Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HETEROCYCLIC COMPOUNDS AS RSV INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2020/210246
Kind Code:
A1
Abstract:
The present invention discloses compounds of Formula (I), or pharmaceutically acceptable salts, esters, or prodrugs thereof: which inhibit Respiratory Syncytial Virus (RSV). The present invention further relates to pharmaceutical compositions comprising the aforementioned compounds for administration to a subject suffering from RSV infection. The invention also relates to methods of treating an RSV infection in a subject by administering a pharmaceutical composition comprising the compounds of the present invention.

Inventors:
OR YAT SUN (US)
YU JIANMING (US)
KIM IN JONG (US)
Application Number:
PCT/US2020/027090
Publication Date:
October 15, 2020
Filing Date:
April 07, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ENANTA PHARM INC (US)
International Classes:
A61K31/397; A61K31/55; C07D205/00; C07D233/16; C07D311/96
Foreign References:
US20190040084A12019-02-07
US20170355717A12017-12-14
US20120196846A12012-08-02
Attorney, Agent or Firm:
HARLAN, Edgar, W. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed: 1. A compound represented by Formula (I):

,

or a pharmaceutically acceptable salt thereof, wherein:

A is selected from the group consisting of:

1) optionally substituted aryl;

2) optionally substituted heteroaryl;

3) optionally substituted–C3-C12 cycloalkyl;

4) optionally substituted–C3-C12 cycloalkenyl; and

5) optionally substituted 3- to 12- membered heterocycloalkyl;

B is selected from the group consisting of:

1) optionally substituted aryl;

2) optionally substituted heteroaryl;

3) optionally substituted–C3-C12 cycloalkyl;

4) optionally substituted–C3-C12 cycloalkenyl; and

5) optionally substituted 3- to 12- membered heterocycloalkyl;

L is absent, -CONH-, -NHCO-, -NHCO2-, or -NHS(O)2-;

Each R1, R2, and R3 is independently selected from the group consisting of:

1) halogen;

2) cyano;

3) nitro;

4) optionally substituted–C1-C8 alkoxy;

5) optionally substituted–C1-C8 alkyl;

6) optionally substituted–C2-C8 alkenyl;

7) optionally substituted–C2-C8 alkynyl;

8) optionally substituted–C3-C6 cycloalkyl; 9) optionally substituted–C3-C6 cycloalkenyl; and

10) optionally substituted 3- to 6-membered heterocycloalkyl;

R4 is selected from the group consisting of:

1) hydrogen;

2) halogen;

3) cyano;

4) nitro;

5) optionally substituted–C1-C8 alkoxy;

6) optionally substituted–C1-C8 alkyl;

7) optionally substituted–C2-C8 alkenyl;

8) optionally substituted–C2-C8 alkynyl;

9) optionally substituted–C3-C8 cycloalkyl;

10) optionally substituted–C3-C8 cycloalkenyl; and

11) optionally substituted 3- to 8-membered heterocycloalkyl;

n is 0, 1, 2, 3 or 4; m is 0, 1, 2, 3 or 4; and v is 0, 1, 2, or 3. 2. The compound of claim 1, wherein L is absent or -CONH-. 3. The compound of claim 1, wherein A is selected from one of the following groups by removal of two hydrogen atoms, wherein each of the groups is optionally substituted when possible:

4. The compound of claim 1, wherein A is selected from the following groups, wherein each group is optionally substituted:

5. The compound of claim 1, wherein B is selected from the following groups by removal of a hydrogen atom, wherein each group is optionally substituted when possible:

.

6. The compound of claim 1, represented by one of Formulas (IIa), (IIb, and (IIc), or a pharmaceutically acceptable salt thereof:

wherein A, B, R1, R2, R3, R4, n, m, and v are as defined in claim 1. 7. The compound of claim 1, represented by one of Formulas (IVa), (IVb), and (IVc), or a pharmaceutically acceptable salt thereof:

wherein A, B, R1, R3, and R4 are as defined in claim 1. 8. The compound of claim 1, represented by one of Formulas (Va), (Vb), and (Vc), or a pharmaceutically acceptable salt thereof:

wherein A, B, R3, and R4 are as defined in claim 1. 9. The compound of claim 1, represented by one of Formulas (VIa), (VIb), and (VIc), or a pharmaceutically acceptable salt thereof:

wherein B, R1, R3, and R4 are as defined in claim 1.

10. The compound of claim 1, represented by one of Formulas (VIIa), (VIIb), and (VIIc), or a pharmaceutically acceptable salt thereof:

wherein B, R3, and R4 are as defined in claim 1. 11. The compound of claim 1, selected from the compounds set forth below or a pharmaceutically acceptable salt thereof:

12. A pharmaceutical composition comprising a compound according to any one of claims 1 to 11 and a pharmaceutically acceptable carrier, diluent or excipient.

13. A method of treating or preventing an RSV infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1 to 11. 14. The method of claim 13, further comprising the step of administering to the subject an additional anti-RSV agent.

15. The method of claim 13, further comprising administering to the subject a steroid anti-inflammatory compound.

16. A method of treating RSV and influenza in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1 to 11 and a therapeutically effective amount of an anti-influenza agent. 17. The method of claim 14, wherein the compound and the additional anti-RSV agent are co-formulated.

18. The method of claim 14, wherein the compound and the additional anti-RSV agent are co-administered.

19. The method of claim 14, wherein administering the additional anti-RSV agent is administered at a lower dose and/or frequency compared to the dose and/or frequency with which it is administered as a monotherapy.

Description:
HETEROCYCLIC COMPOUNDS AS RSV INHIBITORS

RELATED APPLICATION

This application claims the benefit of U.S. Provisional Application No. 62/831,470, filed on April 9, 2019. The entire teachings of the above application are incorporated herein by reference.

TECHNICAL FIELD

The present invention relates generally to compounds and pharmaceutical

compositions useful as Respiratory Syncytial Virus (RSV) inhibitors.

BACKGROUND OF THE INVENTION

Human respiratory syncytial virus (HRSV) is a negative-sense, single stranded, RNA paramyxovirus (KM. Empey, et al., Rev. Anti-Infective Agents , 2010, 50(1 May), 1258-1267). RSV is the leading cause of acute lower respiratory tract infections (ALRI) and affects patients of all ages. The symptoms in adults are usually not severe and are typically analogous to a mild cold. However, in infants and toddlers the virus can cause lower respiratory tract infections including bronchiolitis or pneumonia with many of them requiring hospitalization. Nearly all children have been infected by age 3. There are known high-risk groups that infection with RSV is more likely to progress into the ALRI. Premature infants and/or infants suffering from lung or cardiac disease are at the highest risk to develop ALRI. Additional high-risk groups include the elderly, adults with chronic heart and/or lung disease, stem cell transplant patients and the immunosuppressed.

Currently, there is no vaccine available to prevent HRSV infection. Palivizumab is a monoclonal antibody that is used prophylactically to prevent HRSV infection in high risk infants, e.g. premature infants, and infants with cardiac and/or lung disease. The high cost of palivizumab treatment limits its use for general purposes. Ribavirin has also been used to treat HRSV infections but its effectiveness is limited. There is a major medical need for new and effective HRSV treatments that can be used generally by all population types and ages.

There have been several RSV fusion inhibitors that have been disclosed in the following publications: W02010/103306, WO2012/068622, WO2013/096681,

WO2014/060411, WO2013/186995, WO2013/186334, WO 2013/186332, WO 2012 080451, WO 2012/080450, WO2012/080449, WO 2012/080447, WO 2012/080446, WO

2016/055792, WO 2016/097761, and J. Med. Chem. 2015, 58, 1630-1643. Examples of other N-protein inhibitors for treatment of HRSV have been disclosed in the following publications: WO 2004/026843, J. Med. Chem.2006, 49, 2311-2319, and J. Med. Chem. 2007, 50, 1685-1692. Examples of L-protein inhibitors for HRSV have been disclosed in the following publications: WO 2011/005842, WO 2005/042530, Antiviral Res.2005, 65, 125- 131, and Bioorg. Med. Chem. Lett.2013, 23, 6789-6793, Bioorg. Med. Chem. Lett.2017, 27, 2201-2206. Examples of nucleosides/polymerase inhibitors have been disclosed in the following publications: WO 2013/242525 and J. Med. Chem.2015, 58, 1862-1878.

There is a need for the development of effective treatments for HRSV. The present invention has identified these novel compounds and their inhibitory activity against HRSV. The invention includes methods to prepare the compounds as well as methods of using these compounds to treat disease. SUMMARY OF THE INVENTION

The present invention provides compounds represented by Formula (I), and pharmaceutically acceptable salts, esters and prodrugs thereof that can be used to treat or prevent viral (particularly HRSV) infection:

,

wherein:

A is selected from the group consisting of:

1) optionally substituted aryl;

2) optionally substituted heteroaryl;

3) optionally substituted–C 3 -C 12 cycloalkyl;

4) optionally substituted–C 3 -C 12 cycloalkenyl; and

5) optionally substituted 3- to 12- membered heterocycloalkyl;

B is selected from the group consisting of:

1) optionally substituted aryl;

2) optionally substituted heteroaryl; 3) optionally substituted–C 3 -C 12 cycloalkyl;

4) optionally substituted–C 3 -C 12 cycloalkenyl; and

5) optionally substituted 3- to 12- membered heterocycloalkyl;

L is absent, -CONH-, -NHCO-, -NHCO 2 -, or -NHS(O) 2 -; preferably L is -CONH-;

Each R 1 , R 2 , and R 3 is independently selected from the group consisting of:

1) halogen;

2) cyano;

3) nitro;

4) optionally substituted–C 1 -C 8 alkoxy;

5) optionally substituted–C 1 -C 8 alkyl;

6) optionally substituted–C 2 -C 8 alkenyl;

7) optionally substituted–C 2 -C 8 alkynyl;

8) optionally substituted–C 3 -C 6 cycloalkyl;

9) optionally substituted–C 3 -C 6 cycloalkenyl; and

10) optionally substituted 3- to 6-membered heterocycloalkyl;

R 4 is selected from the group consisting of:

1) hydrogen;

2) halogen;

3) cyano;

4) nitro;

5) optionally substituted–C 1 -C 8 alkoxy;

6) optionally substituted–C 1 -C 8 alkyl;

7) optionally substituted–C 2 -C 8 alkenyl;

8) optionally substituted–C 2 -C 8 alkynyl;

9) optionally substituted–C 3 -C 8 cycloalkyl;

10) optionally substituted–C 3 -C 8 cycloalkenyl; and

11) optionally substituted 3- to 8-membered heterocycloalkyl;

n is 0, 1, 2, 3 or 4;

m is 0, 1, 2, 3 or 4; and

v is 0, 1, 2, or 3.

Each preferred group stated above can be taken in combination with one, any or all other preferred groups. DETAILED DESCRIPTION OF THE INVENTION

In one embodiment.the present invention provides a compound represented by Formula (I) as described above, or a pharmaceutically acceptable salt, ester or prodrug thereof.

In certain embodiments of the compounds of Formula (I), n is 1 to 4 and each R 1 is independently halogen, optionally substituted–C 1 -C 3 alkoxy, or optionally substituted–C 1 - C 3 alkyl.

In certain embodiments of the compounds of Formula (I), n is 1 or 2 and each R 1 is independently F, optionally substituted–OCH 3 , or optionally substituted–CH 3 .

In certain embodiments of the compounds of Formula (I), n is 0.

In certain embodimentsof the compounds of Formula (I), m is 1 to 4 and each R 2 is independently halogen, optionally substituted–C 1 -C 3 alkoxy, or optionally substituted–C 1 - C 3 alkyl.

In certain embodiments of the compounds of Formula (I), m is 1 or 2 and each R 2 is independently F, optionally substituted–OCH 3 , or optionally substituted–CH 3 .

In certain embodiments of the compounds of Formula (I), m is 0.

In certain embodiments of the compounds of Formula (I), wherein n is 1, m is 0 and R 1 is F.

