Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ADAMATANE DERIVATIVES FOR THE TREATMENT OF FILOVIRUS INFECTION
Document Type and Number:
WIPO Patent Application WO/2017/127306
Kind Code:
A1
Abstract:
Compounds of structural Formula (I) were developed for the treatment of infections by filoviruses including Ebolavirus and Marburgvirus.

Inventors:
PLEWE MICHAEL (US)
BROWN ERIC (US)
GANTLA VIDYASAGAR (US)
HENKEL GREGORY (US)
MCCORMACK KENNETH (US)
SOKOLOVA NADEZDA (US)
Application Number:
PCT/US2017/013560
Publication Date:
July 27, 2017
Filing Date:
January 13, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ARISAN THERAPEUTICS (US)
International Classes:
A61K31/015; A61K31/135; A61K31/165; C07C13/28; C07C13/54; C07C13/573
Domestic Patent References:
WO2015057068A12015-04-23
Foreign References:
US20040209858A12004-10-21
US20150166532A12015-06-18
US20120135954A12012-05-31
US20090218935A12009-09-03
Other References:
See also references of EP 3405183A4
Download PDF:
Claims:
WHAT IS CLAIMED IS:

A method of treating infections associated with viruses of the Filoviridae enveloped virus, or any virus expressing filovirus glycoproteins to mediate cell entry comprising administration of a therapeutically effective amount of a compound of Structural Formula I

I

or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D, and Y is a bond or CR4R5; or

X is CR5 and Y is CR4-A-D;

A is -C(R6aR6b)r, (C6to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R6a group;

D is selected from the group consisting of

R8 R8

Ν R10b ^NL^R 03 R8 O O o o

d/ v , ¾- 10b , ^R1Q , 4R10b , ι<» , ¾^Riob , ¾s.Rl0b t ¾s Rl0b j

R is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cyclo- alkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nCO)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cyclo- alkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R3 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group; R4 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cyclo-alkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and -

NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R5 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cyclo-alkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R6a and R6bis independently selected from hydrogen, halogen, OH, nitro, CF3, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, -C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, - (CH2)nC(0)OR12, -(CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R7aand R7b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, NR aR b, (Csto C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group,

or R7a and R7b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) membered cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein the said (C2to C 0) membered cycloheteroalkyi ring is optionally substituted with at least one R 3 group;

each R8 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to Cio) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group;

each of the R9a, R9b, and R9cis independently selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to Cio) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to Co) heteroaryl, -C(0)R12 -C(0)NR aR b,-S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, - (CH2)nC(0)OR12, -(CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and

-NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 0aand R 0bis independently selected from hydrogen, (C^ to C 0) alkyl, (Cito

C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R aand R b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to C9) heteroaryl, (C6to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group,

or R a and R b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group c consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 3 group; each of the R 2 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to Cio) alkynyl, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6 to C 0) aryl, and (C2to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Csto Cio) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

each R 3 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 4aR 4b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R15, -C(0)NR 4aR 4b, -S(0)mR15, -S(0)mNR 4aR 4b, -NR 4aS(0)mR15, - (CH2)nC(0)OR15, -(CH2)nC(0)N(R 4aR 4b), -(CH2)nN(R 4aR 4b), -OC(0)R15, -0(CH2)nO-, - NR 4aC(0)R15, and -NR 4aC(0)N(R 4aR 4b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3to C10) cycloalkylene, and (C2to C 0) cycloheteroalkylene is optionally substituted with at least one R 6 group;

each of the R 4aand R 4b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 6 group,

or R 4a and R 4b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 6 group; each R 5 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 6 group;

each R 6 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 7aR 7b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R18, -C(0)NR 7aR 7b, -S(0)mR18, -S(0)mNR 7aR 7b, -NR 7aS(0)mR18, -(CH2)nC(0)OR18, -(CH2)nC(0)N(R 7aR 7b), -(CH2)nN(R 7aR 7b), -OC(0)R18, -NR 7aC(0)R18, and -NR 7aC(0)N(R 7aR 7b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C2to C9) heteroaryl, (C6 to C 0) aryl, (C6to C 0) arylene, (C2to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2to C10) cycloheteroalkylene is optionally substituted with at least one R 9 group;

each of the R 7aand R 7b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^o C 0) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 9 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 9 group; each R 8 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi,

(Csto C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C2 to C9) heteroaryl, and (C6 to C 0) aryl is optionally substituted with at least one R 9 group;

each R 9 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR20aR20b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R21, -C(O)NR20aR20b, -S(0)mR21, -S(O)mNR20aR20b, - NR20aS(O)mR21, -(CH2)nC(0)OR21, -(CH2)nC(O)N(R20aR20b), -(CH2)nN(R20aR20b), -OC(0)R21, - NR20aC(O)R21, and -NR20aC(O)N(R20aR20b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R22 group; each of the R20a and R20b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to Cio) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C 0) cycloalkyi, (Cs to Cio) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R22 group,

or R20a and R20b may be taken together with the nitrogen atom to which they are attached to form a (C2 to C 0) cycloheteroalkyi ring, wherein

said (C2 to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2 to C 0) cycloheteroalkyi ring is optionally substituted with at least one R22 group; each R2 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

each R22 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR23aR23b, oxo,

(Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R24,-C(0)NR23aR23b, -S(0)mR24, -S(0)mNR23aR23b, -NR23aS(0)mR24, - -(CH2)nC(0)OR24, -(CH2)nC(0)N(R23aR23b), -(CH2)nN(R23aR23b), -OC(0)R24, -NR23aC(0)R24, and -NR23aC(0)N(R23aR23b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R25 group;

each of the R23a and R23b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C2 to C9) heteroaryl, and (C6 to C 0) aryl;

each R24 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

each R25 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR26aR26b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R27, -C(0)NR26aR26b, -S(0)mR27, -S(0)mNR26aR26b, - NR26aS(0)mR27, -(CH2)nC(0)OR27, -(CH2)nC(0)N(R26aR26b), -(CH2)nN(R26aR26b), -OC(0)R27, - NR26aC(0)R27, and -NR26aC(0)N(R26aR26b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R27 group;

each of the R26a and R26b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl,

or R26a and R26b may be taken together with the nitrogen atom to which they are attached to form a (C2 to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S;

each R27 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

h is 1 or 2;

i is 2, 3, 4, 5, or 6;

j is 0, 1 , 2, 3, 4, or 5;

k is 1 , 2, 3, 4, or 5;

m is 0, 1 or 2;

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

Z is selected from the group consisting of -0-, -S-, -S(O)-, and -S(0)2-;

with the proviso that

when R1 , R2, or R3 is alkyl or hydrogen, X is C-C(R6aR6b)rD, Y is CH2, and D is

then NR R cannot be an optionally substituted piperazine or 1 ,4-diazepane,

and with the proviso that

when X is CR5, Y is CR4-D, D is -NR7aR7b, and R7a and R7b are taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, the said (C2to C 0) membered cycloheteroalkyi ring cannot be selected from the group consisting of

The method of claim 1 , wherein the infection is associated with filovirus selected from the group consisting of Ebolavirus and Marburgvirus.

The method of claim 2, where the filovirus is Ebolavirus.

The method of claim 3, including administrating a therapeutic amount of a therapeutic agent selected from the group consisting of Ribavirin, viral RNA-dependent-RNA-polymerase inhibitors, Favipiravir, Triazavirin, GS-5734, small interfering RNAs (siRNAs) and microRNAs, vaccines, immunomodulators.

The method of claim 4, including the inhibition of Ebolavirus glycoprotein.

The method of claim 1 , wherein

7aR7b is selected from the group consisting of

The method of claim 6, wherein:

X is C-A-D and Y is CR4R5.

The method of claim 7, wherein:

A is -C(R6aR6bV;

The method of claim 8, wherein:

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b),

and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, and (Csto C10) cycloalkenyl, wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Cs to Cio) cycloalkenyl is optionally substituted with at least one R 3 group;

R4 is hydrogen;

R5 is hydrogen.

The method of claim 9, wherein:

R3 is hydrogen.

The method of claim 7, wherein:

A is -C(R6aR6bV;

The method of claim 1 1 , wherein:

R is selected from (C6to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

The method of claim 7, wherein:

A is -C(R6aR6bV;

R7a

I

D is 7b .

The method of claim 13, wherein:

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

15. The method of claim 7, wherein:

A is -C(R R

D is selected from the group consisting of

R8

O. .0 O 7ac7b

I I S NR7aR b ^NR7aR7b l l ' T O

16. The method of claim 15, wherein:

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

17. The method of claim 1 , wherein:

A is -C(R6aR6b)j-;

wherein R a is selected from the group consisting of

169

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

The method of claim 6, wherein:

X is C-A-D and Y is a bond.

The method of claim 18, wherein:

A is -C(R6aR6bV;

D is selected from the group consisting of

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group.

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen.

20. The method of claim 6, wherein:

X is CR5 and Y is CR4-A-D. The method of claim 20, wherein:

A is -C(R6aR6bV;

D is selected from the group consisting of

R is selected from (C6to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (Cito C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen,

R4 is hydrogen;

R5 is hydrogen.

The method of claim 20, wherein:

A is -C(R6aR6b)r;

D is selected from the group consisting of

R is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to

C10) cycloalkyi, (Csto C10) cycloalkenyl, (Cito C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to

C10) cycloalkyi, (Csto C10) cycloalkenyl, (Cito C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to

C10) cycloalkyi, and (Csto C10) cycloalkenyl, wherein

each of the said (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl is optionally substituted with at least one R 3 group.

R4 is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R3 group; R5 is hydrogen.

The method of claim 22, wherein:

R1 is hydrogen;

R2 is hydrogen;

R3 is hydrogen.

The method of claim 1 , wherein:

Y is CR4R5;

X is C-A-D, wherein

A is -C(R6aR6b)r, (Ce to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (Ceto C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R6a group;

D is selected from the group consisting of

The method of claim 24, wherein:

0 is selected from the group consisting of

R9b , and R9b

26. The method of claim 1 , wherein:

Y is a bond;

X is C-A-D, wherein

A is -C(R6aR6b)r, (Ce to C10) arylene, or (C2 to Cg) heteroarylene, wherein

each of the said (Ceto C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R6a group;

D is selected from the group consisting of

R8 O

%10b i - -R10b _ ¾^R10b i and Z R10b 27. The method of claim 26, wherein

R 0a is selected from the group consisting of

R9b , and R . R9b

28. A method of treating infections associated with viruses of the Filoviridae enveloped virus, or any virus expressing filovirus glycoproteins to mediate cell entry comprising administration of a therapeutically effective amount of a compound or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, selected from the group consisting of:

29. The method of claim 28, wherein the compound is selected from the group consisting of:

30. The method of claim 28, wherein the infection is associated with filovirus selected from the group consisting of Ebolavirus and Marburgvirus.

31 . The method of claim 30, wherein the filovirus is Ebolavirus.

32. The method of claim 31 , including administrating a therapeutic amount of a therapeutic agent selected from the group consisting of Ribavirin, viral RNA-dependent-RNA-polymerase inhibitors, Favipiravir, Triazavirin, GS-5734, small interfering RNAs (siRNAs) and microRNAs, vaccines, and immunomodulators.

33. The method of claim 29, wherein the infection is associated with filovirus selected from the group consisting of Ebolavirus and Marburgvirus.

34. The method of claim 33, wherein the filovirus is Ebolavirus.

35. The method of claim 34, including administrating a therapeutic amount of a therapeutic agent selected from the group consisting of Ribavirin, viral RNA-dependent-RNA-polymerase inhibitors, Favipiravir, Triazavirin, GS-5734, small interfering RNAs (siRNAs) and microRNAs, vaccines, and immunomodulators.

36. A compound represented by Structural Formula I

I

or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D, and Y is a bond or CR4R5; or

X is CR5 and Y is CR4-A-D;

A is -C(R6aR6b)j-;

D is selected from the group consisting of

wherein NR aR is selected from the group consisting of 179

and wherein R is selected from the group consisting of

R is selected from (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio), (Csto Cio) cycloalkyi, (Cs to Cio) cydoalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2 to C9) heteroaryl, wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cydoalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group

e proviso that R is not selected from the group consisting of

R2 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cydoalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cydoalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R3 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cydoalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, -

C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cydoalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group; R4 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and -

NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R5 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R6a and R6bis independently selected from hydrogen, halogen, OH, nitro, CF3, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to Co) heteroaryl, -C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, - (CH2)nC(0)OR12, -(CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, - NR aC(0)R12, and -NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each R8 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group;

each of the R9a, R9b, and R9cis independently selected from hydrogen, halogen, OH, nitro, CF3, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to Co) heteroaryl, -C(0)R12 -C(0)NR aR b,-S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, - (CH2)nC(0)OR12, -(CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and -NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R aand R b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group,

or R a and R b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 3 group; each of the R 2 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6 to C 0) aryl, and (C2to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 3group;

each R 3 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 4aR 4b, oxo,

(Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R 5,-C(0)NR 4aR 4b, -S(0)mR15, -S(0)mNR 4aR 4b, -NR 4aS(0)mR15, - (CH2)nC(0)OR15, -(CH2)nC(0)N(R 4aR 4b), -(CH2)nN(R 4aR 4b), -OC(0)R15, -0(CH2)nO-, -

NR 4aC(0)R15, and -NR 4aC(0)N(R 4aR 4b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3to C10) cycloalkylene, and (C2to C 0) cycloheteroalkylene is optionally substituted with at least one R 6 group;

each of the R 4a and R 4b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 6 group,

or R 4a and R 4b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 6 group; each R 5 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to Cio) alkenyl, (Ci to

Cio) alkynyl, (C3 to C 0) cycloalkyi, (Csto Cio) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C3 to C 0) cycloalkyi, (Csto Cio) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 6 group;

each R 6 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 7aR 7b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R18, -C(0)NR 7aR 7b, -S(0)mR18, -S(0)mNR 7aR 7b, -NR 7aS(0)mR18,

-(CH2)nC(0)OR18, -(CH2)nC(0)N(R 7aR 7b), -(CH2)nN(R 7aR 7b), -OC(0)R18, -NR 7aC(0)R18, and -NR 7aC(0)N(R 7aR 7b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C2to C9) heteroaryl, (C6 to C 0) aryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2to C10) cycloheteroalkylene is optionally substituted with at least one R 9 group;

each of the R 7aand R 7b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^o C 0) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 9 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 9 group; each R 8 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi,

(Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C2 to C9) heteroaryl, and (C6 to C 0) aryl is optionally substituted with at least one R 9 group;

each R 9 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR20aR20b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R21 , -C(O)NR20aR20b, -S(0)mR21 , -S(O)mNR20aR20b, - NR20aS(O)mR21 , -(CH2)nC(0)OR21 , -(CH2)nC(O)N(R20aR20b), -(CH2)nN(R20aR20b), -OC(0)R21 , - NR20aC(O)R21 , and -NR20aC(O)N(R20aR20b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to Cg) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R22 group;

each of the R20a and R20b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C 0) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R22 group,

or R20a and R20b may be taken together with the nitrogen atom to which they are attached to form a (C2 to C 0) cycloheteroalkyi ring, wherein

said (C2 to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2 to C 0) cycloheteroalkyi ring is optionally substituted with at least one R22 group; each R2 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl,

(Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

each R22 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR23aR23b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R24,-C(0)NR23aR23b, -S(0)mR24, -S(0)mNR23aR23b, -NR23aS(0)mR24, - -(CH2)nC(0)OR24, -(CH2)nC(0)N(R23aR23b), -(CH2)nN(R23aR23b), -OC(0)R24, -NR23aC(0)R24, and -NR23aC(0)N(R23aR23b), wherein each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R25 group;

each of the R23a and R23b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to

C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C2 to C9) heteroaryl, and (C6 to C 0) aryl;

each R24 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

each R25 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR26aR26b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R27, -C(0)NR26aR26b, -S(0)mR27, -S(0)mNR26aR26b, -NR26aS(0)mR27,

-(CH2)nC(0)OR27, -(CH2)nC(0)N(R26aR26b), -(CH2)nN(R26aR26b), -OC(0)R27, -NR26aC(0)R27, and - NR26aC(0)N(R26aR26b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and

(C2 to C10) cycloheteroalkylene is optionally substituted with at least one R27 group;

each of the R26a and R26b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl,

or R26a and R26b may be taken together with the nitrogen atom to which they are attached to form a (C2 to C 0) cycloheteroalkyi ring, wherein

said (C2 to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S;

each R27 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

h is 1 or 2;

i is 2, 3, 4, 5, or 6;

j is 0, 1 , 2, 3, 4, or 5;

k is 1 , 2, 3, 4, or 5;

m is 0, 1 or 2;

n is 0, 1 , 2, 3, or 4; with the proviso that when R is phenyl, R2 is hydrogen, R3 is hydrogen, X is C-A-D, Y is CH2,

R8 R11a

A is a bond, and D is ^ NR7aR7b , then NR7aR7b is not R9a R9b

and with the proviso that when R is selected from the group consisting of 4-methylphenyl, 3,4-dimethylphenyl, and 4-methoxyphenyl, R2 is hydrogen, R3 is hydrogen, X is C-A-D, Y is CH2,

O

A is a bond, then D is not selected from the group consisting of ^ NR7aR7b ,

and with the proviso that when R is 4-chlorophenyl, R2 is hydrogen, R3 is hydrogen, X is C-A- D, Y is CH2, A is a bond, and D is selected from the group consisting of

'!\^NR7aR7b , and > ^NR7aR7b

NR aR is not selected from the group consisting of

and with the proviso that the following compounds shall be excluded :

189

37. The compound of claim 36, wherein:

X is C-A-D and Y is CR4R5.

38. The compound of claim 37, wherein:

A is -C(R6aR6bV;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R4 is hydrogen;

R5 is hydrogen.

39. The compound of claim 38, wherein:

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group.

40. The compound of claim 39, wherein:

R3 is hydrogen. The compound of claim 40, wherein:

A is -C(R6aR6bV;

O 9a p9b ^N¾NR7Wb

D iS R8 O ;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen ;

R4 is hydrogen;

R5 is hydrogen.

The compound of claim 37, wherein:

A is -C(R6aR6b)j-;

R7a

I

D is

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen ;

R4 is hydrogen;

R5 is hydrogen.

The compound of claim 37, wherein:

A is -C(R6aR6bV;

D is selected from the group consisting of

The compound of claim 43, wherein:

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

45. The compound of claim 37, wherein:

A is -C(R R )j-;

D is selected from the group consisting of

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12 wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

46. The compound of claim 36, wherein:

X is C-A-D and Y is a bond.

47. The compound of claim 46, wherein:

A is -C(R R )j-;

D is selected from the group consisting of

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group.

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to Cio) cycloalkyi, (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (C5 to Cio) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen.

48. The compound of claim 36, wherein:

X is CR5 and Y is CR4-A-D.

49. The compound of claim 48, wherein:

A is -C(R6aR6bV;

D is selected from the group consisting of

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R3 is hydrogen;

R4 is hydrogen;

R5 is hydrogen.

The compound of claim 48, wherein:

A is -C(R6aR6bV;

D is selected from the group consisting of

R is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH2)nC(0)N(R aR b), and -C(0)R12, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group; R3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R4 is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R5 is hydrogen.

51 . The compound of claim 50, wherein:

R is hydrogen;

R2 is hydrogen;

R3 is hydrogen.

52. A compound represented by Structural Formula I

I

or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D, and Y is a bond or CR4R5; or

X is CR5 and Y is CR4-A-D;

A is -C(R6aR6b)r, (C6 to C10) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6 to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R6a group;

D is selected from the group consisting of

' , wherein

R 0b is selected from the group consisting of

, and

R is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to Cio) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R 2,-C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nCO)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R2 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R3 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R4 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, - C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R5 is selected from hydrogen, halogen, OH, nitro, CFs, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, -

C(0)R12 -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -(CH2)nC(0)OR12, - (CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and - NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R6a and R6bis independently selected from hydrogen, halogen, OH, nitro, CF3, -NR aR b, (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, -C(0)R12, -C(0)NR aR b, -S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, -

(CH2)nC(0)OR12, -(CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and -NR aC(0)N(R aR b), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each R8 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group;

each of the R9a, R9b, and R9cis independently selected from hydrogen, halogen, OH, nitro, CF3, -NR aR b, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, -C(0)R12 -C(0)NR aR b,-S(0)mR12, -S(0)mNR aR b, -NR aS(0)mR12, - (CH2)nC(0)OR12, -(CH2)nC(0)N(R aR b), -(CH2)nN(R aR b), -OC(0)R12, -NR aC(0)R12, and -NR aC(0)N(R aR b), wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R aand R b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 3 group,

or R a and R b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 3 group; each of the R 2 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6 to C 0) aryl, and (C2to C9) heteroaryl, wherein

each of the said (^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi,

(Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 3group;

each R 3 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 4aR 4b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R 5,-C(0)NR 4aR 4b, -S(0)mR15, -S(0)mNR 4aR 4b, -NR 4aS(0)mR15, - (CH2)nC(0)OR15, -(CH2)nC(0)N(R 4aR 4b), -(CH2)nN(R 4aR 4b), -OC(0)R15, -0(CH2)nO-, - NR 4aC(0)R15, and -NR 4aC(0)N(R 4aR 4b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3to C10) cycloalkylene, and (C2to C 0) cycloheteroalkylene is optionally substituted with at least one R 6 group;

each of the R 4a and R 4b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2to Cg) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 6 group,

or R 4a and R 4b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 6 group; each R 5 is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C3 to C 0) cycloalkyi, (Csto Cio) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C3 to C 0) cycloalkyi, (Csto Cio) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R 6 group;

each R 6 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 7aR 7b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R18, -C(0)NR 7aR 7b, -S(0)mR18, -S(0)mNR 7aR 7b, -NR 7aS(0)mR18, -(CH2)nC(0)OR18, -(CH2)nC(0)N(R 7aR 7b), -(CH2)nN(R 7aR 7b), -OC(0)R18, -NR 7aC(0)R18, and -NR 7aC(0)N(R 7aR 7b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C2to C9) heteroaryl, (C6 to C 0) aryl, (C6to C 0) arylene, (C2to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2to C10) cycloheteroalkylene is optionally substituted with at least one R 9 group;

each of the R 7aand R 7b is independently selected from hydrogen, (C^ to C 0) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene, wherein

each of the said (C^o C 0) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C 0) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C 0) cycloheteroalkyi, (C6to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, and (C2 to C9) heteroarylene is optionally substituted with at least one R 9 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C2to C 0) cycloheteroalkyi ring, wherein

said (C2to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2to C 0) cycloheteroalkyi ring is optionally substituted with at least one R 9 group; each R 8 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to Cio) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C 0) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C2 to C9) heteroaryl, and (C6 to C 0) aryl is optionally substituted with at least one R 9 group;

each R 9 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR20aR20b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R21 , -C(O)NR20aR20b, -S(0)mR21 , -S(O)mNR20aR20b, - NR20aS(O)mR21 , -(CH2)nC(0)OR21 , -(CH2)nC(O)N(R20aR20b), -(CH2)nN(R20aR20b), -OC(0)R21 , - NR20aC(O)R21 , and -NR20aC(O)N(R20aR20b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R22 group;

each of the R20a and R20b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl, wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C 0) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl is optionally substituted with at least one R22 group,

or R20a and R20b may be taken together with the nitrogen atom to which they are attached to form a (C2 to C 0) cycloheteroalkyi ring, wherein

said (C2 to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C2 to C 0) cycloheteroalkyi ring is optionally substituted with at least one R22 group; each R2 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

each R22 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR23aR23b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R24,-C(0)NR23aR23b, -S(0)mR24, -S(0)mNR23aR23b, -NR23aS(0)mR24, - -(CH2)nC(0)OR24, -(CH2)nC(0)N(R23aR23b), -(CH2)nN(R23aR23b), -OC(0)R24, -NR23aC(0)R24, and -NR23aC(0)N(R23aR23b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to Cg) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R25 group;

each of the R23a and R23b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C2 to C9) heteroaryl, and (C6 to C 0) aryl;

each R24 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

each R25 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR26aR26b, oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to

C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R27, -C(0)NR26aR26b, -S(0)mR27, -S(0)mNR26aR26b, -NR26aS(0)mR27, -(CH2)nC(0)OR27, -(CH2)nC(0)N(R26aR26b), -(CH2)nN(R26aR26b), -OC(0)R27, -NR26aC(0)R27, and - NR26aC(0)N(R26aR26b), wherein

each of the said (C^ to C 0) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, (C2 to Cg) heteroaryl, (C6 to C 0) arylene, (C2 to C9) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R27 group;

each of the R26a and R26b is independently selected from hydrogen, (C^ to C 0) alkyl, (Ci to

C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C 0) cycloheteroalkyi, (C6 to C 0) aryl, and (C2 to C9) heteroaryl,

or R26a and R26b may be taken together with the nitrogen atom to which they are attached to form a (C2 to C 0) cycloheteroalkyi ring, wherein

said (C2 to C 0) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S;

each R27 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

h is 1 or 2;

i is 2, 3, 4, 5, or 6;

j is 0, 1 , 2, 3, 4, or 5;

k is 1 , 2, 3, 4, or 5;

m is 0, 1 or 2;

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

with the proviso that when X is C-A-D, and Y is CH2,

then R is not selected from the group consisting of hydrogen and methyl,

and with the proviso that the following compounds shall be excluded:

53. The compound of claim 52, wherein:

A is -C(R6aR6b)r, (C6 to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6 to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R6a group;

D is of C b.

54. A compound selected from the group consisting of:

55. The compound of claim 54 selected from the group consisting of

Description:
PATENT APPLICATION

ADAMANTANE DERIVATIVES FOR THE TREATMENT OF FILOVIRUS INFECTION

ADAMANTANE DERIVATIVES FOR THE TREATMENT OF FILOVIRUS

INFECTION

CROSS REFERENCES TO RELATED APPLICATIONS

This patent application is a continuation in part of and claims the benefit of priority to United States Provisional Patent Applications serial numbers 62/279,917, filed January 18, 2016 and 62/351 ,839, filed June 17, 2016, both applications are herein incorporated by reference for all purposes.

GOVERNMENT SUPPORT

This invention was made with government support under 1 R43AI1 18207-01 (Ken McCormack) awarded by U.S. National Institutes of Health. The government has certain rights in the invention.

REFERENCE TO A "SEQUENCE LISTING, A TABLE, OR A COMPUTER PROGRAM

LISTING APPENDIX SUBMITTED ON A COMPACT DISK

NOT APPLICABLE

FIELD OF THE INVENTION

The present invention relates to methods of inhibiting infection by viruses of the Filoviridae family (filoviruses) in humans, other mammals, or in cell culture, to treating infection by filoviruses, to methods of inhibiting the replication of filoviruses, to methods of reducing the amount of filoviruses, and to compositions that can be employed for such methods. These methods, applications, and compositions apply not only to Filoviridae viruses but also to any virus, whether naturally emerging or engineered, whose cell entry properties are determined by filovirus glycoproteins.

BACKGROUND OF THE INVENTION

The invention relates to the use of compounds for the treatment and/or prophylaxis of infection of humans or other mammals by one or more of a number of enveloped viruses of the Filoviridae family (filoviruses) or any other native or engineered enveloped virus utilizing filovirus glycoproteins to mediate cell entry. Enveloped viruses are comprised of an outer host-derived lipid membrane and an inner nucleoprotein core, which contains the viral genetic material (whether RNA or DNA). Virus-cell fusion is the means by which all enveloped viruses enter cells and initiate disease-causing cycles of replication. In all cases virus-cell fusion is executed by one or more viral surface glycoproteins that are anchored within the lipid membrane envelope. One or more glycoproteins from a given virus may form a glycoprotein complex that interacts with a number of different surface and/or intracellular receptors of infected host cells to initiate the association between virus and host cell. However, one glycoprotein is generally denoted as the protein primarily driving the fusion of viral and host cell membranes. At least three distinct classes of viral membrane fusion proteins have been determined (classes I, II , and III) [Weissenhorn, W.; Carfi, A.; Lee, K.H.; Skehel, J.J., and Wiley, D. C. Crystal structure of the Ebola virus membrane fusion subunit, GP2, from the envelope glycoprotein ectodomain. Mol. Cell (1998) 2:605-616; White, J.M.; Delos, S.E.; Brecher, M.; Schornberg K. Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit. Rev. Biochem. Mol. Biol. (2008) 43:189-219; Igonet, S.; Vaney, M.C.; Vonrhein, C; Bricogne, G.; Stura, E.A.;Hengartner H.; Eschli, B.; Rey, F.A. X-ray structure of the arenavirus glycoprotein GP2 in its postfusion hairpin conformation, Proc. Natl. Acad. Sci. (201 1) 108:19967-19972]. Class I fusion proteins are found in viruses from the Orthomyxoviridae, Retroviridae, Paramyxoviridae, Coronaviridae, Filoviridae, and Arenaviridae familes, Class II proteins from Togaviridae, Flaviviridae, and Bunyaviridae while Class III or other types are from Rhadboviridae, Herpesviridae, Poxviridae, and Hepadnaviridae. Given that viral cell entry is an essential step in the viral replication process the identification of compounds that inhibit virus cell entry could provide attractive antivirals for viruses that are pathogenic to humans and/or other mammals. Chemical compounds that act as inhibitors of one enveloped virus may also act as inhibitors of other enveloped viruses. However, while enveloped

Table 1 Family and Genera of Envelope Viruses and Glycoprotein Classification

viruses share some common functional and structural features with regard to glycoprotein-dependent cell entry and fusion the specific host targets and mechanisms of cell entry differ among enveloped viruses: between and even within different virus families as a function of their unique glycoprotein (GP) sequences and structures, and the cellular host proteins that they interact with [White, J.M.; Delos, S.E.; Brecher, M., Schornberg K. Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit. Rev. Biochem. Mol. Biol. (2008) 43:189-219]. The invention described herein relates to the use of compounds for the treatment and/or prophylaxis of infection as mediated by the cell entry and fusion process of filovirus glycoproteins whether native or engineered.

