO 97/12687 (particularly Figure 6 thereof, which is the basis for the commercial RESPIMAT® nebulizer); WO 94/07607; WO 97/12683; and WO 97/20590, to which reference is hereby made and each of which is incorporated herein by reference in their entireties.

Rectal administration can be effected utilizing unit dose suppositories in which the compound is admixed with low-melting water-soluble or insoluble solids such as fats, cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights, or fatty acid esters of polyethylene glycols, or the like. The active compound is usually a minor component, often from about 0.05 to 10% by weight, with the remainder being the base component.

In all of the above pharmaceutical compositions, the compounds of the invention are formulated with an acceptable carrier or excipient. The carriers or excipients used must, of course, be acceptable in the sense of being compatible with the other ingredients of the composition and must not be deleterious to the patient. The carrier or excipient can be a solid or a liquid, or both, and is preferably formulated with the compound of the invention as a unit- dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compound. Such carriers or excipients include inert fillers or diluents, binders, lubricants, disintegrating agents, solution retardants, resorption accelerators, absorption agents, and coloring agents. Suitable binders include starch, gelatin, natural sugars such as glucose or β-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. Lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.

Pharmaceutically acceptable carriers and excipients encompass all the foregoing additives and the like.