In certain embodiments of the compounds of Formula (I), v is 1 to 3, and each R 3 is independently halogen, optionally substituted–C 1 -C 3 alkoxy, optionally substituted–C 1 -C 3 alkyl, or optionally substituted–C 3 -C 6 cycloalkyl.

In certain embodiments of the compounds of Formula (I), v is 1 and R 3 is F, optionally substituted–OCH 3 , optionally substituted–CH 3 , or optionally substituted cyclopropyl.

In certain embodiments of the compounds of Formula (I), v is 0.

In certain embodiments of the compounds of Formula (I), n is 0 and m is 0.

In certain embodiments of the compounds of Formula (I), n is 0, m is 0, and v is 0. In certain embodiments of the compounds of Formula (I), R 4 is halogen, optionally substituted–C 1 -C 3 alkoxy, or optionally substituted–C 1 -C 3 alkyl.

In certain embodiments of the compounds of Formula (I), R 4 is H, F, optionally substituted–OCH 3 , or optionally substituted–CH 3 . In certain embodiments, R 4 is–CH 3 ,– CF 3 or–CHF 2 .

In certain embodiments of the compounds of Formula (I), L is absent, -CONH-, or - NHCO-. In certain embodiments, L is -CONH-. In certain embodiments of the compounds of Formula (I), A is optionally substituted aryl; preferably A is optionally substituted phenyl. The optional substituents are preferably independently selected from, but not limited to, halogen, -CN, -OH, -NH 2 , -NO 2 , -CH 3 , -CF 3 , -OCH 3 , -OCF 3 , -SO 2 CH 3 , -CH 2 N(CH 3 ) 2 , and -C(O)CH 3 . In preferred embodiments, there are 0 to 2 substituents and, more preferably, 0 or 1 substituent.

In certain embodiments of the compounds of Formula (I), A is optionally substituted heteroaryl; preferably A is optionally substituted thiophenyl. When present the substituent or substituents are preferably independently selected from halogen, -CN, -OH, -NH 2 , -NO 2 , - CH 3, - CF 3 , -OCH 3 , -OCF 3 , -SO 2 CH 3 , -CH 2 N(CH 3 ) 2 , and -C(O)CH 3 . In preferred

embodiments, there are 0 to 2 substituents and, more preferably, 0 or 1 substituent.

In certain embodiments of the compounds of Formula (I), B is optionally substituted aryl; preferably B is optionally substituted phenyl. The optional substituents are preferably independently selected from, but not limited to, halogen, -CN, -OH, -NH 2 , -NO 2 , -CH 3 , -CF 3 , -OCH 3 , -OCF 3 , -SO 2 CH 3 , -CH 2 N(CH 3 ) 2 , and -C(O)CH 3 . In preferred embodiments, there are 0 to 2 substituents and, more preferably, 0 or 1 substituent.

In certain embodiments of the compounds of Formula (I), B is optionally substituted heteroaryl. When present the substituent or substituents are preferably independently selected from halogen, -CN, -OH, -NH 2 , -NO 2 , -CH 3, - CF 3 , -OCH 3 , -OCF 3 , -SO 2 CH 3 , -CH 2 N(CH 3 ) 2 , and -C(O)CH 3 . In preferred embodiments, there are 0 to 2 substituents and, more preferably, 0 or 1 substituent.

In certain embodiments of the compounds of Formula (I), B is optionally substituted –C 3 -C 12 cycloalkyl. When present the substituent or substituents are preferably

independently selected from halogen, -CN, -OH, -NH 2 , -NO 2 , -CH 3, -CF 3 , -OCH 3 , -OCF 3 , - SO 2 CH 3 , -CH 2 N(CH 3 ) 2 , and -C(O)CH 3 . In preferred embodiments, there are 0 to 2 substituents and, more preferably, 0 or 1 substituent.

In another embodiment, the invention provides a compound represented by Formula (IIa) ~ (IIc) or a pharmaceutically acceptable salt, ester or prodrug thereof:

wherein A, B, R 1 , R 2 , R 3 , R 4 , n, m, and v are as previously defined.

In another embodiment, the invention provides a compound represented by one of Formulas (IIIa) ~ (IIIc) or a pharmaceutically acceptable salt, ester or prodrug thereof:

wherein A, B, R 1 , R 3 , R 4 , n, and v are as previously defined.

In another embodiment, the invention provides a compound represented by one of Formulas (IVa) ~ (IVc) or a pharmaceutically acceptable salt, ester or prodrug thereof:

wherein A, B, R 1 , R 3 , and R 4 are as previously defined.

In another embodiment, the invention provides a compound represented by one of Formulas (Va) ~ (Vc) or a pharmaceutically acceptable salt, ester or prodrug thereof:

wherein A, B, R 3 , and R 4 are as previously defined.

In certain embodiments of the compounds of Formulas (I), (IIa)-(IIc), (IIIa)-(IIIc), (IVa)-(IVc) and (Va)-(Vc), A is derived from one of the following by removal of two hydrogen atoms:

wherein each of the above is optionally substituted when possible. Preferably, when present, the substituents are independently selected from, but not limited to, -CN, -F, -Cl, -CH 3, - CF 3 , -OCH 3 , and -OCF 3 .

In certain embodiments of the compounds of Formulas (I), (IIa)-(IIc), (IIIa)-(IIIc), (IVa)-(IVc) and (Va)-(Vc), A is selected from the following:

wherein each of the above is optionally substituted. Preferably, when present, the substituents are independently selected from, but not limited to, -CN, -F, -Cl, -CH 3, - CF 3 , -OCH 3 , and - OCF 3 .

In another embodiment, the invention provides a compound represented by one of Formulas (VIa) ~ (VIc) or a pharmaceutically acceptable salt, ester or prodrug thereof:

wherein B, R 1 , R 3 , and R 4 are as previously defined.

In another embodiment, the invention provides a compound represented by one of Formulas (VIIa) ~ (VIIc) or a pharmaceutically acceptable salt, ester or prodrug thereof:

wherein B, R 3 , and R 4 are as previously defined. In certain embodiments of the compounds of Formulas (I), (IIa)-(IIc), (IIIa)-(IIIc), (IVa)-(IVc), (Va)-(Vc), (VIa)-(VIc) and (VIIa)-(VIIc), B is derived from one of the following by removal of a ring hydrogen atom:

wherein each of the above is optionally substituted when possible. Preferably, when present, the substituents are independently selected from, but not limited to, -CN, -F, -Cl, -CH 3, -CF 3 , - OCH 3 , and -OCF 3 .

It will be appreciated that the description of the present invention herein should be construed in congruity with the laws and principles of chemical bonding. In some instances it may be necessary to remove a hydrogen atom in order to accommodate a substituent at any given location.

It is intended that the definition of any substituent or variable (e.g., R 1 , R 2 , etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule.

It will be yet appreciated that the compounds of the present invention may contain one or more asymmetric carbon atoms and may exist in racemic, diastereoisomeric, and optically active forms. It will still be appreciated that certain compounds of the present invention may exist in different tautomeric forms. All tautomers are contemplated to be within the scope of the present invention.

In certain embodiments, the present invention provides a method for the prevention or treatment of RSV activities and for treating RSV infection in a subject in need thereof. The method comprises administering to the subject a therapeutically effective amount of a compound of Formula (I).

The present invention also provides the use of a compound of Formula (I) for the preparation of a medicament for the prevention or treatment of RSV.

Thus, in one embodiment, a compound of Formula (I), or pharmaceutically acceptable salt thereof, is combined with a steroid anti-inflammatory compound, for example budesonide or fluticasone. In a preferred embodiment, the steroid is administered in low doses to minimize immuno- suppressant effects. In another embodiment a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is combined with a non-steroid anti-inflammatory compound, for example leukotriene antagonists such as Singulair (Merck) or Accolate (Astra Zeneca), phosphodiesterase 4 inhibitors such as roflumilast (Altana), TNF alpha inhibitors such as Enbrel (Amgen), Remicade (Centocor), Humira (Abbott) or CDP870 (Celltech) or NSAFDS.

In a further embodiment, a compound of Formula (I) is combined with interleukin 8 or interleukin 9 inhibitors. The present invention thus also relates to a product containing a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and an anti

inflammatory compound for simultaneous, separate or sequential use in the treatment of RSV.

The present invention also relates to a combination of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, with an anti-influenza compound and the use of such a combination in the treatment of concomitant RSVand influenza infections. The present invention thus also relates to a product containing a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and an anti-influenza compound for simultaneous, separate or sequential use in the treatment of concomitant RSV and influenza infections. The compounds of the invention may be administered in a variety of dosage forms. Thus, they can be administered orally, for example as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules. The compounds of the invention may also be administered parenterally, whether subcutaneously, intravenously, intramuscularly, intrastemally,

transdermally or by infusion techniques. The compounds may also be administered as suppositories. In an embodiment, the compounds of the invention are administered by intranasal or intrabronchial administration. The present invention also provides an inhaler or nebuliser containing a medicament which comprises (a) a benzodiazepine derivative of the Formula (I), as defined above, or a pharmaceutically acceptable salt thereof, and (b) a pharmaceutically acceptable carrier or diluent.

The present invention also provides a pharmaceutical composition containing such a benzodiazepine derivative, or a pharmaceutically acceptable salt thereof, and a

pharmaceutically acceptable carrier or diluent.

The compounds of the invention are typically formulated for administration with a pharmaceutically acceptable carrier or diluent. For example, solid oral forms may contain, together with the active compound, diluents, e.g. lactose, dextrose, saccharose, cellulose, com starch or potato starch; lubricants, e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents; e.g. starches, arabic gums, gelatin, methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disaggregating agents, e.g. starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuffs; sweeteners; wetting agents, such as lecithin, polysorbates, laurylsulphates; and, in general, non toxic and pharmacologically inactive substances used in pharmaceutical formulations. Such pharmaceutical preparations may be manufactured in known manner, for example, by means of mixing, granulating, tableting, sugar coating, or film coating processes.

Liquid dispersions for oral administration may be syrups, emulsions and suspensions. The syrups may contain as carriers, for example, saccharose or saccharose with glycerine and/or mannitol and/or sorbitol.

Suspensions and emulsions may contain as carrier, for example a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, or polyvinyl alcohol. The suspension or solutions for intramuscular injections may contain, together with the active compound, a pharmaceutically acceptable carrier, e.g., sterile water, olive oil, ethyl oleate, glycols, e.g. propylene glycol, and if desired, a suitable amount of lidocaine hydrochloride.

Solutions for injection or infusion may contain as carrier, for example, sterile water or preferably they may be in the form of sterile, aqueous, isotonic saline solutions.

The present invention also relates to the novel compounds, as defined above; or a pharmaceutically acceptable salt thereof, for use in a method of treating the human or animal body. The present invention also relates to a pharmaceutical composition comprising a novel compound as defined above and a pharmaceutically acceptable diluant or carrier. Preferably, the pharmaceutical composition comprises a pharmaceutically acceptable salt of a novel compound as defined above. A pharmaceutically acceptable salt is as defined above. The novel compounds of the invention are typically administered in the manner defined above and the compounds are typically formulated for administration in the manner defined above.

Preferably, the pharmaceutical compositions comprise optically active isomers of the novel compounds of the invention. Thus, for example, preferred novel compounds of the invention containing only one chiral centre include an R enantiomer in substantially pure form, an S enantiomer in substantially pure form and enantiomeric mixtures which contain an excess of the R enantiomer or an excess of the S enantiomer. It is particularly preferred that pharmaceutical contains a compound of the invention which is a substantially pure optical isomer. For the avoidance of doubt, the novel compounds of the invention can, if desired, be used in the form of solvates.

Yet a further aspect of the present invention is a process of making any of the compounds delineated herein employing any of the synthetic means delineated herein.

Definitions

Listed below are definitions of various terms used to describe this invention. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.