One viral expression system that may be utilized to identify inhibitors of enveloped viruses based on their glycoprotein sequences and functional properties is the vesicular stomatitis virus (VSV) system. This approach uses VSV, a virus in the Rhadboviridae family (expressing Class III fusion proteins), lacking a native VSV glycoprotein. "Pseudotyped" viruses that are infective and functionally replicative in cell culture can be generated by substituting the VSV glycoprotein with a glycoprotein originating from other enveloped viruses. The cell entry properties and functions of these pseudotyped viruses are determined by the viral glycoprotein that has been introduced . The cell entry and infectivity properites of pseudotyped VSV viruses have been shown to be determined by the introduced glycoprotein from a host of envelope viruses including Ebola, Lassa, Hanta, Hepatitis B, and other viruses [Ogino, M., et al. Use of vesicular stomatitis virus pseudotypes bearing hantaan or seoul virus envelope proteins in a rapid and safe neutralization test. Clin. Diagn. Lab. immunol.(2003) 10(1):154-60; Saba, M.N., et al., Formation of vesicular stomatitis virus pseudotypes bearing surface proteins of hepatitis B virus. J. Virol. (2005) 79(19):12566-74; Takada, A., et al., A system for functional analysis of Ebola virus glycoprotein, Proc. Natl. Acad. Sci. (1997) 94:14764-69; Garbutt, ., et al., Properties of replication-competent vesicular stomatitis virus vectors expressing glycoproteins of filoviruses and arenaviruses. J. Viroi. (2004) 78(10):5458-65]. When the pseudotype virion also expresses a reporter gene such as green fluorescent protein (GFP) or Renilla luciferase, virion infectivity and replication may be monitored using high-throughput optical methods in cultured mammalian cell lines, including Vera and HEK-293 cells [Cote, M.; Misasi, J.; Ren, T.; Bruchez, A., Lee, K., Filone, C. M.; Hensley, L.; Li, Q.; Ory, D.; Chandran, K.; Cunningham, J., Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection, Nature (201 1) 477: 344-348]. While VSV does not infect humans and may not be a virus of particular interest for the development of therapeutic antivirals, VSV pseudotyped viruses expressing glycoproteins from other enveloped viruses may be used to screen chemical libraries to identify compounds that inhibit the glycoproteins, cell entry, and infectivity of enveloped viruses associated with significant human health concerns. [Cunningham, J. et al. US patent application, publication number US2013/0231332; WO

2012/031090, 8 Mar 2012; WO2013/022550, 14 Feb 2013; Warren, T. K., et al. Antiviral activity of a small-molecule inhibitor of Filovirus infection. Antimicrob. Agents Chemother. (2010) 54: 2152-2159; Yermolina, M., et al. Discovery, synthesis, and biological evaluation of a novel group of selective inhibitors of filovirus entry.J. Med. Chem. (201 1) 54: 765 - 781 ; Basu, A., et al. Identification of a small-molecule entry inhibitor for Filoviruses. J. Virol. (201 1) 85: 3106-31 19; Lee, K., et al., Inhibition of Ebola virus infection: identification of Niemann-Pick as the target by optimization of a chemical probe. ACS Med. Chem. Lett. (2013) 4: 239-243; Madrid, P. B., et al. A Systematic screen of FDA- approved drugs for inhibitors of biological threat agents Plos One (2013) 8: 1 -14; Elshabrawy, H. A., et al. Identification of a broad-spectrum antiviral amall molecule against severe scute respiratory syndrome Coronavirus and Ebola, Hendra, and Nipah Viruses by using a novel high-throughput screening assay. J. Virol. (2014) 88: 4353-4365].

Filovirus infections are associated with hemorrhagic fevers, the clinical manifestations of which may be severe and/or fatal. As described herein, for the current invention, VSV pseudotyped viruses expressing filovirus glycoproteins can be generated and screened with a collection of chemical compounds to identify those compounds that inhibit infectivity. The identification of inhibitors of filovirus glycoprotein-mediated virus cell entry may may be utilized to treat infections of filoviruses to provide effective therapeutic regimens for the prophylaxis and/or treatment of filoviruses or any newly emerging virus, whether native or engineered, whose cell entry properties may be determined by filovirus glycoproteins. The Filoviridae virus family is comprised of at least three genera: Ebolavirus, which currently includes five species Zaire (EBOV), Sudan (SUDV), Bundibygo (BDBV), Tai Forest (TAFV) and Reston (RESTV), Marburgvirus, which currently includes two species Marburg (MARV) and Ravn (RAVV), and Cuervavirus, which currently includes a single species LLovia virus (LLOV). RAVV and LLOV are examples of filoviruses that have been identified only recently and a number of additional new species and genera may continue to emerge.

Ifidae: currentl identified filovirus genera, species, and nomencl

Glycoproteins from Filoviridae family members can be expressed in pseudotyped viruses (e.g.

VSV pseudotype) to identify compounds that inhibit filovirus infection. Based on the structural similarities and/or differences between the viral glycoprotein target and/or host cell targets, the inhibitor compounds may act on only a single filovirus glycoprotein or on a broad spectrum of filoviruses. Furthermore, given the basic functional and structural similarities of glycoproteins among different families of enveloped viruses it is proable that a given compound class may act across a broad range of enveloped viruses.

Alignments of representative filovirus glycoprotein sequences were generated to illustrate the amino acid homology among different filovirus species.

Table 3: Homology of filovirus glycoproteins - created by Clustal2.1

Species/Genbank ID Zaire Bundi T Forest Reston Sudan Marburg

Zaire/AAB81004 100 68.0 66.5 59.9 56.8 32.7

Bundibugyo/AGL73453 68.0 100 73.6 60.4 57.7 33.0

Tai Forest/YP_003815426 66.5 73.6 100 59.5 57.7 33.9

Reston/BAB69006 59.9 60.4 59.5 100 61 .4 32.7

Sudan/AAB37096 56.8 57.7 57.7 61 .4 100 33.0

Marburg/AAC40460 32.7 33.0 33.9 32.7 33.0 100 A matrix comparison of the amino acid homology (homology is defined as the number of identities between any two sequences, divided by the length of the alignment, and represented as a percentage) as determined from the Clustal2.1 program (http://www.ebi. ac.uk/Tools/msa/clustalo/) among and between distinct filovirus genus and species is illustrated in Table 3. Glycoproteins among virus species within the same filovirus genus (e.g., Ebolavirus) are more homologous to each other than to those in another genus (Marburgvirus). However, currently available filovirus glycoproteins exhibit significant homology (>30% identity from any one member to another). Given this homology for some chemical series it is possible to identify compounds that exhibit activity against a broad- spectrum of filoviruses.

Similar alignments were subsequently carried out with a number of class I glycoproteins from other enveloped virus families. Each of the glycoproteins from the other enveloped viruses exhibit <20% identity with any of the filovirus glycoproteins. Although there are similarities in functional and structural characteristics among the class I glycoproteins, there are clear distinctions including dependence on low pH, receptor binding, location of the fusion peptide [White, J.M.; Delos, S.E.,; Brecher, M.; Schornberg, K. Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit. Rev. Biochem. Mol., Biol. (2008) 43:189-219] and given the low amino acid sequence homology across class I virus families it becomes unlikely that a given chemical series that inhibits filovirus cell entry/fusion would also exhibit similar inhibitory activities with other envelope class I glycoprotein virus families.

Table 4: Homology matrix between filoviruses and other class I glycoprotein viruses-created by Clustal2.1

Abbreviations: M: Marburg, Z: Zaire, T: Tai Forest, B: Bundibugyo, S: Sudan, R: Reston, INF:

Influenza, LASV: Lassa virus, JUNV: Junin virus; Genbank ID in bold BRIEF SUMMARY OF THE INVENTION

The present invention relates to methods of inhibiting filoviruses (or any virus whose cell entry is mediated by filovirus glycoproteins) infection in humans, other mammals, or in cell culture, to treating filovirus infection, to methods of inhibiting the replication of filoviruses, to methods of reducing the amount of filoviruses, and to compositions that can be employed for such methods. These methods, applications, and compositions apply not only to Filoviridae viruses but also to any virus, whether naturally emerging or engineered, whose cell entry properties are determined by filovirus glycoproteins.

In one embodiment, the method comprises administering to humans, other mammals, cell culture, or biological sample an effective amount of a compound represented by Structural Formula I for treatment of filovirus infection

I

or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D, and Y is a bond or CR 4 R 5 ; or

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) r , (C6 to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6 to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R 6a group;

D is selected from the group consisting of

R is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to Cio) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nCO)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to Co) cycloheteroalkyi, (Ceto C10) aryl, (C2 to Co) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 5 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , -

(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 6a and R 6b is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR a R b , (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 -C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , -(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group; each of the R and R is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, NR a R b , (Csto C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) membered cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) membered cycloheteroalkyi ring is optionally substituted with at least one R 3 group;

each R 8 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to

Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 )

heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group;

each of the R 9a , R 9b , and R 9c is independently selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to Cio) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to Co) heteroaryl, -C(0)R 12 -C(0)NR a R b ,-S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , - (CH 2 )nC(0)OR 12 , -(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and - NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 0a and R 0b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 )

cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi,

(Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 3 group;

each of the R a and R b is independently selected from hydrogen, (Ci to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group,

or R a and R b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 3 group; each of the R 2 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi,

(Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 3 group;

each R 3 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 4a R 4b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10)

cycloheteroalkylene, -C(0)R 15 , -C(0)NR 4a R 4b , -S(0)mR 15 , -S(0)mNR 4a R 4b , -NR 4a S(0)mR 15 , - (CH 2 )nC(0)OR 15 , -(CH 2 )nC(0)N(R 4a R 4b ), -(CH 2 )nN(R 4a R 4b ), -OC(0)R 15 , -0(CH 2 ) n O-, - NR 4a C(0)R 15 , and -NR 4a C(0)N(R 4a R 4b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3to C10) cycloalkylene, and (C 2 to C 0 ) cycloheteroalkylene is optionally substituted with at least one R 6 group;

each of the R 4a and R 4b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 6 group,

or R 4a and R 4b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 6 group; each R 5 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi,

(Csto Cio) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 6 group;

each R 6 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 7a R 7b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10)

cycloheteroalkylene, -C(0)R 18 , -C(0)NR 7a R 7b , -S(0)mR 18 , -S(0)mNR 7a R 7b , -NR 7a S(0)mR 18 , -(CH 2 )nC(0)OR 18 , -(CH 2 )nC(0)N(R 7a R 7b ), -(CH 2 )nN(R 7a R 7b ), -OC(0)R 18 , -NR 7a C(0)R 18 , and -NR 7a C(0)N(R 7a R 7b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) aryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2to C10) cycloheteroalkylene is optionally substituted with at least one R 9 group;

each of the R 7a and R 7b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^o C 0 ) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C 3 to C 0 ) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 9 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 9 group; each R 8 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, and (C2 to C9) heteroaryl, wherein each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C 3 to C 0 ) cycloalkyi,

(Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, and (C 6 to C 0 ) aryl is optionally substituted with at least one R 9 group;

each R 9 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 20a R 20b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10)

cycloheteroalkylene, -C(0)R 21 , -C(O)NR 20a R 20b , -S(0)mR 21 , -S(O)mNR 20a R 20b , -NR 20a S(O)mR 21 ,

-(CH 2 )nC(0)OR 21 , -(CH 2 )nC(O)N(R 20a R 20b ), -(CH 2 )nN(R 20a R 20b ), -OC(0)R 21 , -NR 20a C(O)R 21 , and -NR 20a C(O)N(R 20a R 20b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C 2 to C10) cycloheteroalkylene is optionally substituted with at least one R 22 group;

each of the R 20a and R 20b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C 3 to C 0 ) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 22 group,

or R 20a and R 20b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 22 group; each R 2 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl,

(Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C 2 to C9) heteroaryl;

each R 22 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR 23a R 23b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C 2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C 2 to C10)

cycloheteroalkylene, -C(0)R 24 ,-C(0)NR 23a R 23b , -S(0)mR 24 , -S(0)mNR 23a R 23b , -NR 23a S(0)mR 24 , - -(CH 2 )nC(0)OR 24 , -(CH 2 )nC(0)N(R 23a R 23b ), -(CH 2 )nN(R 23a R 23b ), -OC(0)R 24 , -NR 23a C(0)R 24 , and -NR 23a C(0)N(R 23a R 23b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C 2 to C10) cycloheteroalkylene is optionally substituted with at least one R 25 group;

each of the R 23a and R 23b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 )

cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, and (C 6 to C 0 ) aryl;

each R 24 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C 2 to C9) heteroaryl; each R is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR R , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene,(C2 to C10) cycloheteroalkylene, -C(0)R 27 , -C(0)NR 26a R 26b , -S(0)mR 27 , -S(0)mNR 26a R 26b , -NR 26a S(0)mR 27 , -(CH 2 )nC(0)OR 27 ,

-(CH 2 )nC(0)N(R 26a R 26b ), -(CH 2 )nN(R 26a R 26b ), -OC(0)R 27 , -NR 26a C(0)R 27 , and -NR 26a C(0)N(R 26a R 26b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 27 group;

each of the R 26a and R 26b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl,

or R 26a and R 26b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S;

each R 27 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

h is 1 or 2;

i is 2, 3, 4, 5, or 6;

j is 0, 1 , 2, 3, 4, or 5;

k is 1 , 2, 3, 4, or 5;

m is 0, 1 or 2;

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

Z is selected from the group consisting of -0-, -S-, -S(O)-, and -S(0) 2 -;

with the proviso that

when R 1 R 2 or R 3 is alk l or h dro en X is C-C R 6a R 6b D Y is CH and D is then NR R cannot be an optionally substituted piperazine or 1 ,4-diazepane,

and with the proviso that

when X is CR 5 , Y is CR 4 -D, D is -NR 7a R 7b , and R 7a and R 7b are taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, the said (C 2 to C 0 ) membered cycloheteroalkyi ring cannot be selected from the group consisting of

DESCRIPTION OF THE DRAWINGS Figure 1 : Depicts results of a pharmacokinetics (PK) study of compound A78 in male CD-1 mice. Shown are plasma concentrations of A78 after intravenous (iv) administration of 1 mg/kg or oral (po) administration at 10 mg/kg dose over a 24 h period.