The term "aryl," as used herein, refers to a mono-, bi-, or polycyclic carbocyclic ring system comprising at least one aromatic ring, including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, and indenyl. A polycyclic aryl is a polycyclic ring system that comprises at least one aromatic ring. Polycyclic aryls can comprise fused rings, covalently attached rings or a combination thereof.

The term "heteroaryl," as used herein, refers to a mono-, bi-, or polycyclic aromatic radical having one or more ring atom selected from S, O and N; and the remaining ring atoms are carbon, wherein any N or S contained within the ring may be optionally oxidized.

Heteroaryl includes, but is not limited to, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isoxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, quinoxalinyl. A polycyclic heteroaryl can comprise fused rings, covalently attached rings or a combination thereof.

In accordance with the invention, aromatic groups can be substituted or unsubstituted.

The term“bicyclic aryl” or“bicyclic heteroaryl” refers to a ring system consisting of two rings wherein at least one ring is aromatic; and the two rings can be fused or covalently attached. The term“alkyl” as used herein, refers to saturated, straight- or branched-chain hydrocarbon radicals. "C 1 -C 3 alkyl,””C 1 -C 6 alkyl,”“C 1 -C 10 alkyl”C 2 -C 4 alkyl,” or”C 3 -C 6 alkyl,” refer to alkyl groups containing from one to three, one to six, one to ten carbon atoms, 2 to 4 and 3 to 6 carbon atoms respectively. Examples of C 1 -C 8 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl, heptyl and octyl radicals.

The term“alkenyl” as used herein, refers to straight- or branched-chain hydrocarbon radicals having at least one carbon-carbon double bond by the removal of a single hydrogen atom.“C 2 -C 10 alkenyl,”“C 2 -C 8 alkenyl,”“C 2 -C 4 alkenyl,” or“C 3 -C 6 alkenyl,” refer to alkenyl groups containing from two to ten, two to eight, two to four or three to six carbon atoms respectively. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, heptenyl, octenyl, and the like.

The term“alkynyl” as used herein, refers to straight- or branched-chain hydrocarbon radicals having at least one carbon-carbon triple bond by the removal of a single hydrogen atom.“C 2 -C 10 alkynyl,”“C 2 -C 8 alkynyl,”“C 2 -C 4 alkynyl,” or“C 3 -C 6 alkynyl,” refer to alkynyl groups containing from two to ten, two to eight, two to four or three to six carbon atoms respectively. Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl, and the like.

The term“cycloalkyl”, as used herein, refers to a monocyclic or polycyclic saturated carbocyclic ring or a bi- or tri-cyclic group fused, bridged or spiro system, and the carbon atoms may be optionally oxo-substituted or optionally substituted with exocyclic olefinic, iminic or oximic double bond. Preferred cycloalkyl groups include C 3 -C 12 cycloalkyl, C 3 -C 6 cycloalkyl, C 3 -C 8 cycloalkyl and C 4 -C 7 cycloalkyl. Examples of C 3 -C 12 cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl, cyclooctyl, 4-methylene-cyclohexyl, bicyclo[2.2.1]heptyl, bicyclo[3.1.0]hexyl, spiro[2.5]octyl, 3- methylenebicyclo[3.2.1]octyl, spiro[4.4]nonanyl, and the like.

The term“cycloalkenyl”, as used herein, refers to monocyclic or polycyclic carbocyclic ring or a bi- or tri-cyclic group fused, bridged or spiro system having at least one carbon-carbon double bond and the carbon atoms may be optionally oxo-substituted or optionally substituted with exocyclic olefinic, iminic or oximic double bond. Preferred cycloalkenyl groups include C 3 -C 12 cycloalkenyl, C 3 -C 8 cycloalkenyl or C 5 -C 7 cycloalkenyl groups. Examples of C 3 -C 12 cycloalkenyl include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, bicyclo[2.2.1]hept- 2-enyl, bicyclo[3.1.0]hex-2-enyl, spiro[2.5]oct-4-enyl, spiro[4.4]non-1-enyl, bicyclo[4.2.1]non-3-en-9-yl, and the like.

As used herein, the term“arylalkyl” means a functional group wherein an alkylene chain is attached to an aryl group, e.g., -CH 2 CH 2 -phenyl. The term“substituted arylalkyl” means an arylalkyl functional group in which the aryl group is substituted. Similarly, the term“heteroarylalkyl” means a functional group wherein an alkylene chain is attached to a heteroaryl group. The term“substituted heteroarylalkyl” means a heteroarylalkyl functional group in which the heteroaryl group is substituted.

As used herein, the term“alkoxy” employed alone or in combination with other terms means, unless otherwise stated, an alkyl group having the designated number of carbon atoms connected to the rest of the molecule via an oxygen atom, such as, for example, methoxy, ethoxy, 1-propoxy, 2-propoxy (isopropoxy) and the higher homologs and isomers. Preferred alkoxy are (C 1 -C 3 ) alkoxy.

It is understood that any alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclic and cycloalkenyl moiety described herein can also be an aliphatic group or an alicyclic group.

An“aliphatic” group is a non-aromatic moiety comprised of any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contains one or more units of unsaturation, e.g., double and/or triple bonds.

Examples of aliphatic groups are functional groups, such as alkyl, alkenyl, alkynyl, O, OH, NH, NH 2 , C(O), S(O) 2 , C(O)O, C(O)NH, OC(O)O, OC(O)NH, OC(O)NH 2 , S(O) 2 NH, S(O) 2 NH 2 , NHC(O)NH 2 , NHC(O)C(O)NH, NHS(O) 2 NH, NHS(O) 2 NH 2 , C(O)NHS(O) 2, C(O)NHS(O) 2 NH or C(O)NHS(O) 2 NH 2 , and the like, groups comprising one or more functional groups, non-aromatic hydrocarbons (optionally substituted), and groups wherein one or more carbons of a non-aromatic hydrocarbon (optionally substituted) is replaced by a functional group. Carbon atoms of an aliphatic group can be optionally oxo-substituted. An aliphatic group may be straight chained, branched, cyclic, or a combination thereof and preferably contains between about 1 and about 24 carbon atoms, more typically between about 1 and about 12 carbon atoms. In addition to aliphatic hydrocarbon groups, as used herein, aliphatic groups expressly include, for example, alkoxyalkyls, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Aliphatic groups may be optionally substituted.

The terms“heterocyclic” or“heterocycloalkyl” can be used interchangeably and referred to a non-aromatic ring or a bi- or tri-cyclic group fused, bridged or spiro system, where (i) each ring system contains at least one heteroatom independently selected from oxygen, sulfur and nitrogen, (ii) each ring system can be saturated or unsaturated (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (v) any of the above rings may be fused to an aromatic ring, and (vi) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted or optionally substituted with exocyclic olefinic, iminic or oximic double bond.

Representative heterocycloalkyl groups include, but are not limited to, 1,3-dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl,

piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, 2-azabicyclo[2.2.1]-heptyl, 8-azabicyclo[3.2.1]octyl, 5- azaspiro[2.5]octyl, 1-oxa-7-azaspiro[4.4]nonanyl, 7-oxooxepan-4-yl, and tetrahydrofuryl. Such heterocyclic groups may be further substituted. Heteroaryl or heterocyclic groups can be C-attached or N-attached (where possible).

It is understood that any alkyl, alkenyl, alkynyl, alicyclic, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclic, aliphatic moiety or the like, described herein can also be a divalent or multivalent group when used as a linkage to connect two or more groups or substituents, which can be at the same or different atom(s). One of skill in the art can readily determine the valence of any such group from the context in which it occurs.

The term“substituted” refers to substitution by independent replacement of one, two, or three or more of the hydrogen atoms with substituents including, but not limited to, -F, -Cl, -Br, -I, -OH, C 1 -C 12 -alkyl; C 2 -C 12 -alkenyl, C 2 -C 12 -alkynyl, -C 3 -C 12 -cycloalkyl, protected hydroxy, -NO 2 , -N 3 , -CN, -NH 2 , protected amino, oxo, thioxo, -NH-C 1 -C 12 -alkyl, -NH-C 2 -C 8 - alkenyl, -NH-C 2 -C 8 -alkynyl, -NH-C 3 -C 12 -cycloalkyl, -NH-aryl, -NH-heteroaryl, -NH- heterocycloalkyl, -dialkylamino, -diarylamino, -diheteroarylamino, -O-C 1 -C 12 -alkyl, -O-C 2 - C 8 -alkenyl, -O-C 2 -C 8 -alkynyl, -O-C 3 -C 12 -cycloalkyl, -O-aryl, -O-heteroaryl, -O- heterocycloalkyl, -C(O)-C 1 -C 12 -alkyl, -C(O)-C 2 -C 8 -alkenyl, -C(O)-C 2 -C 8 -alkynyl, -C(O)-C 3 - C 12 -cycloalkyl, -C(O)-aryl, -C(O)-heteroaryl, -C(O)-heterocycloalkyl, -CONH 2 , -CONH-C 1 - C 12 -alkyl, -CONH-C 2 -C 8 -alkenyl, -CONH-C 2 -C 8 -alkynyl, -CONH-C 3 -C 12 -cycloalkyl, - CONH-aryl, -CONH-heteroaryl, -CONH-heterocycloalkyl, -OCO 2 -C 1 -C 12 -alkyl, -OCO 2 -C 2 - C 8 -alkenyl, -OCO 2 -C 2 -C 8 -alkynyl, -OCO 2 -C 3 -C 12 -cycloalkyl, -OCO 2 -aryl, -OCO 2 -heteroaryl, -OCO 2 -heterocycloalkyl, -CO 2 -C 1 -C 12 alkyl, -CO 2 -C 2 -C 8 alkenyl, -CO 2 -C 2 -C 8 alkynyl, CO 2 - C 3 -C 12 -cycloalkyl, -CO 2 - aryl, CO 2 -heteroaryl, CO 2 -heterocyloalkyl, -OCONH 2 , -OCONH- C 1 -C 12 -alkyl, -OCONH-C 2 -C 8 -alkenyl, -OCONH-C 2 -C 8 -alkynyl, -OCONH-C 3 -C 12 - cycloalkyl, -OCONH-aryl, -OCONH-heteroaryl, -OCONH- heterocyclo-alkyl, -NHC(O)H, - NHC(O)-C 1 -C 12 -alkyl, -NHC(O)-C 2 -C 8 -alkenyl, -NHC(O)-C 2 -C 8 -alkynyl, -NHC(O)-C 3 -C 12 - cycloalkyl, -NHC(O)-aryl, -NHC(O)-heteroaryl, -NHC(O)-heterocyclo-alkyl, -NHCO 2 -C 1 - C 12 -alkyl, -NHCO 2 -C 2 -C 8 -alkenyl, -NHCO 2 - C 2 -C 8 -alkynyl, -NHCO 2 -C 3 -C 12 -cycloalkyl, - NHCO 2 -aryl, -NHCO 2 -heteroaryl, -NHCO 2 - heterocycloalkyl, -NHC(O)NH 2 , -NHC(O)NH- C 1 -C 12 -alkyl, -NHC(O)NH-C 2 -C 8 -alkenyl, -NHC(O)NH-C 2 -C 8 -alkynyl, -NHC(O)NH-C 3 -C 12 - cycloalkyl, -NHC(O)NH-aryl, -NHC(O)NH-heteroaryl, -NHC(O)NH-heterocycloalkyl, NHC(S)NH 2 , -NHC(S)NH-C 1 -C 12 -alkyl, -NHC(S)NH-C 2 -C 8 -alkenyl, -NHC(S)NH-C 2 -C 8 - alkynyl, -NHC(S)NH-C 3 -C 12 -cycloalkyl, -NHC(S)NH-aryl, -NHC(S)NH-heteroaryl, - NHC(S)NH-heterocycloalkyl, -NHC(NH)NH 2 , -NHC(NH)NH-C 1 -C 12 -alkyl, -NHC(NH)NH- C 2 -C 8 -alkenyl, -NHC(NH)NH-C 2 -C 8 -alkynyl, -NHC(NH)NH-C 3 -C 12 -cycloalkyl, - NHC(NH)NH-aryl, -NHC(NH)NH-heteroaryl, -NHC(NH)NH-heterocycloalkyl, -NHC(NH)- C 1 -C 12 -alkyl, -NHC(NH)-C 2 -C 8 -alkenyl, -NHC(NH)-C 2 -C 8 -alkynyl, -NHC(NH)-C 3 -C 12 - cycloalkyl, -NHC(NH)-aryl, -NHC(NH)-heteroaryl, -NHC(NH)-heterocycloalkyl, - C(NH)NH-C 1 -C 12 -alkyl, -C(NH)NH-C 2 -C 8 -alkenyl, -C(NH)NH-C 2 -C 8 -alkynyl, -C(NH)NH- C 3 -C 12 -cycloalkyl, -C(NH)NH-aryl, -C(NH)NH-heteroaryl, -C(NH)NH-heterocycloalkyl, - S(O)-C 1 -C 12 -alkyl, -S(O)-C 2 -C 8 -alkenyl, - S(O)-C 2 -C 8 -alkynyl, -S(O)-C 3 -C 12 -cycloalkyl, - S(O)-aryl, -S(O)-heteroaryl, -S(O)-heterocycloalkyl, -SO 2 NH 2 , -SO 2 NH-C 1 -C 12 -alkyl, - SO 2 NH-C 2 -C 8 -alkenyl, -SO 2 NH- C 2 -C 8 -alkynyl, -SO 2 NH-C 3 -C 12 -cycloalkyl, -SO 2 NH-aryl, - SO 2 NH-heteroaryl, -SO 2 NH- heterocycloalkyl, -NHSO 2 -C 1 -C 12 -alkyl, -NHSO 2 -C 2 -C 8 - alkenyl, - NHSO 2 -C 2 -C 8 -alkynyl, -NHSO 2 -C 3 -C 12 -cycloalkyl, -NHSO 2 -aryl, -NHSO 2 - heteroaryl, -NHSO 2 -heterocycloalkyl, -CH 2 NH 2 , -CH 2 SO 2 CH 3 , -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, -heterocycloalkyl, -C 3 -C 12 -cycloalkyl, polyalkoxyalkyl, polyalkoxy, - methoxymethoxy, -methoxyethoxy, -SH, -S-C 1 -C 12 -alkyl, -S-C 2 -C 8 -alkenyl, -S-C 2 -C 8 - alkynyl, -S-C 3 -C 12 -cycloalkyl, -S-aryl, -S-heteroaryl, -S-heterocycloalkyl, or methylthio- methyl. In certain embodiments, the substituents are independently selected from halo, preferably Cl and F; C 1- C 4 -alkyl, preferably methyl and ethyl; halo-C 1- C 4 -alkyl, such as fluoromethyl, difluoromethyl, and trifluoromethyl; C 2 -C 4 -alkenyl; halo-C 2 -C 4 -alkenyl; C 3 -C 6 - cycloalkyl, such as cyclopropyl; C 1- C 4 -alkoxy, such as methoxy and ethoxy; halo-C 1- C 4 - alkoxy, such as fluoromethoxy, difluoromethoxy, and trifluoromethoxy, -CN; -OH; NH 2 ; C 1- C 4 -alkylamino; di(C 1- C 4 -alkyl)amino; and NO 2 . It is understood that the aryls, heteroaryls, alkyls, and the like can be further substituted. In some cases, each substituent in a substituted moiety is additionally optionally substituted when possible with one or more groups, each group being independently selected from C 1- C 4 -alkyl; -CF 3 , -OCH 3 , -OCF 3 , -F, -Cl, -Br, -I, - OH, -NO 2 , -CN, and -NH 2 . Preferably, a substituted alkyl group, such as a substituted methyl group, is substituted with one or more halogen atoms, more preferably one or more fluorine or chlorine atoms.