DETAILED DESCRIPTION OF THE INVENTION

The present invention relates to methods of inhibiting filoviruses (or any virus whose cell entry is mediated by filovirus glycoproteins) infection in humans, other mammals, or in cell culture, to treating filovirus infection, to methods of inhibiting the replication of filoviruses, to methods of reducing the amount of filoviruses, and to compositions that can be employed for such methods. These methods, applications, and compositions apply not only to Filoviridae viruses but also to any virus, whether naturally emerging or engineered, whose cell entry properties are determined by filovirus glycoproteins.

In one embodiment, the method comprises administering to humans, other mammals, cell culture, or biological sample an effective amount of a compound represented by Structural Formula I for treatment of filovirus infection

I

or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D, and Y is a bond or CR 4 R 5 ; or

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) r , (C6to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R 6a group; D is selected from the group consisting of

R is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to Cio) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nCO)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group; R 2 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 5 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 6a and R 6b is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR a R b , (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 , -C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , -(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 7a and R 7b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, NR a R b , (Csto C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (Ci to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) membered cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) membered cycloheteroalkyi ring is optionally substituted with at least one R 3 group;

each R 8 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to

C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 )

heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group;

each of the R 9a , R 9b , and R 9c is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to Co) heteroaryl, -C(0)R 12 -C(0)NR a R b ,-S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , - (CH 2 )nC(0)OR 12 , -(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and - NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 0a and R 0b is independently selected from hydrogen, (Ci to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 )

cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, wherein each of the said (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 3 group;

each of the R a and R b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (Ci to Cio) alkoxy, aryloxy, (C3to Cio) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group,

or R a and R b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 3 group; each of the R 2 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi,

(Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 3 group;

each R 3 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 4a R 4b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10)

cycloheteroalkylene, -C(0)R 15 , -C(0)NR 4a R 4b , -S(0)mR 15 , -S(0)mNR 4a R 4b , -NR 4a S(0)mR 15 , - (CH 2 )nC(0)OR 15 , -(CH 2 )nC(0)N(R 4a R 4b ), -(CH 2 )nN(R 4a R 4b ), -OC(0)R 15 , -0(CH 2 ) n O-, - NR 4a C(0)R 15 , and -NR 4a C(0)N(R 4a R 4b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3to C10) cycloalkylene, and (C 2 to C 0 ) cycloheteroalkylene is optionally substituted with at least one R 6 group;

each of the R 4a and R 4b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 6 group,

or R 4a and R 4b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 6 group; each R 5 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 6 group;

each R 6 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 7a R 7b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10)

cycloheteroalkylene, -C(0)R 18 , -C(0)NR 7a R 7b , -S(0)mR 18 , -S(0)mNR 7a R 7b , -NR 7a S(0)mR 18 , -(CH 2 )nC(0)OR 18 , -(CH 2 )nC(0)N(R 7a R 7b ), -(CH 2 )nN(R 7a R 7b ), -OC(0)R 18 , -NR 7a C(0)R 18 , and -NR 7a C(0)N(R 7a R 7b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) aryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2to C10) cycloheteroalkylene is optionally substituted with at least one R 9 group;

each of the R 7a and R 7b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to

C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^o C 0 ) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C 3 to C 0 ) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 9 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 9 group; each R 8 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, and (C2 to C9) heteroaryl, wherein each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C 3 to C 0 ) cycloalkyi, (Cs to Cio) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, and (C 6 to C 0 ) aryl is optionally substituted with at least one R 9 group;

each R 9 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR 20a R 20b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10)

cycloheteroalkylene, -C(0)R 21 , -C(O)NR 20a R 20b , -S(0)mR 21 , -S(O)mNR 20a R 20b , -NR 20a S(O)mR 21 , -(CH 2 )nC(0)OR 21 , -(CH 2 )nC(O)N(R 20a R 20b ), -(CH 2 )nN(R 20a R 20b ), -OC(0)R 21 , -NR 20a C(O)R 21 , and -NR 20a C(O)N(R 20a R 20b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 22 group;

each of the R 20a and R 20b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to

C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C 3 to C 0 ) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 22 group,

or R 20a and R 20b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 22 group; each R 2 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

each R 22 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR 23a R 23b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10)

cycloheteroalkylene, -C(0)R 24 ,-C(0)NR 23a R 23b , -S(0)mR 24 , -S(0)mNR 23a R 23b , -NR 23a S(0)mR 24 , - -(CH 2 )nC(0)OR 24 , -(CH 2 )nC(0)N(R 23a R 23b ), -(CH 2 )nN(R 23a R 23b ), -OC(0)R 24 , -NR 23a C(0)R 24 , and -NR 23a C(0)N(R 23a R 23b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 25 group; each of the R and R is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 )

cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, and (C 6 to C 0 ) aryl;

each R 24 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

each R 25 is independently selected from hydrogen, halogen , OH, nitro, CF3, -NR 26a R 26b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene,(C2 to C10) cycloheteroalkylene, -C(0)R 27 , -C(0)NR 26a R 26b , -S(0)mR 27 , -S(0)mNR 26a R 26b , -NR 26a S(0)mR 27 , -(CH 2 )nC(0)OR 27 ,

-(CH 2 )nC(0)N(R 26a R 26b ), -(CH 2 )nN(R 26a R 26b ), -OC(0)R 27 , -NR 26a C(0)R 27 , and -NR 26a C(0)N(R 26a R 26b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 27 group;

each of the R 26a and R 26b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl,

or R 26a and R 26b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S;

each R 27 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl;

h is 1 or 2;

i is 2, 3, 4, 5, or 6;

j is 0, 1 , 2, 3, 4, or 5;

k is 1 , 2, 3, 4, or 5;

m is 0, 1 or 2;

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

Z is selected from the group consisting of -0-, -S-, -S(O)-, and -S(0) 2 -;

with the proviso that

when R 1 , R 2 , or R 3 is alkyl or hydrogen, X is C-C(R 6a R 6b ) r D, Y is CH 2 , and D is then NR R cannot be an optionally substituted piperazine or 1 ,4-diazepane, and with the proviso that

when X is CR 5 , Y is CR 4 -D, D is -NR 7a R 7b , and R 7a and R 7b are taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyl ring, the said (C 2 to C 0 ) membered cycloheteroalkyl ring cannot be selected from the group consisting of

In another embodiment, the method comprises administering to humans, other mammals, cell culture, or biological sample an effective amount of a compound represented by Structural Formula I for treatment of Ebolavirus infection.

In another embodiment, the method comprises administering to humans, other mammals, cell culture, or biological sample an effective amount of a compound represented by Structural Formula I for treatment of Marburgvirus infection.

In another embodiment, the method comprises of inhibiting Ebolavirus.

In another embodiment, the method comprises of including administrating a therapeutic amount of a therapeutic agent selected from the group consisting of Ribavirin, viral RNA-dependent- RNA-polymerase inhibitors, Favipiravir, Triazavirin, GS-5734, small interfering RNAs (siRNAs) and microRNAs, vaccines, and immunomodulators.

In another embodiment, the method comprises of inhibiting of Ebolavirus glycoprotein.

In another embodiment, the invention relates to compounds of Structural Formula I,

or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 , wherein A-D is defined as before.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond, wherein A-D is defined as before.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D, wherein A-D is defined as before. In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R R and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is hydrogen; R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b ) j -;

R is selected from (Ceto C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (Ceto C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is hydrogen;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is hydrogen;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond or CR 4 R 5 ; or

X is CR 5 and Y is CR 4 -A-D;

A is -C(R R and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond or CR 4 R 5 ;

A is -C(R 6a R 6b ) r , (C6to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at ne R 6a group; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is a bond or CR 4 R 5 ;

A is (C6to C10) arylene or (C2 to C9) heteroarylene, wherein

each of the said (C6to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R 6a group; and

D is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

D is selected from the group consisting of wherein NR R is selected from the group consisting of

R is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , -

(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is selected from hydrogen, halogen, OH, nitro, CFs, -NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 ,

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (Ceto C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 5 is selected from hydrogen, halogen, OH, nitro, CF3,-NR a R b , (Ci to C10) alkyl, (Ci to C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (Ceto C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12

-C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , - (CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each of the R 6a and R 6b is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR a R b , (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, -C(0)R 12 -C(0)NR a R b , -S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , -(CH 2 )nC(0)OR 12 , -(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and -NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

each R 8 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 )

heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group;

each of the R 9a , R 9b , and R 9c is independently selected from hydrogen, halogen, OH, nitro, CF3,-NR a R b , (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Cito C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2 to Co) heteroaryl, -C(0)R 12 -C(0)NR a R b ,-S(0)mR 12 , -S(0)mNR a R b , -NR a S(0)mR 12 , - (CH 2 )nC(0)OR 12 , -(CH 2 )nC(0)N(R a R b ), -(CH 2 )nN(R a R b ), -OC(0)R 12 , -NR a C(0)R 12 , and - NR a C(0)N(R a R b ), wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2to C9) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 0 is selected from the group consisting of

each of the R a and R b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 3 group,

or R a and R b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 3 group; each R 2 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to

Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 3 group;

each R 3 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 4a R 4b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C9) cycloheteroalkyi, (C6to C10) aryl, (C2to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2to C10) cycloheteroalkylene, -C(0)R 5 ,-C(0)NR 4a R 4b , -S(0)mR 15 , -S(0)mNR 4a R 4b , -NR 4a S(0)mR 15 , -(CH 2 )nC(0)OR 15 ,

-(CH 2 )nC(0)N(R 4a R 4b ), -(CH 2 )nN(R 4a R 4b ), -OC(0)R 15 , -0(CH 2 ) n O-, -NR 4a C(0)R 15 , and

-NR 4a C(0)N(R 4a R 4b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3to C10) cycloalkylene, and (C 2 to C 0 ) cycloheteroalkylene is optionally substituted with at least one R 6 group;

each of the R 4a and R 4b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10)

cycloalkenyl, (C 2 to C 9 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 6 group,

or R 4a and R 4b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 6 group; each R 5 is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to Cio) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto Cio) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 6 group;

each R 6 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 7a R 7b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)NR 7a R 7b , C(0)R 18 , S(0)mR 18 , -S(0)mNR 7a R 7b , -NR 7a S(0)mR 18 , -(CH 2 )nC(0)OR 18 ,

-(CH 2 )nC(0)N(R 7a R 7b ), -(CH 2 )nN(R 7a R 7b ), -OC(0)R 18 , -NR 7a C(0)R 18 , and -NR 7a C(0)N(R 7a R 7b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) aryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2to C10) cycloheteroalkylene is optionally substituted with at least one R 9 group;

each of the R 7a and R 7b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Cito C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, C 6 to C 0 ) aryl, (C 2 to C 9 ) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to Cg) heteroarylene, wherein

each of the said (C^o C 0 ) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C 3 to C 0 ) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, and (C 2 to C 9 ) heteroarylene is optionally substituted with at least one R 9 group,

or R 7a and R 7b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 9 group; each R 8 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, and (C2to C9) heteroaryl, wherein each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C 3 to C 0 ) cycloalkyi, (Csto C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, and (C 6 to C 0 ) aryl is optionally substituted with at least one R 9 group;

each R 9 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 20a R 20b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Csto C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R ,-C(0)NR^ ua R , -S(0)mR , -S(0)mNR^ ua R , -NR^ ua S(0)mR , -(CH 2 )nC(0)OR , - (CH 2 )nC(O)N(R 20a R 20b ), -(CH 2 )nN(R 20a R 20b ), -OC(0)R 21 , -NR 20a C(O)R 21 , and -NR 20a C(O)N(R 20a R 20b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to Cio) cycloalkyi, (Cs to Cio) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 22 group;

each of the R 20a and R 20b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl, wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C 3 to C 0 ) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl is optionally substituted with at least one R 22 group,

or R 20a and R 20b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S, and wherein

the said (C 2 to C 0 ) cycloheteroalkyi ring is optionally substituted with at least one R 22 group; each R 2 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

each R 22 is independently selected from hydrogen, halogen, OH, nitro, CF3, -NR 23a R 23b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)NR 23a R 23b , -C(0)R 24 , -S(0)mR 24 , -S(0)mNR 23a R 23b , -NR 23a S(0)mR 24 , -(CH 2 )nC(0)OR 24 ,

-(CH 2 )nC(0)N(R 23a R 23b ), -(CH 2 )nN(R 23a R 23b ), -OC(0)R 24 , -NR 23a C(0)R 24 , and -NR 23a C(0)N(R 23a R 23b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 25 group;

each of the R 23a and R 23b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 )

cycloheteroalkyi, (C 2 to C 9 ) heteroaryl, and (C 6 to C 0 ) aryl;

each R 24 is independently selected from hydrogen, halogen, OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

R 25 is selected from hydrogen, halogen, OH, nitro, CF3, -NR 26a R 26b , oxo, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, (C2 to C9) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to Cg) heteroarylene, (C3 to C10) cycloalkylene, (C2 to C10) cycloheteroalkylene, -C(0)R 27 ,

-C(0)NR 26a R 26b , -S(0)mR 27 , -S(0)mNR 26a R 26b , -NR 26a S(0)mR 27 , -(CH 2 )nC(0)OR 27 ,

-(CH 2 )nC(0)N(R 26a R 26b ), -(CH 2 )nN(R 26a R 26b ), -OC(0)R 27 , -NR 26a C(0)R 27 , and -NR 26a C(0)N(R 26a R 26b ), wherein

each of the said (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, (C 2 to Cg) heteroaryl, (C 6 to C 0 ) arylene, (C 2 to C 9 ) heteroarylene, (C3 to C10) cycloalkylene, and (C2 to C10) cycloheteroalkylene is optionally substituted with at least one R 27 group;

each of the R 26a and R 26b is independently selected from hydrogen, (C^ to C 0 ) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C 2 to C 0 ) cycloheteroalkyi, (C 6 to C 0 ) aryl, and (C 2 to C 9 ) heteroaryl,

or R 26a and R 26b may be taken together with the nitrogen atom to which they are attached to form a (C 2 to C 0 ) cycloheteroalkyi ring, wherein

said (C 2 to C 0 ) cycloheteroalkyi ring has 1 to 3 ring heteroatoms selected from the group consisting of N, O, and S;

each R 27 is independently selected from hydrogen, halogen , OH, nitro, CF3, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (Ci to C10) alkoxy, aryloxy, cyano, (C3 to C10) cycloalkyi, (Cs to C10) cycloalkenyl, (C2 to C10) cycloheteroalkyi, (C6 to C10) aryl, and (C2 to C9) heteroaryl;

h is 1 or 2;

i is 2, 3, 4, 5, or 6;

j is 0, 1 , 2, 3, 4, or 5;

k is 1 , 2, 3, 4, or 5;

m is 0, 1 or 2;

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

Z is selected from the group consisting of -0-, -S-, -S(O)-, and -S(0) 2 -;

with the proviso that

when R 1 R 2 or R 3 is alk l or h dro en X is C-C(R 6a R 6b ) r D, Y is CH 2 , and D is then NR R cannot be an optionally substituted piperazine or 1 ,4-diazepane.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 , wherein A-D is defined as above.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ; A is -C(R 6a R 6b ) j -; and