In certain embodiments, a substituted alkyl, alkenyl or alkoxy group is substituted with one or more halogen atoms, preferably fluorine atoms. Such substituted alkyl groups include fluoromethyl, difluoromethyl and trifluoromethyl. Such substituted alkoxy groups include fluoromethoxy, difluoromethoxy and trifluorom ethoxy.

The term“halo” or halogen” alone or as part of another substituent, as used herein, refers to a fluorine, chlorine, bromine, or iodine atom.

The term“optionally substituted”, as used herein, means that the referenced group may be substituted or unsubstituted. In one embodiment, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In another embodiment, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein.

The term“hydrogen” includes hydrogen and deuterium. In addition, the recitation of an atom includes other isotopes of that atom so long as the resulting compound is pharmaceutically acceptable.

In certain embodiments, the compounds of each formula herein are defined to include isotopically labelled compounds. An“isotopically labelled compound” is a compound in which at least one atomic position is enriched in a specific isotope of the designated element to a level which is significantly greater than the natural abundance of that isotope. For example, one or more hydrogen atom positions in a compound can be enriched with deuterium to a level which is significantly greater than the natural abundance of deuterium, for example, enrichment to a level of at least 1%, preferably at least 20% or at least 50%. Such a deuterated compound may, for example, be metabolized more slowly than its non- deuterated analog, and therefore exhibit a longer half-life when administered to a subject. Such compounds can synthesize using methods known in the art, for example by employing deuterated starting materials. Unless stated to the contrary, isotopically labelled compounds are pharmaceutically acceptable.

The term“hydroxy activating group,” as used herein, refers to a labile chemical moiety which is known in the art to activate a hydroxyl group so that it will depart during synthetic procedures such as in a substitution or an elimination reaction. Examples of hydroxyl activating group include, but are not limited to, mesylate, tosylate, trifl ate, p- nitrobenzoate, phosphonate and the like. The term“activated hydroxyl,” as used herein, refers to a hydroxy group activated with a hydroxyl activating group, as defined above, including mesylate, tosylate, triflate, p- nitrobenzoate, phosphonate groups, for example.

The term“hydroxy protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect a hydroxyl group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the hydroxy protecting group as described herein may be selectively removed. Hydroxy protecting groups as known in the art are described generally in T.H. Greene and P.G. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of hydroxyl protecting groups include benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, tert-butoxy- carbonyl, isopropoxycarbonyl, diphenylmethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, allyloxycarbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t-butyl, 2,2,2-trichloroethyl, 2 -trimethyl silyl ethyl, allyl, benzyl, triphenyl- methyl (trityl), m ethoxy methyl, methylthiomethyl, benzyloxymethyl, 2-(trimethylsilyl)- ethoxymethyl, methanesulfonyl, trimethylsilyl, triisopropyl silyl, and the like.

The term "protected hydroxy," as used herein, refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including benzoyl, acetyl, trimethylsilyl, triethyl silyl, methoxymethyl groups, for example.

The term“hydroxy prodrug group,” as used herein, refers to a promoiety group which is known in the art to change the physicochemical, and hence the biological properties of a parent drug in a transient manner by covering or masking the hydroxy group. After said synthetic procedure(s), the hydroxy prodrug group as described herein must be capable of reverting back to hydroxy group in vivo. Hydroxy prodrug groups as known in the art are described generally in Kenneth B. Sloan, Prodrugs. Topical and Ocular Drug Delivery

(Drugs and the Pharmaceutical Sciences; Volume 53), Marcel Dekker, Inc., New York (1992) and in“Prodrugs of Alcohols and Phenols” by S. S. Dhareshwar and V. J. Stella, in Prodrugs Challenges and Rewards Part-2 (Biotechnology: Pharmaceutical Aspects), edited by V. J. Stella, et al, Springer and AAPSPress, 2007, pp 31-99.

The term“amino protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect an amino group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the amino protecting group as described herein may be selectively removed. Amino protecting groups as known in the art are described generally in T.H. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis 3rd edition, John Wiley & Sons, New York (1999). Examples of amino protecting groups include, but are not limited to, methoxycarbonyl, t-butoxycarbonyl, 9-fluorenyl- methoxycarbonyl, benzyloxycarbonyl, and the like.

The term“protected amino,” as used herein, refers to an amino group protected with an amino protecting group as defined above.

The term "leaving group" means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction. By way of example, representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; and acyloxy groups, such as acetoxy, trifluoroacetoxy and the like.

The term "aprotic solvent," as used herein, refers to a solvent that is relatively inert to proton activity, i.e., not acting as a proton-donor. Examples include, but are not limited to, hydrocarbons, such as hexane and toluene, for example, halogenated hydrocarbons, such as, for example, methylene chloride, ethylene chloride, chloroform, and the like, heterocyclic compounds, such as, for example, tetrahydrofuran and N-methylpyrrolidinone, and ethers such as diethyl ether, bis-methoxymethyl ether. Such compounds are well known to those skilled in the art, and it will be obvious to those skilled in the art that individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of aprotic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification, 4th ed., edited by John A. Riddick et al .,

Vol. II, in the Techniques of Chemistry Series. John Wiley & Sons, NY, 1986.

The term“protic solvent,” as used herein, refers to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like. Such solvents are well known to those skilled in the art, and it will be obvious to those skilled in the art that individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of protogenic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification. 4th ed., edited by John A. Riddick et al., Vol. II, in the Techniques of Chemistry Series. John Wiley & Sons, NY, 1986.

Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term“stable,” as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).

The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. As can be appreciated by the skilled artisan, further methods of synthesizing the compounds of the Formula herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations. 2 nd Ed. Wiley-VCH (1999); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley and Sons (1999); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic

Synthesis John Wiley and Sons (1995), and subsequent editions thereof.

The term“subject,” as used herein, refers to an animal. Preferably, the animal is a mammal. More preferably, the mammal is a human. A subject also refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, fish, birds and the like.

The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and may include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.

The compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-, or as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optical isomers may be prepared from their respective optically active precursors by the procedures described above, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et al ., Enantiomers, Racemates, and Resolutions (John Wiley & Sons, 1981). When the compounds described herein contain olefmic double bonds, other unsaturation, or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers or cis- and trans- isomers. Likewise, all tautomeric forms are also intended to be included. Tautomers may be in cyclic or acyclic. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon-carbon double bond or carbon-heteroatom double bond depicted arbitrarily herein as trans may be cis, trans , or a mixture of the two in any proportion.

Certain compounds of the present invention may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present invention includes each conformational isomer of these compounds and mixtures thereof.

As used herein, the term "pharmaceutically acceptable salt," refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, etal. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977).

The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid. Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentane-propionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemi sulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further

pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate.

Pharmaceutically acceptable salts can also be prepared by deprotonation of the parent compound with a suitable base, thereby forming the anionic conjugate base of the parent compound. In such salts the counter ion is a cation. Suitable cations include ammonium and metal cations, such as alkali metal cations, including Li + , Na + , K + and Cs + , and alkaline earth metal cations, such as Mg 2+ and Ca 2+ .

As used herein, the term "pharmaceutically acceptable ester" refers to esters which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, esters of C 1 -C 6 -alkanoic acids, such as acetate, propionate, butyrate and pivalate esters.

In certain embodiments, the invention provides pharmaceutically acceptable prodrugs of the compounds disclosed herein. The term“pharmaceutically acceptable prodrugs” as used herein refers to those prodrugs of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the present invention. "Prodrug", as used herein means a compound, which is convertible in vivo by metabolic means (e.g. by hydrolysis) to afford any compound delineated by the formulae of the instant invention. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, Vol.4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed.). "Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 8:1-38(1992);

Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer,“Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzymology,” John Wiley and Sons, Ltd. (2002).

Additional types of prodrugs are also encompassed. For instance, free carboxyl groups can be derivatized as amides or alkyl esters. Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, ethyl succinate, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxycarbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 115. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers wherein the acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities. In certain embodiments, a compound of the invention can incorporate two or more groups that are metabolically removed in vivo to yield the active parent compound. For example, a compound of Formula I wherein R 1 is an amino acid residue can also be esterified, for example at a hydroxyl group of the sugar residue, to form a compound with two groups that can be removed in vivo to yield the active compound.