O

D is -X^NR 7a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

O

U

D is *i W a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl,

(Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl.wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

O

D is -^N ^ 7 " .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein : X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl.wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R is hydrogen;

R 5 is hydrogen; and

X O

D is ' NR^R 7 ^

except when R is phenyl, R 2 is hydrogen, and j is 0,

then NR 7a R 7b cannot be

and when R is hydrogen, halogen, or hydroxy, R 2 is hydrogen, R 3 is hydrogen, X is C-A-D, Y is

then j is 2,3,4, or 5, but not 0 or 1 .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen; R 5 is hydrogen; and

O

D is -X^NR 7a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V; and

D is

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to Cio) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -; and

R 7a

D is ^R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 ggrroouupp;;

R 4 is hydrogen; R 5 is hydrogen; and

R 7a

D is ^% 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyl, and (Csto C10) cycloalkenyl, (Cito C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyl, (Csto C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3to C10) cycloalkyl, and (Csto C10) cycloalkenyl, wherein

each of the said (Cito C10) alkyl, (Cito C10) alkenyl, (Cito C10) alkynyl, (C3to C10) cycloalkyl, (Csto C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

R 7a

I

D is ^ 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

D is ^ 7b . In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V; and

° °

D is ^ S NR 7a R 7b or S NR 7a R 7b

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

0 0 o

D is ^NR 7a R 7b or S a R 7b

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

P °

D is ^NR 7a R 7b or S ^NR 7a R 7b

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R is hydrogen

R 5 is hydrogen; and

R 8

I

Χ \ NLγ .NR 7a R 7b Χ "vNγ NR 7a R 7b

D is S or O In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

D i

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

D is In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D, and Y is a bond or CR 4 R 5 ; or

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6 ) j -; and

wherein R 0a is selected from the group consisting

R is selected from (C6to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group; R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen; and

R 5 is hydrogen.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b ) j -; and

O

D is-'O NR 7a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

O

D is -X^NR 7a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group; R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

O

D is ^ NR^R 7 " .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen; and

O

X

D is 'V"NR 7a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

6 a R 6b ) j -; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R group; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl.wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen; and In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V; and

R 7a

I

D is ^ 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 7a

I

D is 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group; and

R 7a

I

D is 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein : X is C-A-D and Y is a bond;

A is -C(R 6a R 6b ) j -;

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group; and

R 3 is hydrogen;

R 7a

D is ¾- 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -; and

X O

D is ' NRV

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b V;

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6to C10) aryl and (C2to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

O

D is ^NR 7a R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -

R is selected from (C6to C10) aryl and (C2 to C9) heteroaryl, wherein each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

O

U

D is

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -; and In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryland (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryland (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Csto C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Csto C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Csto C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Csto C10) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -; R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen; and

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -; and

D is ^R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

D is ^R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b V;

R is selected from (C6 to C10) aryland (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to Cio) cycloalkenyl is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to Cio) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to Cio) alkyl, (Ci to Cio) alkenyl, (Ci to Cio) alkynyl, (C3 to Cio) cycloalkyl,

(Cs to Cio) cycloalkenyl is optionally substituted with at least one R 3 group;

R 4 is hydrogen;

R 5 is hydrogen; and

D is ^R 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -;

R is selected from (C6 to Cio) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 2 is selected from hydrogen and (Ci to C10) alkyl, wherein the said (Ci to C10) alkyl is optionally substituted with at least one R 3 group;

R 3 is hydrogen;

R 4 is hydrogen;

R 5 is hydrogen; and

R 7a

I

D is ^ 7b .

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) j -;

D is selected from the group consisting of

R is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group; R 2 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, (Ci to C10) alkoxy, -(CH 2 )nC(0)N(R a R b ), and -C(0)R 12 , wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl, and (Ci to C10) alkoxy is optionally substituted with at least one R 3 group;

R 3 is selected from hydrogen, (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, and (Cs to C10) cycloalkenyl, wherein

each of the said (Ci to C10) alkyl, (Ci to C10) alkenyl, (Ci to C10) alkynyl, (C3 to C10) cycloalkyl, (Cs to C10) cycloalkenyl is optionally substituted with at least one R 3 group.

R 4 is selected from (C6 to C10) aryl and (C2 to C9) heteroaryl, wherein

each of the said (C6 to C10) aryl and (C2 to C9) heteroaryl is optionally substituted with at least one R 3 group;

R 5 is hydrogen.

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is C-A-D and Y is CR 4 R 5 ;

A is -C(R 6a R 6b ) r , (C6 to C10) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6 to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R 6a group; and

D is selected from the group consisting of

R 8 O

<·». z

¾!^R 10b , and Z ^ R ( > .

, wherein

R 0b is selected from the group consisting of

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein :

X is C-A-D and Y is a bond; A is -C(R 6a R 6b ) r , (C6to Cio) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R 6a group; and

D is selected from the group consisting of n

R 0b is selected from the group consisting of

b

In another embodiment, the invention relates to compounds of Structural Formula I, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, wherein:

X is CR 5 and Y is CR 4 -A-D;

A is -C(R 6a R 6b ) r , (C6to C10) arylene, or (C2 to C9) heteroarylene, wherein

each of the said (C6to C10) arylene or (C2 to C9) heteroarylene is optionally substituted with at least one R 6a group; and

D is selected from the group consisting of

R 8 O

X% 10b > -|-R i o b ¾ Rl o b j and , wherein

R 0b is selected from the group consisting of 1 b

R 9b , and R . R 9b

In another embodiment, the invention relates to compounds, or a pharmaceutically acceptable salt, and a pharmaceutically acceptable carrier, diluent, or vehicle thereof, selected from the group consisting the compounds described as examples A1 to A1 16, A201 to A205, C1 to C19, E1 to E6 in the method of preparation section, and examples B1 to B19 in table 5 and D1 to D47 in Table 7.

DEFINITIONS

As used herein, the terms "comprising" and "including" are used in their open, non-limiting sense.

The terms "halo" and/or "halogen" refer to fluorine, chlorine, bromine or iodine.

The term "(C-i to C 0 ) alkyl" refers to a saturated aliphatic hydrocarbon radical including straight chain and branched chain groups of 1 to 8 carbon atoms. Examples of (C-i to C 0 ) alkyl groups includemethyl, ethyl, propyl, 2-propyl, n-butyl, / ' so-butyl, fe/ -butyl, pentyl, and the like. The terms "Me" and"methyl," as used herein, mean a -CH 3 group. The terms "Et" and "ethyl," as used herein, mean a - C 2 H 5 group.

The term "(C 2 to C 0 ) alkenyl", as used herein, means an alkyl moiety comprising 2 to 10 carbons having at least one carbon-carbon double bond. The carbon-carbon double bond in such a group may be anywhere along the 2 to 10 carbon chain that will result in a stable compound. Such groups include both the E and Z isomers of said alkenyl moiety. Examples of such groups include, but are not limited to, ethene, propene, 1 -butene, 2-butene, 1 -pentene, 2-pentene, 1 -hexene, 2-hexene, and 3-hexyene Examples of such groups include, but are not limited to, ethenyl, propenyl, butenyl, allyl, and pentenyl.

The term "allyl," as used herein, means a -CH 2 CH=CH 2 group.

As used herein, the term "(C 2 to C 0 ) alkynyl" means an alkyl moiety comprising from 2 to 8 carbon atoms and having at least one carbon-carbon triple bond. The carbon-carbon triple bond in such a group may be anywhere along the 2 to 10 carbon chain that will result in a stable compound. Examples of such groups include, but are not limited to, ethyne, propyne, 1 -butyne, 2-butyne, 1 - pentyne, 2-pentyne, 1 -hexyne, 2-hexyne, and 3-hexyne.

The term "(CT to C 0 ) alkoxy", as used herein, means an O-alkyl group wherein said alkyl group contains from 1 to 8 carbon atoms and is straight, branched, or cyclic. Examples of such groups include, but are not limited to, methoxy, ethoxy, n-propyloxy, iso-propyloxy, n-butoxy, 5 iso-butoxy, tert-butoxy, cyclopentyloxy, and cyclohexyloxy.

The term "(C 6 to C 0 ) aryl", as used herein, means a group derived from an aromatic hydrocarboncontaining from 6 to 10 carbon atoms. Examples of such groups include, but are not limited to, phenyl or naphthyl. The terms "Ph" and "phenyl," as used herein, mean a -C 6 H 5 group. The term "benzyl," as used herein, means a -CH 2 C 6 H 5 group.

The term "(C 6 to C 0 ) arylene" is art-regognized, and as used hereinpertains to a bivalent moiety obtained by removing a hydrogen atom from a (C 6 to C 0 ) aryl ring, as defined above.

"(C 2 to C 9 ) heteroaryl", as used herein, means an aromatic heterocyclic group having a total of from5 to 10 atoms in its ring, and containing from 2 to 9 carbon atoms and from one to four heteroatoms each independently selected from O, S and N, and with the proviso that the ring of said group does not containtwo adjacent O atoms or two adjacent S atoms. The heterocyclic groups include benzo-fused ring systems. Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl.pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. The C 2 to C 9 heteroaryl groups may be C-attached or N-attached where such is possible. For instance, a group derived from pyrrole may be pyrrol-1 -yl (N- attached) or pyrrol-3-yl (C-attached). Further, a group derived from imidazole may be imidazol-1 -yl (N- attached) or imidazol-3-yl (C-attached).

The term "(C 2 to C 0 ) heteroarylene" is art-recognized, and as used herein pertains to a bivalent moiety obtained by removing a hydrogen atom from a (C 6 to C 0 ) heteroaryl ring, as defined above.

The term "(C 2 to C 0 ) cycloheteroalkyi", as used herein, means a non-aromatic, monocyclic, bicyclic, tricyclic.spirocyclic, or tetracyclic group having a total of from 4 to 13 atoms in its ring system, and containing from 5 to 10 carbon atoms and from 1 to 4 heteroatoms each independently selected from O, S and N, and with the proviso that the ring of said group does not contain two adjacent O atoms or two adjacent S atoms. Furthermore, such (C 2 to C 0 ) cycloheteroalkyi groups may contain an oxo substituent at any available atom that will result in a stable compound. For example, such a group may contain an oxo atom at an available carbon or nitrogen atom. Such a group may contain more than one oxo substituent if chemically feasible. In addition, it is to be understood that when such a (C 2 to C 0 ) cycloheteroalkyi group contains a sulfur atom, said sulfur atom may be oxidized with one or two oxygen atoms to afford either a sulfoxide or sulfone. An example of a 4 membered

cycloheteroalkyi group is azetidinyl (derived from azetidine). An example of a 5 membered cycloheteroalkyi group is pyrrolidinyl. An example of a 6 membered cycloheteroalkyi group is piperidinyl. An example of a 9 membered cycloheteroalkyi group is indolinyl. An example of a 10 membered cycloheteroalkyi group is 4H-quinolizinyl. Further examples of such (C 2 to C 0 ) cycloheteroalkyi groups include, but are not limited to, tetrahydrofuranyl, dihydrofuranyl,

tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4/-/-pyranyl, dioxanyl, 1 ,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3- azabicyclo[3.1 .0]hexanyl, 3-azabicyclo[4.1 .0]heptanyl, 3/-/-indolyl, quinolizinyl, 3-oxopiperazinyl, 4- methylpiperazinyl, 4-ethylpiperazinyl, and 1 -oxo-2,8,diazaspiro[4.5]dec-8-yl.The (C 2 to C 0 )heteroaryl groups may be C-attached or N-attached where such is possible. For instance, a group derived from piperazine may be piperazin-1 -yl (N-attached) or piperazin-2-yl (C-attached).

The term "(C 2 to C 0 ) cycloheteroalkylene" is art-recognized, and as used herein pertains to a bidentate moiety obtained by removing a hydrogen atom from a (C 6 to C 0 ) cycloheteroalkyl ring, as defined above.

The term "(C 3 to C 0 ) cycloalkyl group" means a saturated, monocyclic, fused, spirocyclic, or polycyclic ring structure having a total of from 3 to 10 carbon 5 ring atoms. Examples of such groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptyl, and adamantyl.

The term "(C 3 to C 0 ) cycloalkylene" is art-recognized, and as used herein pertains to a bidentate moiety obtained by removing a hydrogen atom from a (C 3 to C 0 ) cycloalkyl ring, as defined above.

The term "spirocyclic" as used herein has its conventional meaning, that is, any compound containing two or more rings wherein two of the rings have one ring carbon in common. The rings of a 10 spirocyclic compound, as herein defined, independently have 3 to 20 ring atoms. Preferably, they have 3 to 10 ring atoms. Non-limiting examples of a spirocyclic compound include spiro[3.3]heptane, spiro[3.4]octane, and spiro[4.5]decane.

The term "(C 5 to C 8 ) cycloalkenyl" means an unsaturated, monocyclic, fused, spirocyclic ring structures having a total of from 5 to 8 carbon ring atoms. Examples of such groups include, but not limited to, cyclopentenyl, cyclohexenyl.

The term cyano" refers to a -C≡N group.

An "aldehyde" group refers to a carbonyl group where R is hydrogen.

An "alkoxy" group refers to both an -O-alkyl and an -O-cycloalkyl group, as defined herein. An "alkoxycarbonyl" refers to a -C(0)OR.

An "alkylaminoalkyl" group refers to an -alkyl-NR-alkyl group.

An "alkylsulfonyl" group refer to a -S0 2 alkyl.

An "amino" group refers to an -NH 2 or an -NRR'group.

An "aminoalkyl" group refers to an -alky-NRR' group.

An "aminocarbonyl" refers to a -C(0)NRR'.

An "arylalkyl" group refers to -alkylaryl, where alkyl and aryl are defined herein.

An "aryloxy" group refers to both an -O-aryl and an -O-heteroaryl group, as defined herein.

An "aryloxycarbonyl" refers to -C(0)Oaryl.

An "arylsulfonyl" group refers to a -S0 2 aryl.

A "C-amido" group refers to a -C(0)NRR' group. A "carbonyl" group refers to a -C(0)R.

A "C-carboxyl" group refers to a -C(0)OR groups.

A "carboxylic acid" group refers to a C-carboxyl group in which R is hydrogen.

A "cyano" group refers to a -CN group.

A "dialkylamionalkyl" group refers to an— (alkyl)N(alkyl)2 group.

A "halo" or "halogen" group refers to fluorine, chlorine, bromine or iodine.

A "haloalkyl" group refers to an alkylgroup substituted with one or more halogen atoms.

A "heteroalicycloxy" group refers to a heteroalicyclic-O group with heteroalicyclic as defined herein.

A "heteroaryloxyl" group refers to a heteroaryl-O group with heteroaryl as defined herein.

A "hydroxy" group refers to an -OH group.

An "N-amido" group refers to a -R'C(0)NR group.

An "N-carbamyl" group refers to a -ROC(0)NR-group.

A "nitro" group refers to a -N0 2 group.

An "N-Sulfonamido" group refers to a -NR-S(0) 2 R group.

An "N-thiocarbamyl" group refers to a ROC(S)NR' group.

An "O-carbamyl" group refers to a -OC(0)NRR' group.

An "O-carboxyl" group refers to a RC(0)0 group.

An "O-thiocarbamyl" group refers to a -OC(S)NRR' group.

An "oxo" group refers to a carbonyl moiety such that alkyl substituted by oxo refers to a ketone group.

A "perfluoroalkyl group" refers to an alkyl group where all of the hydrogen atoms have been replaced with fluorine atoms.

A "phosphonyl" group refers to a -P(0)(OR) 2 group.

A "silyl" group refers to a -SiR 3 group.

An "S-sulfonamido" group refers to a -S(0) 2 NR-group.

A "sulfinyl" group refers to a -S(0)R group.

A "sulfonyl" group refers to a -S(0) 2 R group.

A "thiocarbonyl" group refers to a -C(=S)-R group.

A "trihalomethanecarbonyl" group refers to a Z 3 CC(0) group, where Z is halogen.

A "trihalomethanesulfonamido" group refers to a Z 3 CS(0) 2 NR-group, where Z is halogen.

A "trihalomethanesulfonyl" group refers to a Z 3 CS(0) 2 group, where Z is halogen.

A "trihalomethyl" group refers to a -CZ 3 group.

A "C-carboxyl" group refers to a -C(0)OR groups.

The term "substituted," means that the specified group or moiety bears one or more substituents.

The term "unsubstituted," means that the specified group bears no substituents. The term "optionally substituted" means that the specified group is unsubstituted or substituted by one or more substituents. Itis to be understood that in the compounds of the present invention when a group is said to be"unsubstituted," or is "substituted" with fewer groups than would fill the valencies of all the atoms in thecompound, the remaining valencies on such a group are filled by hydrogen. For example, if a C 6 arylgroup, also called "phenyl" herein, is substituted with one additional substituent, one of ordinary skill in theart would understand that such a group has 4 open positions left on carbon atoms of the C 6 aryl ring (6 initial positions, minus one to which the remainder of the compound of the present invention is bonded, minus an additional substituent, to leave 4). In such cases, the remaining 4 carbon atoms are each bound to one hydrogen atom to fill their valencies. Similarly, if a C 6 aryl group in the present compounds is said to be "disubstituted," one of ordinary skill in the art would understand it to mean that the C 6 aryl has 3 carbon atoms remaining that are unsubstituted. Those three unsubstituted carbon atoms are each bound to one hydrogen atom to fill their valencies.

The term "solvate," is used to describe a molecular complex between compounds of the presentinvention and solvent molecules. Examples of solvates include, but are not limited to, compounds of theinvention in combination water, isopropanol, ethanol, methanol, dimethylsulfoxide (DMSO), ethyl acetate, acetic acid, ethanolamine, or mixtures thereof.

The term "hydrate" can be used when said solvent is water. It is specifically contemplated that in the present invention one solvent molecule can be associatedwith one molecule of the compounds of the present invention, such as a hydrate. Furthermore, it isspecifically contemplated that in the present invention, more than one solvent molecule may beassociated with one molecule of the compounds of the present invention, such as a dihydrate.

Additionally, it is specifically contemplated that in the present invention less than one solvent molecule may be associated with one molecule of the compounds of the present invention, such as a hemihydrate. Furthermore, solvates of the present invention are contemplated as solvates of compounds of the present invention that retain the biological effectiveness of the non-hydrate form of the compounds.

The term "pharmaceutically acceptable salt," as used herein, means a salt of a compound of thepresent invention that retains the biological effectiveness of the free acids and bases of the specifiedderivative and that is not biologically or otherwise undesirable.

The term "pharmaceutically acceptable formulation", as used herein, means a combination of acompound of the invention, or a salt or solvate thereof, and a carrier, diluent, and/or excipient(s) that arecompatible with a compound of the present invention, and is not deleterious to the recipient thereof. Pharmaceutical formulations can be prepared by procedures known to those of ordinary skill in the art. For example, the compounds of the present invention can be formulated with common excipients.diluents, or carriers, and formed into tablets, capsules, and the like. Examples of excipients, diluents, andcarriers that are suitable for such formulations include the following: fillers and extenders such as starch.sugars, mannitol, and silicic derivatives; binding agents such as

carboxymethyl cellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl pyrrolidone; moisturizing agents such as glycerol;disintegrating agents such as povidone, sodium starch glycolate, sodium carboxymethylcellulose, agar.calcium carbonate, and sodium bicarbonate; agents forretarding dissolution such as paraffin; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol, glycerol monostearate; adsorptive carriers such as kaolin and bentonite; and lubricants such as talc, calcium and magnesium stearate and solid polyethylene glycols. Final pharmaceutical forms may be pills, tablets, powders, lozenges, saches, cachets, or sterile packaged powders, and the like, depending on the type of excipient used. Additionally, it is specifically contemplated that pharmaceutically acceptable formulations of the present invention can contain more than one active ingredient. For example, such formulations may contain more than one compound according to the present invention. Alternatively, such formulations may contain one or more compounds of the present invention and one or more additional agents that reduce abnormal cell growth.

The term "virus inhibiting amount" as used herein, refers to the amount of a compound of thepresent invention, or a salt or solvate thereof, required to inhibit the cell entry of an enveloped virus in vivo, such as in a mammal, or in vitro. The amount of such compounds required to cause such inhibition can be determined without undue experimentation using methods described herein and those known to those of ordinary skill in the art.

The terms "treat", "treating", and "treatment" with reference to enveloped virus infection, in mammals, particularly a human, include: (i) preventing the disease or condition from occurring in a subject which may be predisposed to the condition, such that the treatment constitutes prophylactic treatment for the pathologic condition; (ii) modulating or inhibiting the disease or condition, i.e., arresting its development; (iii) relieving the disease or condition, i.e., causing regression of the disease or condition; or (iv) relieving and/or alleviating the disease or condition or the symptoms resulting from the disease or condition.

The compositions are delivered in effective amounts. The term "effective amount" refers to the amount necessary or sufficient to realize a desired biologic effect and/or reduce the viral load.

Combined with the teachings provided herein, by choosing among the various active compounds and weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side- effects and preferred mode of administration, an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is effective to treat the particular subject. In addition, based on testing, toxicity of the inhibitor is expected to be low. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular inhibitor being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular inhibitor and/or other therapeutic agent without necessitating undue experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to some medical judgment. Multiple doses per day may be contemplated to achieve appropriate systemic levels of compounds. Appropriate systemic levels can be determined by, for example, measurement of the patient's peak or sustained plasma level of the drug.

"Dose"and"dosage" are used interchangeably herein. For any compound described herein, the therapeutically effective amount can be initially determined from preliminary in vitro studies and/or animal models. At herapeutically effective dose can also be determined from human data for inhibitors that have been tested in humans and for compounds, which are known to exhibit similar

pharmacological activities, such as other related active agents. The applied dose can be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other methods well-known in the art, is well within the capabilities of the ordinarily skilled artisan. In certain embodiments, the methods of the invention are useful for treating infection with enveloped viruses.

Unless indicated otherwise, all references herein to the inventive compounds include references to salts, solvates, and complexes thereof, including polymorphs, stereoisomers, tautomers, and isotopically labeled versions thereof. For example, compounds of the present invention can be pharmaceutically acceptable salts and/or pharmaceutically acceptable solvates.

The term "stereoisomers" refers to compounds that have identical chemical constitution, but differ with regard to the arrangement of their atoms or groups in space. In particular, the term

"enantiomers" refers to two stereoisomers of a compound that are non-superimposable mirror images of one another.

A pure enantiomer can be contaminated with up to 2% of the opposite enantiomer.

The terms "racemic" or "racemic mixture," as used herein, refer to a 1 :1 mixture of enantiomers of aparticular compound. The term "diastereomers", on the other hand, refers to the relationship between a pair of stereoisomers that comprise two or more asymmetric centers and are not mirror images of one another. In accordance with a convention used in the art, the symbol is used in structural formulas herein to depict the bond that is the point of attachment of the moiety or substituent to the core or backbone structure. In accordance with another convention, in some structural formulae herein the carbon atoms and their bound hydrogen atoms are not explicitly depicted, e.g., ¾ represents a methyl group, ¾ represents an ethyl group, represents a cyclopentyl group, etc.

The compounds of the present invention may have asymmetric carbon atoms. The carbon- carbonbonds of the compounds of the present invention may be depicted herein using a solid line ( — ), a solid wedge (— ), or a dotted wedge ( ). The use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers (e.g. specific enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of either a solid or dotted wedge to depict bonds to asymmetric carbon atoms is meant to indicate that only the stereoisomer shown is meant to be included. It is possible that compounds of the invention may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included. For example, unless stated otherwise, it is intended that the compounds of the present invention can exist as enantiomers and diastereomers or as racemates and mixtures thereof. The use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of the invention and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is present.

(R) z then, unless otherwise defined, a substituent "R" may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the reacemate using, for example, chiral high pressure liquid chromatography (HPLC). Alternatively, the racemate (or a racemic precursor) maybe reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound contains an acidic or basic moiety, an acid or base such as tartaric acid or 1 -phenyl ethyl amine.The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the

corresponding pure enantiomer(s) by means well known to one skilled in the art. Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% isopropanol, typically from 2 to 20%, and from 0 to 5% of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture. Stereoisomeric conglomerates may be separated by conventional techniques known to those skilled in the art. See, e.g. "Stereochemistry of Organic Compounds" by E L Eliel (Wiley, New York, 1994), the disclosure of which is incorporated herein by reference in its entirety.

Where a compound of the invention contains an alkenyl or alkenylene group, geometric cis/trans (or Z/E) isomers are possible. Where the compound contains, for example, a keto or oxime group or an aromatic moiety, tautomeric isomerism ('tautomerism') can occur. Examples of tautomerism include keto and enol tautomers. A single compound may exhibit more than one type of isomerism. Included within the scope of the invention are all stereoisomers, geometric isomers and tautomeric forms of the inventive compounds, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallization.

The compounds of the present invention may be administered as prodrugs. Thus certain derivatives of compounds of Formula I which may have little or no pharmacological activity themselves can, when administered to a mammal, be converted into a compound of Formula I having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as

"prodrugs". Prodrugs can, for example, be produced by replacing appropriate functionalities present in the compound of Formula I with certain moieties known to those skilled in the art. See, e.g. "Pro-drugs as Novel Delivery Systems", Vol. 14, ACS Symposium Series (T Higuchi and W Stella) and

"Bioreversible Carriers in Drug Design", Pergamon Press, 1987 (ed. E B Roche, American

Pharmaceutical Association), the disclosures of which are incorporated herein by reference in their entireties. Some examples of such prodrugs include: an ester moiety in the place of a carboxylic acid functional group; an ether moiety or an amide moiety in place of an alcohol functional group; and an amide moiety in place of a primary or secondary amino functional group. Further examples of replacement groups are known to those of skill in the art. See, e.g. "Design of Prodrugs" by H Bundgaard (Elsevier, 1985), the disclosure of which is incorporated herein by reference in its entirety. It is also possible that certain compounds of Formula I may themselves act as prodrugs of other compounds of Formula I.

Salts of the present invention can be prepared according to methods known to those of skill in the art. Examples of salts include, but are not limited to, acetate, acrylate, benzenesulfonate, benzoate (such as chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, and methoxybenzoate), bicarbonate, bisulfate, bisulfite, bitartrate, borate, bromide, butyne-1 ,4-dioate, calcium edetate, camsylate, carbonate, chloride, caproate, caprylate, clavulanate, citrate, decanoate, dihydrochloride, dihydrogenphosphate, edetate, edislyate, estolate, esylate, ethylsuccinate, formate, fumarate, gluceptate, gluconate, glutamate, glycollate, glycollylarsanilate, heptanoate, hexyne-1 ,6- dioate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, γ-hydroxybutyrate, iodide, isobutyrate, isothionate, lactate, lactobionate, laurate, malate, maleate, malonate, mandelate, mesylate, metaphosphate, methanesulfonate, methylsulfate, monohydrogenphosphate, mucate, napsylate, naphthalene-1 -sulfonate, naphthalene-2-sulfonate, nitrate, oleate, oxalate, pamoate (embonate), palmitate, pantothenate, phenylacetates, phenylbutyrate, phenylpropionate, phthalate, phospate/diphosphate, polygalacturonate.propanesulfonate, propionate, propiolate, pyrophosphate, pyrosulfate, salicylate, stearate, subacetate, suberate, succinate, sulfate, sulfonate, sulfite, tannate, tartrate, teoclate, tosylate, triethiodode, and valerate salts.

The compounds of the present invention that are basic in nature are capable of forming a wide variety of different salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animals, it is often desirable in practice to initially isolate the compound of the present invention from the reaction mixture as a pharmaceutically unacceptable salt and then simply convert the latter back to the free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition salt. The acid addition salts of the base compounds of this invention can be prepared by treating the base compound with a substantially equivalent amount of the selected mineral or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or ethanol. Upon evaporation of the solvent, the desired solid salt is obtained. The desired acid salt can also be precipitated from a solution of the free base in an organic solvent by adding an appropriate mineral or organic acid to the solution.

Those compounds of the present invention that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include the alkali metal or alkaline-earth metal salts and particularly, the sodium and potassium salts. These salts are all prepared by conventional techniques. The chemical bases which are used as reagents to prepare the pharmaceutically acceptable base salts of this invention are those which form non-toxic base salts with the acidic compounds of the present invention. Such non-toxic base salts include those derived from such pharmacologically acceptable cations as sodium, potassium calcium and magnesium, etc. These salts can be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, preferably under reduced pressure. Alternatively, they may also be prepared by mixing lower alkanolic solutions of the acidic compounds and the desired alkali metal alkoxide together, and then evaporating the resulting solutionto dryness in the same manner as before. In either case, stoichiometric quantities of reagents are preferably employed in order to ensure completeness of reaction and maximum yields of the desired final product.

If the inventive compound is a base, the desired salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, suchas acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalicacid, glycolic acid, salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic acid, or the like.

If the inventive compound is an acid, the desired salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative examples of suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, primary, secondary, and tertiary amines, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.

In the case of agents that are solids, it is understood by those skilled in the art that the inventive compounds, agents and salts may exist in different crystal or polymorphic forms, all of which are intended to be within the scope of the present invention and specified formulas.

The invention also includes isotopically-labeled compounds of the invention, wherein one or moreatoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as C, 3 C and 4 C, chlorine, such as 36 CI, fluorine, such as 8 F, iodine, such as 23 l and 25 l, nitrogen, such as 3 N and 5 N, oxygen, such as 5 0, 7 0 and 8 0, phosphorus, such as 32 P, and sulfur, such as 35 S.