The term "treating", as used herein, means relieving, lessening, reducing, eliminating, modulating, or ameliorating, i.e. causing regression of the disease state or condition. Treating can also include inhibiting, i.e. arresting the development, of a existing disease state or condition, and relieving or ameliorating, i.e. causing regression of an existing disease state or condition, for example when the disease state or condition may already be present.

The term "preventing", as used herein means, to completely or almost completely stop a disease state or condition, from occurring in a patient or subject, especially when the patient or subject is predisposed to such or at risk of contracting a disease state or condition.

Additionally, the compounds of the present invention, for example, the salts of the compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules. Nonlimiting examples of hydrates include monohydrates, dihydrates, etc. Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc. "Solvates" means solvent addition forms that contain either stoichiometric or non stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H 2 O, such combination being able to form one or more hydrate.

As used herein, the term "analog" refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the replacement of one functional group by another functional group). Thus, an analog is a compound that is similar to or comparable in function and appearance to the reference compound.

The term "aprotic solvent," as used herein, refers to a solvent that is relatively inert to proton activity, i.e., not acting as a proton-donor. Examples include, but are not limited to, hydrocarbons, such as hexane and toluene, for example, halogenated hydrocarbons, such as, for example, methylene chloride, ethylene chloride, chloroform, and the like, heterocyclic compounds, such as, for example, tetrahydrofuran and N-methylpyrrolidinone, and ethers such as diethyl ether, bis-methoxymethyl ether. Such solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of aprotic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of

Purification . 4th ed., edited by John A. Riddick et al. , Vol. II, in the Techniques of Chemistry Series , John Wiley & Sons, NY, 1986.

The terms "protogenic organic solvent” or“protic solvent” as used herein, refer to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like. Such solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of protogenic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification , 4th ed., edited by John A. Riddick et al., Vol. II, in the Techniques of Chemistry Series . John Wiley & Sons, NY, 1986.

Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term“stable”, as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).

The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and variation of the reaction conditions can produce the desired bridged macrocyclic products of the present invention. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein include, for example, those described in R. Larock, Comprehensive Orsanic

Transformations , VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Orsanic Synthesis . 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reasents for Orsanic Synthesis . John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reasents for Orsanic Synthesis . John Wiley and Sons (1995).

The compounds of this invention may be modified by appending various

functionalities via synthetic means delineated herein to enhance selective biological properties. Such modifications include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.

PHARMACEUTICAL COMPOSITIONS

The pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term "pharmaceutically acceptable carrier" means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are sugars such as lactose, glucose and sucrose; starches such as com starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol; esters such as ethyl oleate and ethyl laurate; agar;

buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid;

pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. The pharmaceutical compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, or as an oral or nasal spray.

The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol,

tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.

The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.

Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.

Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.

The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to the compounds of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.

Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art. All publications, patents, published patent applications, and other references mentioned herein are hereby incorporated by reference in their entirety. Abbreviations

Abbreviations which have been used in the descriptions of the schemes and the examples that follow are:

ACN for acetonitrile;

BOP for benzotriazol-1-yloxy-tris(dimethylamino)phosphonium hexafluorophosphate;

Bn for benzyl;

BTC for bis(trichloromethyl)carbonate; triphosgene;

BzCl for benzoyl chloride;

CDI for carbonyldiimidazole;

DABCO for 1,4-diazabicyclo[2.2.2]octane;

DAST for diethylaminosulfur trifluoride;

DABCYL for 6-(N-4'-carboxy-4-(dimethylamino)azobenzene)- aminohexyl-;

1-O-(2-cyanoethyl)-(N,N-diisopropyl)-phosphoramidite;

DBU for 1, 8-Diazabicycloundec-7-ene; DCC for N, N'-dicyclohexylcarbodiimide;

DCM for dichloromethane;

DIAD for diisopropyl azodicarboxylate;

DIBAL-H for diisobutylaluminum hydride;

DIPEA for diisopropyl ethylamine;

DMAP for N,N-dimethylaminopyridine;

DME for ethylene glycol dimethyl ether;

DMEM for Dulbecco’s Modified Eagles Media;

DMF for N,N-dimethyl formamide;

DMSO for dimethylsulfoxide;

DPPA for diphenylphosphoryl azide or diphenyl phosphorylazidate;

DSC for N, N'-disuccinimidyl carbonate;

EDCI or EDC for 1-(3-diethylaminopropyl)-3-ethylcarbodiimide hydrochloride; EtOAc for ethyl acetate;

EtOH for ethyl alcohol;

HATU for O (7-Azabenzotriazole-1-yl)-N,N,N’,N’– tetramethyluronium hexafluorophosphate;

HCl for hydrochloric acid;

KHMDS is potassium bis(trimethylsilyl) amide;

PyBrOP for Bromo-tri-pyrolidino-phosphonium hexafluorophosphate;

Ph for phenyl;

RT for reverse transcription;

RT-PCR for reverse transcription-polymerase chain reaction;

TBME for tert-butyl methyl ether;

TCDI for 1,1’-thiocarbonyldiimidazole;

TEA for triethylamine;

Tf 2 O for trifluoromethanesulfonic anhydride;

TFA for trifluoroacetic acid;

THF for tetrahydrofuran;

TLC for thin layer chromatography;

(TMS) 2 NH for hexamethyldisilazane;

TMSOTf for trimethylsilyl trifluoromethanesulfonate;

TBS for t-Butyldimethylsilyl;

TMS for trimethylsilyl; TPAP tetrapropylammonium perruthenate;

TPP or PPh 3 for triphenylphosphine;

TrCl for trityl chloride;

Ts for p-CH 3 C 6 H 4 SO 2 - tBOC or Boc for tert-butyloxy carbonyl; and

Xantphos for 4,5-Bis-diphenylphosphanyl-9,9-dimethyl-9H-xanthene. Synthetic Methods

The compounds and processes of the present invention will be better understood in connection with the following synthetic schemes that illustrate the methods by which the compounds of the invention may be prepared, which are intended as an illustration only and not to limit the scope of the invention. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims. Scheme 1

Scheme 1 illustrates methods for preparing a compound of formula 11, wherein X is OTf or halogen; Y is hydrogen or methyl; n, R 1 , m, R 2 , v, R 3 , ring A and ring B are defined as previously. Deprotection as described in scheme 1 followed by installation of substituted or unsubstituted 4-nitrobenzoyl group provides compound 3. Compound 3 is converted to vinyl triflate or other vinyl halogen 4, which is coupled with using a suitable reagent such as a boronic ester, boronic acid, organotin reagent, organozinc reagent, organomagnesium reagent or the like, catalyzed by an appropriate Pd, Ni, or Cu catalyst or the like, to afford compounds of formula 5 (5 is converted to ester 6 if Y is H). Ester 6 is subsequently reduced with reducing agent such as but not limited to, iron, to give compound 7, which is coupled with functionalized acyl chloride 8 to afford compounds of formula 9. Hydrolysis the ester 9 with a base such as but not limited to, LiOH or NaOH, gives acid compound 10. Compounds of formula 11 are prepared from the reaction of compound 10 with Scheme 2

Scheme 2 illustrates methods to prepare compounds of formula 21, wherein X is OTf or halogen; Y is hydrogen or methyl; n, R 1 , m, R 2 , v, R 3 , ring A and ring B are defined as previously described. Selective methylation of 1 with MeI and a base, such as, but not limited to, t-BuONa, t-BuOK or NaH, gives a–methyl ketone 12. Deprotection as described in Scheme 1 followed by installation of substituted or unsubstituted 4-nitrobenzoyl group provides compound 14. With 14 in hand, compounds of formula 21 are prepared by adapting the similar conditions as described in scheme 1 for compounds of formula 11. Scheme 3

Scheme 3 illustrates another method to prepare compounds of formula 11, wherein X is OTf or halogen; Y is hydrogen or methyl; n, R 1 , m, R 2 , v, R 3 , ring A and ring B are defined as previously described. Protection of 2 with TsCl in the presence of a base, such as but not limited to, pyridine, affords 22 which is converted to vinyl triflate or other vinyl halogen 23. Compound 23 is coupled with using a suitable reagent such as a boronic ester, boronic acid, organotin reagent, organozinc reagent, organomagnesium reagent or the like, catalyzed by an appropriate Pd, Ni, Cu catalyst or the like to afford a compound of formula 24. Deprotection of tosyl group of 24 in acidic conditions such as but not limited to, H 2 SO 4 , provides 25, which reacts with to provide compound 26. Compound 26 is coupled with functionalized acyl chloride 27 to afford compounds of formula 11. EXAMPLES

The compounds and processes of the present invention will be better understood in connection with the following examples, which are intended as an illustration only and not limiting of the scope of the invention. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, formulations and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims.

Example 1

The synthesis of this compound is illustrated in Scheme 4. Scheme 4

Example 1 step a:

A solution of 1-benzyl-1,2,3,5-tetrahydro-4H-benzo[b]azepin-4-one (3.0 g, 11.9 mmol) and Pd/C (2.0 g) in EtOAc/t-BuOH (9:1, 30 mL) was stirred with hydrogen balloon for 16 h at room temperature. The palladium was filtered through Celite and the filtrate was concentrated to afford the desired product (crude) as yellow oil which was used directly for the next step. ESI-MS m/z: 162.15 [M+H] + .

Example 1 step b:

A solution of the compound from step a (crude), 4-nitrobenzoyl

chloride (5.76 g, 31.01 mmol), pyridine (2.45g, 31 mmol) in CH 2 Cl 2 (10 mL) was stirred for 2 h at room temperature. The resulting solution was concentrated by rotary evaporation. The crude product was purified by reverse phase C18 column chromatography (CH 3 CN/H 2 O) to afford the desired product (3.4 g) as yellow oil. ESI-MS m/z: 311.05 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) d 3.02-2.65 (m, 2H), 3.59 (s, 2H), 4.19 (s, 1H), 4.86 (s, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.06 (t, J = 8.0 Hz, 1H), 7.23 (t, J = 8.0 Hz, 1H), 7.32 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 8.0 Hz, 2H), 8.05 (d, J = 8.0 Hz, 2H).

Example 1 step c:

A reaction mixture of the compound from step b (3.4 g, 11.0 mmol) and

sodium hydride (1.05 g, 43.8 mmol) in DMF (30 mL) was stirred for 30 min at 0°C. Then 1,1,1-trifluoro-N-phenyl-N-trifluoromethanesulfonylmethanesu lfonamide (4.7 g, 13.1 mmol) was added and the resulting mixture was stirred overnight at room temperature. The residue was purified by reverse flash chromatography (MeCN/H 2 O) to give the desired product as a yellow solid (3.6 g, 74%). ESI-MS m/z: 443.10 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) d 2.92 (d, J = 16.0 Hz, 1H), 3.09 (t, J = 12.0 Hz, 1H), 3.43-3.28 (m, 1H), 5.17-5.02 (m, 1H), 6.63 (d, J = 8.0 Hz, 1H), 6.75 (s, 1H), 7.02 (t, J = 8.0 Hz, 1H), 7.23 (t, J = 8.0 Hz, 1H), 7.41-7.30 (m, 3H), 8.07 (d, J = 8.0 Hz, 2H).

Example 1 step d:

A solution of the compound from step c (2.0 g, 4.5 mmol), 5-(dihydroxyboranyl)thiophene-2- carboxylic acid (933 mg, 5.4 mmol), Cs 2 CO 3 (4.43 g, 13.6 mmol), Pd(PPh 3 ) 2 Cl 2 (317 mg, 0.45 mmol) in DMF (30 mL) was heated for 2 h at 80°C after degassed. The reaction mixture was used directly for the next step. ESI-MS m/z: 421.10 [M+H] + .

Example 1 step e:

The reaction mixture from step d, methyl iodide (2.19 g, 15.46 mmol) in DMF (20 mL) was stirred overnight at room temperature. The residue was purified by reverse flash

chromatography to give the desired product as a yellow solid (1.2 g). ESI-MS m/z: 435.15 [M+H] +

.

Example 1 step f:

A mixture of the compound from step f (1.20 g, 2.76 mmol), iron (1.54 g, 27.62 mmol), NH 4 Cl (1.48 g, 27.62 mmol) in EtOH (30 mL) and water (30 mL) was heated for 2 h at 80°C. After cooled down, the reaction mixture was passed through celite. The filtrate was evaporated and the residue was purified by reverse flash chromatography give the desired product as a yellow solid (950 mg, 85%). ESI-MS m/z: 405.15 [M+H] +

. Example 1 step g:

A mixture of 5-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid (405 mg, 1.0 mmol), (1-chloro-2-methylprop-1-en-1-yl)dimethylamine (401 mg, 3.0 mmol) in CH 2 Cl 2 (30 mL) was stirred for 1 h at room temperature. The solvent was evaporated and the residue was dried in vacuo. The residue was dissolved in CH 2 Cl 2 (30 mL) and cooled down to 0°C. After that, the compound from step f (263 mg, 1.0 mmol) and pyridine (0.5 mL) were added to the above solution and the mixture was stirred for 1 h at room temperature. The solvent was removed by rotary evaporation. The residue was purified by reverse flash chromatography to give the desired product as a yellow solid (510 mg, 79%). ESI-MS m/z: 649.35 [M+H] +

. Example 1 step h:

A mixture of the compound from step g (1.2 g, 1.85 mmol), LiOH (443 mg, 18.5 mmol) in methanol (30 mL) and H 2 O (30 mL) was stirred for 2 h at room temperature. The solution was neutralized to pH 7 and solvents were removed by rotary evaporation. The residue was purified by reverse flash chromatography to give the desired product as a yellow solid (850 mg, 72%). ESI-MS m/z: 635.20 [M+H] +

. Example 1 step i:

A mixture of the compound from step h (300 mg, 0.47 mmol) and (1-chloro-2-methylprop-1- en-1-yl)dimethylamine (189 mg, 1.42 mmol) in CH 2 Cl 2 (20 mL) was stirred for 30 min at room temperature. The solvent was evaporated and the residue was dried in vacuo. The r esidue was dissolved in CH2Cl2 (5 mL) and cooled down to 0°C and then

c yclopropanamine (135 mg, 2.36 mmol) and pyridine (0.5 mL) were added. The resulting mixture was stirred for 30 min at room temperature. After evaporated the solvent, the residue was purified by reverse flash chromatography to give the desired product as a white solid (230 mg, 72%). ESI-MS m/z: 674.40 [M+H] + 1

. H NMR (400 MHz, DMSO-d 6 ) d 0.61-0.55 (m, 2H), 0.76-0.68 (m, 2H), 1.63 (t, J = 4.0 Hz, 4H), 2.18 (s, 3H), 2.85-2.77 (m, 1H), 3.20- 3.00 (m, 4H), 3.46 (t, J = 4.0 Hz, 4H), 3.61 (s, 4H), 4.95 (s, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.0 Hz, 1H), 7.07 (d, J = 8.0 Hz, 2H), 7.20 (t, J = 8.0 Hz, 1H), 7.26 (s, 1H), 7.38 (d, J = 4.0 Hz, 1H), 7.56-7.47 (m, 3H), 7.61 (d, J = 8.0 Hz, 1H), 7.66 (d, J = 4.0 Hz, 1H), 8.04 (s, 1H), 8.49 (d, J = 4.0 Hz, 1H), 10.38 (s, 1H).

Example 2

Example 2 was prepared using a procedure similar to that used to prepare Example 1, where 2,6-difluoroaniline was used in place of cyclopropanamine in step i. ESI-MS m/z: 746.40 [M+H] + 1

. H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 4.0 Hz, 4H), 2.18 (s, 3H), 3.22-3.02 (m, 3H), 3.47 (t, J = 4.0 Hz, 4H), 3.62 (s, 4H), 4.98 (s, 1H), 6.74 (d, J = 8.0 Hz, 1H), 7.02 (t, J = 8.0 Hz, 1H), 7.09 (d, J = 8.0 Hz, 2H), 7.28-7.18 (m, 3H), 7.33 (s, 1H), 7.47-7.39 (m, 1H), 7.58-7.48 (m, 4H), 7.64 (d, J = 8.0 Hz, 1H), 7.97 (d, J = 4.0 Hz, 1H), 8.04 (s, 1H), 10.21 (s, 1H), 10.38 (s, 1H).

Example 3

Example 3 was prepared using a procedure similar to that used to prepare Example 1, where cyclobutanamine was used in place of cyclopropanamine in step i. ESI-MS m/z: 688.40 [M+H] + 1

. H NMR (400 MHz, DMSO-d 6 ) d 1.74-1.58 (m, 6H), 2.14-2.01 (m, 2H), 2.28-2.15 (m, 5H), 3.18-3.01 (m, 3H), 3.46 (t, J = 4.0 Hz, 4H), 3.61 (s, 4H), 4.45-4.31 (m, 1H), 4.94 (s, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 4.0 Hz, 1H), 7.07 (d, J = 8.0 Hz, 2H), 7.20 (t, J = 4.0 Hz, 1H), 7.26 (s, 1H), 7.40 (d, J = 4.0 Hz, 1H), 7.57-7.47 (m, 3H), 7.61 (d, J = 8.0 Hz, 1H), 7.74 (d, J = 4.0 Hz, 1H), 8.05 (s, 1H), 8.64 (d, J = 8.0 Hz, 1H), 10.38 (s, 1H).

Example 4

Example 4 was prepared using a procedure similar to that used to prepare Example 1, where 3,3-difluorocyclobutan-1-amine was used in place of cyclopropanamine in step i. ESI-MS m/z: 724.40 [M+H] + 1

. H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 4.0 Hz, 4H), 2.18 (s, 3H), 2.85-2.68 (m, 2H), 3.13-2.90 (m, 5H), 3.46 (t, J = 4.0 Hz, 4H), 3.61 (s, 4H), 4.29-4.21 (m, 1H), 4.95 (s, 1H), 6.73 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.0 Hz, 1H), 7.07 (d, J = 8.0 Hz, 2H), 7.20 (t, J = 8.0 Hz, 1H), 7.28 (s, 1H), 7.43 (d, J = 4.0 Hz, 1H), 7.58-7.47 (m, 3H), 7.61 (d, J = 8.0 Hz, 1H), 7.73 (d, J = 4.0 Hz, 1H), 8.29 (s, 1H), 8.87 (t, J = 4.0 Hz, 1H), 10.38 (s, 1H).

Example 5

Example 5 was prepared using a procedure similar to that used to prepare Example 1, where bicyclo[1.1.1]pentan-1-amine was used in place of cyclopropanamine in step i. ESI-MS m/z: 700.40 [M+H] + 1 H NMR (400 MHz, DMSO-d 6 ) d 1.66 (t, J = 4.0 Hz, 4H), 2.09 (s, 6H), 2.19 (s, 3H), 3.12-3.01 (m, 3H), 3.47 (t, J = 4.0 Hz, 4H), 3.71 (s, 4H), 4.95 (s, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 7.09 (d, J = 8.0 Hz, 2H), 7.19 (t, J = 8.0 Hz, 1H), 7.25 (s, 1H), 7.38 (d, J = 4.0 Hz, 1H), 7.53 (d, J = 8.0 Hz, 2H), 7.61 (d, J = 8.0 Hz, 2H), 7.72-7.65 (m, 2H), 8.02 (s, 1H), 8.99 (s, 1H), 10.48 (s, 1H). .

Example 6

Example 6 was prepared using a procedure similar to that used to prepare Example 1, where 2-fluoro-6-methylaniline was used in place of cyclopropanamine in step i. ESI-MS m/z: 742.40 [M+H] + 1

. H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 4.0 Hz, 4H), 2.18 (s, 3H), 2.25 (s, 3H), 3.19-3.05 (m, 3H), 3.47 (t, J = 4.0 Hz, 4H), 3.62 (s, 4H), 4.97 (s, 1H), 6.74 (d, J = 8.0 Hz, 1H), 7.02 (t, J = 8.0 Hz, 1H), 7.09 (t, J = 8.0 Hz, 2H), 7.16 (d, J = 8.0 Hz, 2H), 7.20 (t, J = 8.0 Hz, 1H), 7.34-7.25 (m, 2H), 7.49 (d, J = 4.0 Hz, 2H), 7.54 (d, J = 8.0 Hz, 2H), 7.63 (d, J = 8.0 Hz, 1H), 7.95 (s, 1H), 8.04 (s, 1H), 9.96 (s, 1H), 10.38 (s, 1H). Example 7

Example 7 was prepared using a procedure similar to that used to prepare Example 2, where 6-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid was used in place of 5- methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid in step g. ESI-MS m/z: 746.45 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.64 (t, J = 4.0 Hz, 4H), 2.36 (s, 3H), 3.22-3.02 (m, 3H), 3.48 (t, J = 4.0 Hz, 4H), 3.68 (s, 4H), 4.97 (s, 1H), 6.60 (d, J = 8.0 Hz, 1H), 6.74 (d, J = 8.0 Hz, 1H), 7.02 (t, J = 8.0 Hz, 1H), 7.08 (t, J = 8.0 Hz, 2H), 7.28-7.17 (m, 3H), 7.33 (s, 1H), 7.47-7.39 (m, 1H), 7.51 (d, J = 4.0 Hz, 1H), 7.54 (d, J = 8.0 Hz, 2H), 7.68- 7.57 (m, 2H), 7.97 (d, J = 4.0 Hz, 1H), 10.22 (s, 1H), 10.35 (s, 1H). Example 8

Example 8 was prepared using a procedure similar to that used to prepare Example 6, where 6-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid was used in place of 5- methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid in step g. ESI-MS m/z: 742.50 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 4.0 Hz, 4H), 2.25 (s, 3H), 2.33 (s, 3H), 3.19-3.05 (m, 3H), 3.47 (t, J = 4.0 Hz, 4H), 3.64 (s, 4H), 4.97 (s, 1H), 6.57 (d, J = 8.0 Hz, 1H), 6.74 (d, J = 8.0 Hz, 1H), 7.02 (t, J = 8.0 Hz, 1H), 7.08 (t, J = 8.0 Hz, 2H), 7.16 (d, J = 8.0 Hz, 2H), 7.21 (t, J = 8.0 Hz, 1H), 7.34-7.25 (m, 2H), 7.49 (d, J = 4.0 Hz, 1H), 7.54 (d, J = 8.0 Hz, 3H), 7.63 (d, J = 8.0 Hz, 1H), 7.95 (s, 1H), 9.96 (s, 1H), 10.38 (s, 1H). Example 9

Example 9 was prepared using a procedure similar to that used to prepare Example 4, where 6-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid was used in place of 5- methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid in step g. ESI-MS m/z: 724.45 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 4.0 Hz, 4H), 2.33 (s, 3H), 2.85-2.60 (m, 2H), 3.20-2.90 (m, 5H), 3.47 (t, J = 4.0 Hz, 4H), 3.64 (s, 4H), 4.35-4.15 (m, 1H), 4.97 (s, 1H), 6.57 (d, J = 8.0 Hz, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.15-7.01 (m, 3H), 7.22 (t, J = 8.0 Hz, 1H), 7.26 (s, 1H), 7.40 (d, J = 4.0 Hz, 1H), 7.56-7.50 (m, 3H), 7.60 (d, J = 8.0 Hz, 1H), 7.74 (d, J = 4.0 Hz, 1H), 8.84 (d, J = 8.0 Hz, 1H), 10.26 (s, 1H). Example 10

Example 10 was prepared using a procedure similar to that used to prepare Example 3, where 6-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid was used in place of 5- methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid in step g. ESI-MS m/z: 688.50 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.74-1.58 (m, 6H), 2.14-2.02 (m, 2H), 2.27- 2.16 (m, 2H), 2.33 (s, 3H), 3.14-2.98 (m, 3H), 3.47 (t, J = 4.0 Hz, 4H), 3.64 (s, 4H), 4.45- 4.32 (m, 1H), 4.95 (s, 1H), 6.57 (d, J = 8.0 Hz, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.0 Hz, 1H), 7.07 (t, J = 8.0 Hz, 2H), 7.20 (t, J = 8.0 Hz, 1H), 7.26 (s, 1H), 7.40 (d, J = 4.0 Hz, 1H), 7.56-7.50 (m, 3H), 7.60 (d, J = 8.0 Hz, 1H), 7.74 (d, J = 4.0 Hz, 1H), 8.63 (d, J = 8.0 Hz, 1H), 10.29 (s, 1H).