Certain isotopically-labeled compounds of the invention, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, 3 H, and carbon-14, 4 C, are particularly useful for this purpose in view of their ease ofincorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, 35 S increased in vivo half-life or reduced dosage requirements, and hence may be preferred in somecircumstances. Substitution with positron emitting isotopes, such as C, 8 F, 5 0 and 3 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.

Isotopically-labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.

The compounds of the present invention may be formulated into pharmaceutical compositions as described below in any pharmaceutical form recognizable to the skilled artisan as being suitable. Pharmaceutical compositions of the invention comprise a therapeutically effective amount of at least one compound of the present invention and an inert, pharmaceutically acceptable carrier or diluent.

To treat or prevent diseases or conditions mediated in part or whole by enveloped virus infection, a pharmaceutical composition of the invention is administered in a suitable formulation prepared by combining a therapeutically effective amount (i.e., an enveloped virus GP- or host cell partner- modulating, regulating, or inhibiting amount effective to achieve therapeutic efficacy) of at least one compound of the present invention (as an active ingredient) with one or more

pharmaceutically suitable carriers, which may be selected, for example, from diluents, excipients and auxiliaries that facilitate processing of the active compounds into the final pharmaceutical preparations.

The pharmaceutical carriers employed may be either solid or liquid. Exemplary solid carriers are lactose, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemplary liquid carriers are syrup, peanut oil, olive oil, water and the like. Similarly, the inventive compositions may include time-delay or time-release material known in the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate or the like. Further additives or excipients may be added to achieve the desired formulation properties. For example, a bioavailability enhancer, such as Labrasol, Gelucire or the like, or formulator, such as CMC (carboxy-methylcellulose), PG (propyleneglycol), or PEG

(polyethyleneglycol), may be added. Gelucire®, a semi-solid vehicle that protects active ingredients from light, moisture and oxidation, may be added, e.g., when preparing a capsule formulation.

If a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form, or formed into a troche or lozenge. The amount of solid carrier may vary, but generally will be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation may be in the form of syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampoule or vial or non-aqueous liquid suspension. If a semi-solid carrier is used, the preparation may be in the form of hard and soft gelatin capsule formulations. The inventive compositions are prepared in unit-dosage form appropriate for the mode of administration, e.g. parenteral or oral administration.

To obtain a stable water-soluble dose form, a salt of a compound of the present invention may be dissolved in an aqueous solution of an organic or inorganic acid, such as a 0.3 M solution of succinic acid or citric acid. If a soluble salt form is not available, the agent may be dissolved in a suitable co-solvent or combinations of co-solvents. Examples of suitable co-solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0 to 60% of the total volume. In an exemplary embodiment, a compound of the present invention is dissolved in DMSO and diluted with water. The composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle such as water or isotonic saline or dextrose solution.

Proper formulation is dependent upon the route of administration selected. For injection, the agents of the compounds of the present invention may be formulated into aqueous solutions, preferably in physiologically compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer.

For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.

For oral administration, the compounds can be formulated by combining the active compounds with pharmaceutically acceptable carriers known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained using a solid excipient in admixture with the active ingredient (agent), optionally grinding the resulting mixture, and processing the mixture of granules after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include: fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; and cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as crosslinked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, polyvinyl pyrrolidone, Carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.

Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active agents.

Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active agents may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.

For administration intranasally or by inhalation, the compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount.

Capsules and cartridges of gelatin for use in an inhaler or insufflator and the like may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.

The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit-dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such 35 forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.

Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active agents may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.

Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. sterile pyrogen-free water, before use.

In addition to the formulations described above, the compounds of the present invention may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion-exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. A pharmaceutical carrier for hydrophobic compounds is a cosolvent system comprising benzyl alcohol, a non-polar surfactant, a water-miscible organic polymer, and an aqueous phase. The co-solvent system may be a VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the non-polar surfactant polysorbate 80, and 65% w/v polyethylene glycol300, made up to volume in absolute ethanol. The VPD co-solvent system (VPD: 5W) contains VPD diluted 1 :1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. The proportions of a cosolvent system may be suitably varied without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may be substituted for dextrose.

Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at thecost of greater toxicity due to the toxic nature of DMSO. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymerscontaining the therapeutic agent. Various sustained-release materials have been established and are known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.

The pharmaceutical compositions also may comprise suitable solid- or gel-phase carriers or excipients. These carriers and excipients may provide marked improvement in the bioavailability of poorly soluble drugs. Examples of such carriers or excipients include calcium carbonate, calcium phosphate.sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Furthermore, additives or excipients such as Gelucire®, Capryol®, Labrafil®, Labrasol®,

Lauroglycol®, Plurol®, Peceol®, Transcutol® and the like may be used.

Further, the pharmaceutical composition may be incorporated into a skin patch for delivery of the drug directly onto the skin.

It will be appreciated that the actual dosages of the agents of this invention will vary according tothe particular agent being used, the particular composition formulated, the mode of administration, and the particular site, host, and disease being treated. Those skilled in the art using conventional dosage determination tests in view of the experimental data for a given compound may ascertain optimal dosages for a given set of conditions. For oral administration, an exemplary daily dose generally employed will be from about 0.001 to about 1000 mg/kg of body weight, with courses of treatment repeated at appropriate intervals.

Furthermore, the pharmaceutically acceptable formulations of the present invention may contain acompound of the present invention, or a salt or solvate thereof, in an amount of about 10 mg to about 2000 mg, or from about 10 mg to about 1500 mg, or from about 10 mg to about 1000 mg, or from about 10 mg to about 750 mg, or from about 10 mg to about 500 mg, or from about 25 mg to about 500 mg, or from about 50 to about 500 mg, or from about 100 mg to about 500 mg.

Additionally, the pharmaceutically acceptable formulations of the present invention may contain acompound of the present invention, or a salt or solvate thereof, in an amount from about 0.5 w/w% to about 95 w/w%, or from about 1 w/w% to about 95 w/w%, or from about 1 w/w% to about 75 w/w%, or from about 5 w/w% to about 75 w/w%, or from about 10 w/w% to about 75 w/w%, or from about 10 w/w% to about 50 w/w%.

The compounds of the present invention, or salts or solvates thereof, may be administered to amammal, such as a human, suffering from a condition or disease mediated by an enveloped virus, either alone or as part of a pharmaceutically acceptable formulation, once a day, twice a day, three times a day, four times a day, or even more frequently.

The compounds of the present invention, or salts or solvates thereof, may be administered to humans or mammals suffering from a condition or disease mediated by a filovirus, arenavirus, or other enveloped virus in combination with at least one other agent used for treatment, alone or as part of a pharmaceutically acceptable formulation, once a day, twice a day, three times a day, four times a day, or even more frequently.

Those of ordinary skill in the art will understand that with respect to the compounds of the presentinvention, the particular pharmaceutical formulation, the dosage, and the number of doses given per day to humans or mammals requiring such treatment, are all choices within the knowledge of one of ordinary skill in theart and can be determined without undue experimentation.

Combination Therapy

Compounds of Structural Formula I of the invention may be combined with other therapeutic agents. The inhibitor and other therapeutic agent may be administered simultaneously or sequentially. When the other therapeutic agents are administered simultaneously they can be administered in the same or separate formulations, but are administered at the same time. The other therapeutic agents are administered sequentially with one another and with the inhibitors, when the administration of the other therapeutic agents and the inhibitors is temporally separated. The separation in time between the administration of these compounds may be a matter of minutes or it may be longer. Other therapeutic agents include but are not limited to anti-viral vaccines and anti-viral agents. In some instances the inhibitors are administered with multiple therapeutic agents, i.e., 2, 3, 4 or even more different anti-viral agents.

An anti-viral vaccine is a formulation composed of one or more viral antigens and one or more adjuvants. The viral antigens include proteins or fragments thereof as well as whole killed virus.

Adjuvants are well known to those of skill in the art.

Antiviral agents are compounds, which prevent infection of cells by viruses or replication of the virus within the cell. There are many fewer antiviral drugs than antibacterial drugs because viruses are more dependent on host cell factors than bacteria. There are several stages within the process of viral infection, which can be blocked or inhibited by antiviral agents. These stages include, attachment of the virus to the host cell (immunoglobulin or binding peptides), membrane penetration inhibitors, e.g. T-20, uncoating of the virus (e.g. amantadine), synthesis or translation of viral mRNA (e.g.

interferon), replication of viral RNA or DNA (e.g. nucleotide analogues), maturation of new virus proteins (e.g. pro- tease inhibitors), and budding and release of the virus.

Nucleotide analogues are synthetic compounds which are similar to nucleotides, but which have an incomplete or abnormal deoxyribose or ribose group. Once the nucleotide analogues are in the cell, they are phosphorylated, producing the triphosphate formed which competes with normal nucleotides for incorporation into the viral DNA or RNA. Once the triphosphate form of the nucleotide analogue is incorporated into the growing nucleic acid chain, it causes irreversible association with the viral polymerase and thus chain termination. Nucleotide analogues include, but are not limited to, acyclovir (used for the treatment of herpes simplex virus and varicella-zoster virus), gancyclovir (useful for the treatment of cytomegalovirus), idoxuridine, ribavirin (useful for the treatment of respiratory syncitial virus), didcoxyi- nosine, dideoxycytidine, zidovudine (azidothymidine), imiquimod, resimiquimod, favirpiravir, BCX4430, and GS-5374 or their analogues. The interferons are cytokines which are secreted by virus-infected cells as well as immune cells. The interferons function by binding to specific receptors on cells adjacent to the infected cells, causing the change in the cell which protects it from infection by the virus. - and β-interferon also induce the expression of Class I and Class II MHC molecules on the surface of infected cells, resulting in increased antigen presentation for host immune cell recognition, a- and β-interferons are available as recombinant proteins and have been used for the treatment of chronic hepatitis B and C infection. At the dosages that are effective for anti-viral therapy, interferons may have severe side effects such as fever, malaise and weight loss.

Anti-viral agents, which may be useful in combination with Structural Formula I of the invention, include but are not limited to immunoglobulins, amantadine, interferons, nucleotide analogues, small interfering RNAs (siRNAs) and other protease inhibitors (other than the papain-like cysteine protease inhibitors-although combinations of papain-like cysteine protease inhibitors are also useful). Specific examples of anti-viral agents include but are not limited to Acemannan; Acyclovir; Acyclovir Sodium; Adefovir; Alovudine; Alvircept Sudotox; Amantadine Hydro- chloride; Aranotin; Arildone; Atevirdine Mesylate; AVI-7537: Avridine; Cidofovir; Cipamfylline; Cytarabine Hydrochloride; Delavirdine Mesylate; Desciclovir; Didanosine; Disoxaril; Edoxudine; Enviradene; Enviroxime;

Famciclovir; Famotine Hydrochloride; Favipiravir; Fiacitabine; Fialuridine; Fosarilate; Foscar- net SodillH I; Fosfonet Sodium; Ganciclovir; Ganciclovir Sodium; Idoxuridine; Kethoxal; Lamivudinc; Lobucavir; Memotine Hydrochloride; Methisazone; Nevirapine; Penci- clovir; Pirodavir; Ribavirin; Rimantadine Hydrochloride; Saquinavir Mesylate; Somantadine Hydrochloride; Sorivudine; Statolon; Stavudine; Tilorone Hydrochloride; TKM Ebola; Triazavirin; Trifluri- dine; Valacyclovir Hydrochloride; Vidarabine; Vidarabine Phosphate; Vidarabine SodillH I Phosphate; Viroxime; Zalcit- abine;

Zidovudine; Zinviroxime; and ZMapp.

Immunoglobulin therapy is used for the prevention of viral infection. Immunoglobulin therapy for viral infections is different than bacterial infections, because rather than being antigen-specific, the immunoglobulin therapy functions by binding to extracellular virions and preventing them from attaching to and entering cells which are susceptible to the viral infection. The therapy is useful for the prevention of viral infection for the period of time that the antibodies are present in the host. In general there are two types of immunoglobulin therapies, normal immunoglobulin therapy and hyper- immunoglobulin therapy. Normal immune globulin therapy utilizes an antibody product which is prepared from the serum of normal blood donors and pooled. This pooled product contains low titers of antibody to a wide range of human viruses, such as hepatitis A, parvovirus, enterovirus (especially in neonates). Hyper-immune globulin therapy utilizes antibodies which are prepared from the serum of individuals who have high titers of an antibody to a particular virus. Those antibodies are then used against a specific virus. Another type of immunoglobulin therapy is active immunization. This involves the administration of antibodies or antibody fragments to viral surface proteins.

In the following Preparations and Examples, "Ac" means acetyl, "Me" means methyl, "Et" means ethyl, "Ph" means phenyl, "Py" means pyridine, "BOC", "Boc" or "boc" means N-tert- butoxycarbonyl, "Ns" means 2-Nitrophenylsulfonyl, " CMMP" means (cyanomethylene) trimethyl phosphorane", DCM" (CH 2 CI 2 ) means dichloromethane or methylene chloride, "DCE" means dichloroethane or ethylene chloride, "DCM" means dichloromethane, "DIAD" means diisopropylazadicarboxylate, "DIPEA" or "DIEA" means diisopropyl ethyl amine, "DMA" means N,N- dimethylacetamide, "DMAP" means 4-dimethylaminopyridine, "DME" means 1 ,2-dimethoxyethane, "DMF" means Ν,Ν-dimethyl formamide, "DMSO" means dimethylsulfoxide, "DPPA" means diphenylphosphorylazide, "DPPP"means 1 ,3-bis(diphenylphosphino)propane, "EDCI" means 3- (ethyliminomethyleneamino)-/V,/V-dimethylpropan-1 -amine, "EtOAc" means ethyl acetate, "HATU" means 1 -[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5-b]pyridinium 3-oxid

hexafluorophosphate, "HOAt" means 1 -hydroxy-7 azabenzotriazole, "HOAc" means acetic acid, "IPA" means isopropyl alcohol, "LDA" means lithium diisopropylamide, "NMP" means 1 -methyl 2- pyrrolidinone, "TEA" means triethyl amine, "TFA" means trifluoroacetic acid, "TOSMIC" means toluenesulfonylmethyl isocyanide, , "MgS0 4 " means magnesium sulphate, "NaHMDS" or "NHMDS" means sodium hexamethyldisilazide, "Na 2 S0 4 " means sodium sulphate, "MeOH" means methanol, "Et 2 0" means diethyl ether, "EtOH" means ethanol, "H 2 0" means water, "HCI" means hydrochloric acid, "POCI 3 " means phosphorus oxychloride,"SOCI 2 " means thionylchloride, "K 2 C0 3 " means potassium carbonate, "THF" means tetrahydrofuran, "DBU" means 1 ,8-diazabicyclo[5.4.0]undec-7- ene, "LAH" means lithium aluminium hydride, "LiHMDS" or "LHMDS" means lithium

hexamethyldisilazide, "TBABr" means tetra butyl ammonium bromide, "TBME" or"MTBE" means tert- butyl methyl ether, "TMS" means trimethylsilyl , "PMHS" means polymethylhydrosiloxane,"MCPBA" means 3-chloroperoxy benzoic acid, "N" means Normal, "M"means molar, "ml_" means millilitre, "mmol" means millimoles, "μηιοΓ means micromoles, "eq." meansequivalent, "°C" means degrees

Celsius, "Pa" means pascals, "Xanthphos" means 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene, "rt" means room temperature.