Example 11

Example 11 was prepared using a procedure similar to that used to prepare Example 3, where 6-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid was used in place of 5-methyl-2- (7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid in step g. ESI-MS m/z: 700.45 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 4.0 Hz, 4H), 2.09 (s, 6H), 2.33 (s, 3H), 2.47 (s, 1H), 3.18-2.98 (m, 3H), 3.47 (t, J = 4.0 Hz, 4H), 3.64 (s, 4H), 4.94 (s, 1H), 6.57 (d, J = 8.0 Hz, 1H), 6.72 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.0 Hz, 1H), 7.07 (d, J = 8.0 Hz, 2H), 7.19 (t, J = 8.0 Hz, 1H), 7.25 (s, 1H), 7.39 (d, J = 4.0 Hz, 1H), 7.57-7.50 (m, 3H), 7.61 (d, J = 8.0 Hz, 1H), 7.69 (d, J = 4.0 Hz, 1H), 8.99 (s, 1H), 10.30 (s, 1H). Example 12

Example 12 was prepared using a procedure similar to that used to prepare Example 1, where methyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)thiophene-3-c arboxylate was used in place of 5-(dihydroxyboranyl)thiophene-2-carboxylic acid in step b. ESI-MS m/z: 674.35 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 0.51– 0.60 (m, 2H), 0.71 (dt, J = 6.9, 3.3 Hz, 2H), 1.66 (t, J = 5.2 Hz, 4H), 2.19 (s, 3H), 2.81 (m, 1H), 3.05 (s, 3H), 3.47 (t, J = 5.1 Hz, 4H), 3.70 (s, 4H), 4.95 (s, 1H), 6.73 (d, J = 7.9 Hz, 1H), 7.00 (t, J = 7.6 Hz, 1H), 7.08 (d, J = 8.3 Hz, 2H), 7.15 (s, 1H), 7.19 (td, J = 7.6, 1.3 Hz, 1H), 7.53 (d, J = 8.3 Hz, 2H), 7.56– 7.62 (m, 1H), 7.66 (s, 1H), 7.73 (d, J = 1.4 Hz, 1H), 8.03 (dd, J = 4.7, 1.7 Hz, 2H), 8.26 (d, J = 4.1 Hz, 1H), 10.47 (s, 1H). Example 13

Example 13 step a:

To a round-bottom flask (100 mL) were added 1-benzyl-3,5-dihydro-2H-1-benzazepin-4-one (1.50 g, 5.97 mmol), t-BuONa (0.57 g, 5.97 mmol), CH 3 I (0.85 g, 5.97 mmol) and THF (40 mL) at room temperature. The resulting mixture was stirred for 20 min at 70 o C and then diluted with brine (50 mL). After extracted with EtOAc (3 x 100 mL), the combined organic layers were dried, evaporated and purified by reverse flash chromatography to afford the crude product (1.1 g) as yellow oil. ESI-MS m/z: 266.25 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.34 (d, J = 7.0 Hz, 3H), 2.29 (dt, J = 12.5, 6.0 Hz, 1H), 2.58 (dt, J = 12.9, 7.6 Hz, 1H), 3.10 (dd, J = 7.5, 6.0 Hz, 2H), 3.84 (q, J = 7.0 Hz, 1H), 4.21– 4.36 (m, 2H), 7.03 (td, J = 7.3, 1.4 Hz, 1H), 7.13– 7.27 (m, 4H), 7.28– 7.38 (m, 4H).

Example 13 step b:

To a round-bottom flask (100 mL) were added the compound from step a (1.10 g, 4.14 mmol), EtOAc (40 mL) and t-BuOH (5 mL) at room temperature. Then Pd/C (2.21 g) was added under nitrogen atmosphere at room temperature and the resulting mixture was stirred for 2 h at room temperature under H 2 atmosphere. The reaction mixture was passed through Celite and the filter cake was washed with EtOAc (3 x 10 mL). The filtrate was concentrated under reduced pressure to give the crude product (800 mg) as yellow oil. ESI-MS m/z: 176.10 [M+H] + . Example 13 step c:

To a round-bottom flask (100 mL) were added the compound from step b (800 mg, 4.56 mmol), 4-nitro-(1.27 g, 6.84 mmol), pyridine (722 mg, 9.13 mmol) and CH 2 Cl 2 (35 mL) at 0 o C. The resulting mixture was stirred for 30 min at room temperature. Then the mixture was concentrated under reduced pressure and the residue was purified by reverse flash and Prep- TLC (CH 2 Cl 2 / MeOH 15:1) to afford the desired product (900 mg) as a yellow solid. ESI-MS m/z: 325.15 [M+H] + .

Example 13 step d:

To a round-bottom flask (100 mL) were added the compound from step c (1.14 g, 3.51 mmol) and DMF (40 mL) at room temperature. After the solution was cooled down to 0 o C, t-BuONa (1.01 g, 10.54 mmol) and 1,1,1-trifluoro-N-phenyl-N-trifluoromethanesulfonylmethane- sulfonamide (2.51 g, 7.02 mmol) were added. The resulting mixture was slowly warmed to room temperature and stirred further for 30 min. The reaction mixture was quenched with water and extracted with EtOAc (5 x 200 mL). After separation, the combined organic layers were dried, filtered and concentrated under reduced pressure. The residue was purified by reverse flash chromatography to afford the desired product (1.0 g) as a brown yellow solid. ESI-MS m/z: 457.05[M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 2.30 (d, J = 1.6 Hz, 3H), 2.57– 2.82 (m, 2H), 3.71 (ddd, J = 13.0, 6.3, 2.8 Hz, 1H), 4.87 (td, J = 12.6, 6.1 Hz, 1H), 7.04 (dd, J = 7.9, 1.4 Hz, 1H), 7.13 (td, J = 7.6, 1.5 Hz, 1H), 7.23– 7.41 (m, 3H), 7.59 (dd, J = 7.9, 1.5 Hz, 1H), 8.00– 8.13 (m, 2H). Example 13 step e:

To a round-bottom flask (100 mL) were added the compound from step d (500 mg, 1.09 mmol), 5-(dihydroxyboranyl)thiophene-2-carboxylic acid (282 mg, 1.64 mmol,), Pd(dppf)Cl 2 (80 mg, 0.11 mmol), Cs 2 CO 3 (714 mg, 2.19 mmol) and DMF (40 mL) at room temperature. After degassed, the mixture was heated for 2 h at 100 o C. The resulting mixture was used in next step without purification. ESI-MS m/z: 435.10[M+H] + .

Example 13 step f:

The mixture from step e was cooled down to room temperature, and then CH 3 I (1 mL) was added. The resulting mixture was stirred for 1 h at room temperature. The solid was filtered out through celite. The filter cake was washed with EtOAc (3 x 30 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reverse flash

chromatography to afford the crude product (280 mg) as a dark green solid. ESI-MS m/z: 449.10 [M+H] + .

Example 13 step g:

To a round-bottom flask (100 mL) were added the compound from step f (260 mg, 0.58 mmol), Fe (323 mg, 5.79 mmol), NH 4 Cl (310 mg, 5.79 mmol), H 2 O (3 mL) and EtOH (25 mL) at room temperature. The resulting mixture was heated for 30 min at 80 o C. After cooled down, the mixture was concentrated under vacuum. The stick residue was washed with CH 2 Cl 2 (4 x 50 mL). The combined CH 2 Cl 2 was passed through celite and the filtrate was concentrated under reduced pressure. The obtained residue was purified by prep-TLC

(CH 2 Cl 2 /MeOH 15 :1) to afford the desired product (220 mg) as a yellow solid. ESI-MS m/z: 419.10 [M+H] + . Example 13 step h:

A mixture of 5-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinic acid (179 mg, 0.68 mmol) and (1-chloro-2-methylprop-1-en-1-yl)dimethylamine (210 mg, 1.57 mmol) in CH 2 Cl 2 (10 mL) was stirred for 1 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated and the residue was dried in vacuo. The residue was dissolved in CH 2 Cl 2 (5 mL) and cooled down to 0℃. To the solution were added the compound from step g (220 mg, 0.52 mmol), CH 2 Cl 2 (10 mL) and pyridine (1 mL). The resulting mixture was stirred for 2 h at room temperature and then concentrated. The obtained residue was purified by prep-TLC (CH 2 Cl 2 / MeOH 15:1) to get the desired product (300 mg) as a yellow solid. ESI-MS m/z: 663.20 [M+H] + .

Example 13 step i:

To a 100 mL round-bottom flask were added the compound from step h (300 mg, 0.45 mmol), MeOH (30 mL) and a solution of LiOH (108 mg, 4.52 mmol) in H 2 O (5 mL). The mixture was stirred overnight at room temperature and then acidified to pH 5 with HCl (2M aq.). The mixture was concentrated under reduced pressure and the residue was purified by reverse flash chromatography to afford the desired product (270 mg) as a yellow solid. ESI- MS m/z: 649.35 [M+H] + . Example 13 step j:

To a sealed tube (8 mL) were added the compound from step i (60 mg, 0.09 mmol), (1- chloro-2-methylprop-1-en-1-yl)dimethylamine (37 mg, 0.27 mmol) and CH 2 Cl 2 (2 mL) at room temperature. The resulting mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated and the residue was dried in vacuo. The residue was d issolved in CH2Cl2 (2 mL) and cooled down to 0℃. To the mixture were added

c yclopropanamine (31 mg, 0.55 mmol) and pyridine (0.5 mL) which was stirred for 1 h at room temperature. After evaporated the solvent, the residue was purified directly by prep- TLC (CH 2 Cl 2 / MeOH 15:1) and prep-HPLC to afford the desired product as a white solid. ESI-MS m/z: 688.50 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 0.55– 0.62 (m, 2H), 0.71 (dt, J = 6.9, 3.4 Hz, 2H), 1.63 (t, J = 5.2 Hz, 4H), 2.17 (s, 3H), 2.40 (s, 4H), 2.65 (d, J = 17.8 Hz, 1H), 2.81 (td, J = 7.2, 3.7 Hz, 1H), 3.47 (t, J = 5.2 Hz, 4H), 3.61 (s, 4H), 3.76 (s, 1H), 4.56 (s, 1H), 6.87 (s, 1H), 7.08 (s, 3H), 7.31 (d, J = 8.9 Hz, 2H), 7.40– 7.58 (m, 4H), 7.72 (d, J = 4.0 Hz, 1H), 8.04 (d, J = 2.2 Hz, 1H), 8.38– 8.54 (m, 1H), 10.35 (s, 1H).

Example 14

Example 14 was prepared using a procedure similar to that used to prepare Example 13, where 2-fluoro-6-methylaniline was used in place of cyclopropanamine. ESI-MS m/z:

756.50 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.63 (t, J = 5.1 Hz, 4H), 2.17 (s, 3H), 2.26 (s, 3H), 2.43 (s, 4H), 2.67 (s, 1H), 3.47 (d, J = 5.3 Hz, 4H), 3.61 (s, 4H), 3.77 (d, J = 13.6 Hz, 1H), 4.60 (s, 1H), 6.89 (s, 1H), 7.13 (dd, J = 19.5, 10.4 Hz, 5H), 7.24– 7.42 (m, 3H), 7.43– 7.59 (m, 4H), 8.02 (dd, J = 18.2, 3.0 Hz, 2H), 9.98 (s, 1H), 10.35 (s, 1H).

Example 15

Example 15 step a:

A solution of the compound from Example 1 step a (1.35 g, 8.35 mmol) and TsCl (3.5 g, 18.61 mmol) in pyridine (10 mL) was stirred for 2 h at room temperature. The resulting solution was extracted with EtOAc (3 x 50 mL). The combined organic layer was washed with brine, dried, filtered and concentrated. The crude product was purified by reverse phase C18 column chromatography (CH 3 CN/H 2 O) to afford the desired product (1.4 g) as yellow oil. ESI-MS m/z: 316.20 [M+H] + . Example 15 step b:

A solution of the compound of step a (1.4 g, 4.44 mmol) in THF (50 mL) was cooled downed to -78 o C and LiHMDS (5.3 mL, 5.33 mmol) was added. The resulting mixture was stirred for 1 h at -78 o C. After that, 1,1,1-trifluoro-N-phenyl-N- trifluoromethanesulfonylmethanesulfonamide (1.5 g, 4.18 mmol) in THF (20 mL) was gradually added into the solution and stirred for 1 h. The resulting solution was diluted with saturated NH 4 Cl solution and extracted with EtOAc (3 x 50 mL). The combined organic layer was washed with brine, dried, filtered and concentrated. The crude product was purified by reverse phase C18 column chromatography (CH 3 CN/H 2 O) to afford the desired product (980 mg, 49%) as yellow oil. ESI-MS m/z: 448.15 [M+H] + .

Example 15 step c:

A solution of the compound from step b (150 mg, 0.33 mmol) and 4- (methoxycarbonyl)phenylboronic acid (120 mg, 0.67 mmol), Pd(PPh 3 ) 2 Cl 2 (47 mg, 0.07 mmol) and Cs 2 CO 3 (328 mg, 1.00 mmol) in DMF (3 mL) with an inert atmosphere of nitrogen was heated for 2 h at 80℃. The resulting solution was extracted with EtOAc (3 x 20 mL), and the combined organic layer was washed with brine, dried, filtered and concentrated to afford the desired product (crude) as a yellow solid. ESI-MS m/z: 434.25 [M+H] + .

Example 15 step d:

A solution of the compound from step c (crude) in H 2 SO 4 (90%) (4.00 mL) was heated for 16 h at 70℃. The pH of the resulting solution was adjusted to ~6 with NaHCO 3 (aq) and then extracted with EtOAc (3 x 20 mL). The combined organic layer was washed with brine, dried, filtered and concentrated. The crude product was purified by reverse phase C18 column chromatography (CH 3 CN/H 2 O) to afford the desired product (50 mg, 56%) as a yellow solid. ESI-MS m/z: 266.20 [M+H] + . Example 15 step e:

A solution of the compound from step d (50 mg, 0.19 mmol), cyclopropanamine (54 mg, 0.94 mmol), DIPEA (73 mg, 0.56 mmol) and HATU (107 mg, 0.28 mmol) in DMF ( 2 mL) was stirred for 2 h at room temperature. The solution was purified by reverse phase C18 column chromatography (CH 3 CN/H 2 O) to give the desired compound as a white solid (40 mg, 69%). ESI-MS m/z: 305.25 [M+H] + .

Example 15 step f:

A solution of 4-(5-methyl-2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)nicotinamido) benzoic acid (100 mg, 0.26 mmol) and (1-chloro-2-methylprop-1-en-1-yl)dimethylamine (105 mg, 0.79 mmol) in CH 2 Cl 2 (10 mL) was stirred for 2 h at room temperature. Then the resulting mixture was concentrated under vacuum and the residue was dried in vacuo. The residue was dissolved in CH 2 Cl 2 (5 mL) and cooled down to 0℃. After that, the compound from step e (40 mg, 0.13 mmol) and pyridine (31 mg, 0.39 mmol) in CH 2 Cl 2 (10 mL) were added. The resulting solution was stirred for 1 h at 0℃ and then concentrated. The crude product was purified by Prep-HPLC (CH 3 CN/H 2 O) to afford the desired product (25.3 mg, 23%) as a yellow solid. ESI-MS m/z: 668.50 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 0.61 (s, 2H), 0.68– 0.74 (m, 2H), 1.65 (s, 4H), 2.19 (s, 3H), 2.68 (s,1H), 3.05 (s, 2H), 3.47 (s, 6H), 3.65 (s, 4H), 4.95 (s, 1H), 6.74 (d, J = 8.6 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 7.11 (d, J = 6.7 Hz, 3H), 7.20 (t, J = 7.5 Hz, 1H), 7.54 (d, J = 8.3 Hz, 2H), 7.61 (d, J = 7.3 Hz, 1H), 7.75 (d, J = 8.2 Hz, 2H), 7.88 (d, J = 8.2 Hz, 2H), 8.04 (s, 1H), 8.47 (d, J = 4.4 Hz, 1H), 10.41 (s, 1H). Example 16

Example 16 was prepared using a procedure similar to that used to prepare Example 15, where 2-fluoro-6-methylaniline was used in place of cyclopropanamine in step e. ESI-MS m/z: 736.40 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.64 (t, J = 5.2 Hz, 4H), 2.18 (s, 3H), 2.26 (s, 3H), 3.09 (s, 3H), 3.47 (t, J = 5.1 Hz, 4H), 3.62 (s, 4H), 5.00 (s, 1H), 6.75 (d, J = 7.9 Hz, 1H), 7.02 (t, J = 7.7 Hz, 1H), 7.09– 7.32 (m, 7H), 7.47– 7.59 (m, 3H), 7.60– 7.66 (m, 1H), 7.84 (d, J = 8.2 Hz, 2H), 8.02– 8.09 (m, 3H), 9.94 (s, 1H), 10.39 (s, 1H).

Example 17

Example 17 was prepared using a procedure similar to that used to prepare Example 6, where 2-fluoro-6-methylaniline was used in place of 5-(dihydroxyboranyl)thiophene-2-carboxylic acid in step d. ESI-MS m/z: 754.35 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.67 (t, J = 5.2 Hz, 4H), 2.20 (s, 3H), 2.26 (s, 3H), 2.95 (d, J = 71.7 Hz, 3H), 3.48 (t, J = 5.3 Hz, 4H), 3.71 (s, 4H), 4.95 (s, 1H), 6.78 (d, J = 8.0 Hz, 1H), 6.86 (s, 1H), 7.04 (t, J = 7.6 Hz, 1H), 7.16 (t, J = 8.9 Hz, 4H), 7.21 (t, J = 7.5 Hz, 1H), 7.25– 7.34 (m, 1H), 7.55 (t, J = 8.5 Hz, 3H), 7.67 (s, 1H), 7.74 (t, J = 7.9 Hz, 1H), 7.90 (dd, J = 16.6, 9.8 Hz, 2H), 8.04 (d, J = 2.2 Hz, 1H), 10.05 (s, 1H), 10.50 (s, 1H). Example 18

Example 18 was prepared using a procedure similar to that used to prepare Example 6, where 2-chloro-6-methylaniline was used in place of 5-(dihydroxyboranyl)thiophene-2-carboxylic acid in step d. ESI-MS m/z: 770.50 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.64 (t, J = 4.0 Hz, 4H), 2.18 (s, 3H), 2.26 (s, 3H), 3.19-2.73 (m, 3H), 3.48 (t, J = 4.0 Hz, 4H), 3.63 (s, 4H), 4.96 (s, 1H), 6.62 (s, 1H), 6.78 (d, J = 8.0 Hz, 1H), 7.03 (t, J = 8.0 Hz, 1H), 7.22-7.11 (m, 5H), 7.32-7.24 (m, 1H), 7.52-7.46 (m, 2H), 7.56 (d, J = 8.0 Hz, 2H), 7.67 (d, J = 8.0 Hz, 1H), 8.08-7.98 (m, 2H), 8.14 (s, 1H), 10.08 (s, 1H), 10.39 (s, 1H).

Example 19

Example 19 was prepared using a procedure similar to that used to prepare Example 6, where 2-methyl-6-methylaniline was used in place of 5-(dihydroxyboranyl)thiophene-2-carboxylic acid in step d. ESI-MS m/z: 750.60 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) d 1.64 (t, J = 4.0 Hz, 4H), 2.18 (s, 3H), 2.25 (s, 3H), 2.40 (s, 3H), 3.20-2.72 (m, 3H), 3.48 (t, J = 4.0 Hz, 4H), 3.63 (s, 4H), 4.93 (s, 1H), 6.51 (s, 1H), 6.77 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 8.0 Hz, 1H), 7.21-7.10 (m, 5H), 7.42 (d, J = 8.0 Hz, 1H), 7.47 (d, J = 4.0 Hz, 1H), 7.51 (s, 1H), 7.57 (d, J = 8.0 Hz, 2H), 7.86 (d, J = 8.0 Hz, 1H), 7.91 (s, 1H), 8.05 (s, 1H), 9.88 (s, 1H), 10.40 (s, 1H). ASSAYS

INTRODUCTION RSV is a single stranded negative sense RNA virus that causes respiratory tract infections which can be dangerous to infants, the elderly, and immunosuppressed individuals. Currently there is no vaccine, and therapeutic options are both costly and of limited effectiveness. These approved treatments are Ribavirin, and Palivizumab/Synagis (a monoclonal antibody). RSV has two genotypes, A and B, which differ primarily in the structure of the virus’ surface“G” attachment protein. Our current primary screen focusses on RSV-A and uses an in vitro cytoprotection assay where compounds are added in 2-fold dilutions to cells which are then subjected to fully replicative viral particles. Cell viability is measured several days later along with separate measurements of compound cytotoxicity. This report focuses on the results of our most recent screening of compounds. METHODS HEp-2 cells, (originally derived from tumors grown in irradiated-cortisonised weanling rats that had been injected with epidermoid carcinoma tissue from a 56 year old male’s larynx, but later found to be indistinguishable from HeLa cells by PCR DNA analysis), were used for the culturing of genotype A,“Long” strain RSV. Flasks were inoculated with RSV and viral stocks were collected once cytopathic effect (CPE) was greater than 90%. Viral stocks in 25% sucrose media were snap frozen using liquid nitrogen to increase viral stability. Viral stock titers were quantified by tissue culture infectious dose 50% (TCID 50 ) using 8,000 cells per well and 3-fold viral dilutions across a 96-well plate, cultured for 4 days.

The control compound currently used in the RSV assay is RSV-604, a ~2.4 µM EC 50 nucleocapsid inhibitor previously developed by Novartis. Following extensive parameter testing, the final assay is run as follows: HEp-2 cells are seeded into the inner 60 wells of a 96-well plate at 8,000 cells per well in a volume of 50 µL using Growth Media (DMEM without phenol red, 1% L-Glut, 1% Penn/Strep, 1% nonessential amino acids, 10% FBS).2- Fold serial dilutions of control and test compounds are added to the wells in duplicate in a total volume of 25 µL. Viral stock is then added to the wells in a volume of 25 µL, bringing the total volume of each well to 100 µL. Each 96-well plate has a control column of 6 wells with cells and virus but no compound (negative control, max CPE), a column with cells but no compound or virus (positive control, minimum CPE), and a column with no cells or virus or compound (background plate/reagent control). The control wells with cells but no virus are given an additional 25 uL of growth media containing an equal quantity of sucrose as those wells receiving the viral stock in order to keep consistent in media and volume conditions. The outer wells of the plate are filled with 125 µL of growth media to act as a thermal and evaporative moat around the test wells. Following a 4-day incubation period, the plates are read using ATPlite (50 uL added per well), which quantifies the amount of ATP (a measure of cell health) present in each well. Assay plates are read using the Envision luminometer. In parallel, cytotoxicity is examined on an additional 96-well plate treated in an identical manner, but substituting the 25 µL of viral stock for 25 µL of growth media. These data are used to calculate the EC 50 of each compound. EC 50 ranges are as follows: A < 0.01 µM; B 0.01-0.05 µM; C > 0.05 µM.

Table 1 Summary of Activities

. While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.