Methods of Preparation.

Compounds of the present invention may be prepared using the reaction routes and synthetic schemes described below, employing the techniques available in the art using starting materials that are readily available. The preparation of certain embodiments of the present invention is described in detail in the following examples, but those of ordinary skill in the art will recognize that the

preparations described may be readily adapted to prepare other embodiments of the present invention. For example, the synthesis of non-exemplified compounds according to the invention may be performed by modifications apparent to those skilled in the art, e.g. by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. Alternatively, other reactions referred to herein or known in the art will be recognized as having adaptability for preparing other compounds of the invention.

In one general synthetic process, compounds of the Structural Formula I where Y is a bond or

O

CR 4 R 5 and X is C-A NR 7a R 7b t represented by Formula l-a can be prepared according to Scheme 1 by reacting carboxylic acid 1-1 with amine NHR 7a R 7b in the presence of a coupling reagent such as EDCI or HATU and a base such as DIEA or triethylamine in a solvent such as DMF or dichloroethane to provide the desired product of Formula l-a. Alternatively, carboxylic acid 1-1 can react with SOCI 2 to form acid chloride 1 -2 which can react with amine NHR 7a R 7b in presence of a base such as DIEA or triethylamine in a solvent such as DMF or dichloroethane to form amide l-a. Reduction of the amide I- a with a reducing agent such as lithium alumuminum hydride or similar reagents known to those skilled in the arts in a solvent such as THF or dietyl ether can provide a compound of general structure I where Y is a bond or CR 4 R 5 and X is -C-A-CH 2 NR 7a R 7b represented by Formula l-b.

Scheme 1

1-2

In another general synthetic process, compounds of the Structural Formula I where Y is a

bond or CR 4 R 5 and X is , represented by Formula l-c can be prepared according to Scheme 2 by reacting carboxylic acid 2-1 with amine 2-2 in the presence of a coupling reagent such as EDCI or HATU and a base such as trimethylamine in a solvent such as DMF or dichloromethane to provide the desired product of Formula l-c. Alternatively, carboxylic acid 2-1 can react with SOCI 2 to form acid chloride 2-3 which can react with amine 2-2 in presence of a base such as DIEA or triethylamine in a solvent such as DMF or dichloroethane to form amide l-c

In another general synthetic process, compounds of the Structural Formula I where Y is a bond or CR 4 R 5 and X is C-A" S s

"NR 7a R 7b or C-A % s

" ° "NR 7a R 7b , represented by Formula l-d or l-e can be prepared according to Scheme 3 by reacting sulfinyl chloride 3-1 with amine NHR 7a R 7b in the presence a base such as triethylamine in a solvent such as DMF or dichloromethane to provide the desired product of Formula l-d which can be oxidized with an oxidizing agent such as MCPBA to desired product of formula l-e. Alternatively, sulfonyl chloride 3-2 can be reacted with amine

NHR 7a R7 b in the presence of a base such as triethylamine in a solvent such as DMF or

dichloromethane to provide the desired product of Formula l-e.

In another general synthetic process, compounds of the Structural Formula I where X is CR 5 ,

O

and Y is -CR 4 -A NR 7a R 7b t represented by Formula l-f can be prepared according to Scheme 4 by reacting carboxylic acid 4-1 with amine NHR 7a R 7b in the presence of a coupling reagent such as EDCI or HATU and a base such as DIEA or triethylamine in a solvent such as DMF or dichloroethane to provide the desired product of Formula l-f. Alternatively, carboxylic acid 4-1 can react with SOCI 2 to form acid chloride 4-2 which can react with amine NHR 7a R 7b in presence of a base such as DIEA or triethylamine in a solvent such as DMF or dichloroethane to form amide l-f. Reduction of the amide l-f with a reducing agent such as lithium alumuminum hydride or similar reagents known to those skilled in the arts in a solvent such as THF or dietyl ether can provide a compound of general structure I where X is CR 5 and Y is CR 4 -A-CH 2 NR 7a R 7b , represented by Formula l-g.

4-2

In another general synthetic process, compounds of the Structural Formula I where Y is a

R 11a bond or CR 4 R 5 , X is C-A-ZR 0 , Z is O, S, or NR 8 , and R 0 is R % 9a R 9b 11b , represented by Formula l-h can be prepared according to Scheme 5 by reacting 5-1 with a suitable hydroxy alkane containing a leaving group (LG) such as a halogen or a sulfonate 5-2 to provide 5-3. Treatment of 5-3 with triflic anhydride in a presence of a base such as triethylamine in a solvent such as THF or DCM can provide triflate 4-5 which can be treated with amine NHR a R b in a solvent such as THF or DCM to provide a compound of Formula l-h.

Scheme 5

In another general synthetic process, compounds of the Structural Formula I where Y is a

bond or CR 4 R 5 , X is C-A-ZR 10 , Z is O, S, or NR 8 , and , represented by

Formula l-i can be prepared according to Scheme 6 by reacting 6-1 with an alkyl oxirane or oxetane 6-2 containing a leaving group (LG) such as a halogen or a sulfonate in the presence of a base such as K 2 C0 3 in a solvent such as DMF to form oxirane or oxetane 6-3. Alternatively, when Z is oxygen, alcohol 6-1 can be treated with triflic anhydride in the presence of a base such as NEt 3 in a solvent such as DCM to form triflate 6-4 which in the presence of a base such sodium hydride in a solvent such as DMF can react with alcohol 6-5 to form 6-3. Treatment of 6-3 with amine NHR a R b in a solvent such as isopropanol can provide a compound of Formula l-i.

In another general synthetic process, compounds of the Structural Formula I where Y is a

bond or CR 4 R 5 , X is C-A-ZR 0 , Z is O, S, or NR 8 , and R 0 is , represented by Formula l-j can be prepared according to Scheme 7 by reacting 7-1 with an alkyl oxirane or oxetane 7-2 containing a leaving group (LG) such as a halogen or a sulfonate in the presence of a base such K 2 C0 3 in a solvent such as DMF to form oxirane or oxetane 7-3. Alternatively, when Z is oxygen, alcohol 7-1 can be treated with triflic anhydride in the presence of a base such as NEt 3 in a solvent such as DCM to form triflate 7-4 which in the presence of a base such as sodium hydride in a solvent such as DMF can react with alcohol 7-5 to form 7-3. Treatment of 7-3 with amine NHR a R b in a solvent such as isopropanol can provide a compound of Formula l-j.

Scheme 7

In another general synthetic process, compounds of the Structural Formula I where Y is a

bond or CR 4 R 5 , X is C-A-ZR 0 , Z is O, S, or NR 8 , and R 0 represented by

Formula l-k can be prepared according to Scheme 8 by reacting 8-1 with an alkyl oxirane or oxetane 8-2 containing a leaving group (LG) such as a halogen or a sulfonate in the presence of a base such K 2 C0 3 in a solvent such as DMF to form oxirane or oxetane 8-3. Alternatively, when Z is oxygen, alcohol 8-1 can be treated with triflic anhydride in the presence of a base such as NEt 3 in a solvent such as DCM to form triflate 8-4 which in the presence of a base such sodium hydride in a solvent such as DMF can react with alcohol 8-5 to form 8-3. Treatment of 8-3 with amine NHR a R b in a solvent such as isopropanol can provide a compound of Formula l-k.

8-4

In another general synthetic process, compounds of the Structural Formula I where Y is a

bond or CFCFC, X is C-A-D, A is a bond, and D is , represented by Formula l-l and l-m can be prepared according to Scheme 9 from carboxylic acid 9-1 via a Curtius rearrangement using an azide reagent such as diphenylphosphorylazide to form amine 9-2 which can react with an aldehyde in the presence of a reducing agent such as NaCNBH 3 in a solvent such as DCM to form amine 9-3. Reaction of amine with a carboxylic acid R 0a CO 2 H the presence of a coupling reagent such as EDCI or HATU and a base such as triethylamine in a solvent such as DMF or dichloromethane can provide the desired product of Formula l-l. Reaction of amine 9-3 with sulfonyl chloride R 0a SO 2 CI in the presence of a base such as triethylamine in a solvent such as

dichloromethane can provide the desired product of Formula l-m. Scheme 9

l-m

In another general synthetic process, compounds of the Structural Formula I where Y is a

H N NR 7a R 7b

bond or CR R , X is C-A-D, A is a bond, and D is o , represented by Formula l-n can be prepared according to Scheme 10 from carboxylic acid 10-1 via a Curtius rearrangement using an azide reagent such as diphenylphosphorylazide to form amine 9-2 and reacting the intermediate isocyanate with an amine NHR 7a R 7b to form the desired product of Formula l-n.

Preparation of Intermediates fe/f-Butyl 7-benzyl-2,7-diazaspiro[4.4]nonane-2-carboxylate

A mixture of fe/f-butyl 2,7-diazaspiro[4.4]nonane-2-carboxylate (0.15 g, 0.66 mmol) and benzaldehyde (0.07 g, 0.66 mmol) in 3 ml_ 1 ,2-dichloroethane was stirred at rt for 0.5 h. Sodium triacetoxy borohydride (0.35 g, 1 .65 mmol) was then added and the reaction mixture was stirred at rt for 12 h. The reaction mixture was then diluted with CH 2 CI 2 and washed with saturated aq. NaHC0 3 solution. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by Si0 2 column chromatography (EtOAc, then CH 2 CI 2 / CH 3 OH 40:1) to give 0.084 g (40%) of the title compound as a colorless oil. LC/MS m/z: 217.28 (M-Boc+H) + , 261 .29 (M-tBu+H) + , 302.31 (M- CH 3 +H) + , 317.33 (M+H) + , 358.31 (M+H+CH 3 CN) + .

The following intermediates were prepared in the same manner as described above for fe/f-butyl 7- benzyl-2,7-diazaspiro[4.4]nonane-2-carboxylate using the appropriate amine and the appropriate aldehyde as starting materials.

Starting materials Intermediate/Name Analytical Data fe/f-butyl 2,7-diazaspiro LC/MS m/z: 481 .36 (M+H) + [4.4]nonane-2-carboxylate

methyl 4-(2-formylphenoxy

fe/f-butyl 7-[(2-{[4-(methoxycarbonyl)

methyl) benzoate

phenyl] methoxy}phenyl)methyl]-2,7- diazaspiro[4.4]nonane-2-carboxylate

fe/f-butyl piperazine-1 - LC/MS m/z: 441 .24 (M+H) + carboxylate

methyl 4-(2-formyl

fe/f-butyl 4-[(2-{[4-(methoxycarbonyl)

phenoxymethyl) benzoate

phenyl] ethoxy}phenyl)methyl]

piperazine-1 -carboxylate fe/f-butyl A/-(piperidin-4-yl) m LC/MS m/z: 455.28 (M+H) + carbamate

BocHN^ ^

methyl 4-(2-formylphenoxy

methyl 4-{2-[(4-{[(fe/f- methyl) benzoate

butoxy)carbonyl] amino}piperidin-1 - yl)methyl]phenoxy methyl}benzoate

fe/f-butyl 2,5-diazabicyclo LC/MS m/z: 453.28 (M+H) +

[2.2.1 ]heptane-2- carboxylate

methyl 4-(2-formylphenoxy fe/f-butyl 5-[(2-{[4-(methoxycarbonyl)

methyl) benzoate phenyl] methoxy}phenyl)methyl]-2,5- diazabicyclo[2.2.1 ]heptane-2- carboxylate

fe/f-butyl 2,6- LC/MS m/z: 233.23 (M- diazaspiro[3.3]heptane-2- tBu+H) + , 274.23 (M-

carboxylate CH 3 +H) + , 289.28 (M+H) + , fe/f-butyl 6-benzyl-2,6- benzaldehyde 330.32 (M+H+CH 3 CN) + diazaspiro[3.3]heptane-2-carboxylate fe/f-butyl 2,6- LC/MS m/z: 247.2 (M- diazaspiro[3.4]octane-2- tBu+H) + , 288.22 (M- carboxylate fe/f-butyl 6-benzyl-2,6- CH 3 +H) + , 303.33 (M+H) + , benzaldehyde diazaspiro[3.4]octane-2-carboxylate 344.38 (M+H+CH 3 CN) + fe/f-Butyl-/V-(2-{7-benzyl-2,7-diazaspiro[4.4]nonan-2-yl}-2- oxoethyl)carbamate

Stepl : 2-benzyl-2,7-diazaspiro[4.4]nonanedihydrochloride

To a solution of fe/f-butyl 7-benzyl-2,7-diazaspiro[4.4]nonane-2-carboxylate (0.08 g, 0.25 mmol) in 1 .5 mL CH 3 OH was added 0.75 mL cone. HCI. The reaction mixture was stirred at rt for 12 h, then concentrated in vacuoto give 0.073 g (100%) of the product as a viscous oil, which was used in the next step without further purification. LC/MS m/z: 258.09 (M+H+CH 3 CN) + .

To a mixture of 2-benzyl-2,7-diazaspiro[4.4]nonanedihydrochloride (58 mg, 0.2 mmol) in 2 ml_ anhydrous CH 2 CI 2 was added /V,/V-diisopropylethylamine (1 16.6 mg, 0.9 mmol), 2-{[(fe/f- butoxy)carbonyl]amino}acetic acid (35 mg, 0.2 mmol), EDC hydrochloride (50 mg, 0.26 mmol), and 1 - hydroxy-7-azabenzotriazole (HOAt, 30 mg, 0.22 mmol). The reaction mixture was stirred at rt for 12 h, then diluted with CH 2 CI 2 and washed with saturated aq. NaHC0 3 solution. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by Si0 2 column chromatography (EtOAc, then CH 2 CI 2 / CH 3 OH from 40:1 to 10:1) to give 45 mg (60%) of the product as a colorless oil. LC/MS m/z: 21 AAA (M-Boc+H) + , 318.35 (M-tBu+H) + , 374.36 (M+H) + .

The following intermediates 1-1 to I-8 were prepared in the same manner as described for the preparation of fe/f-butyl A/-(2-{7-benzyl-2,7-diazaspiro[4.4]nonan-2-yl}-2-oxoethyl)ca rbamate using the appropriate precursor.

ethyladamantane-1 -carboxylic acid.

The title compound was obtained by bromination of 3,5-dimethyladamantane-1 -carboxylic acid using the procedure described in patent application US 2013/0096120, published April 18, 2013.

3-Bromo-5-ethyladamantane-1 -carboxylic acid.

The title compound was prepared from 3-ethyladamantane-1 -carboxylic acid in the same manner as described for 3-bromo-5,7-dimethyladamantane-1 -carboxylic acid. 3,5-Dimethyl-7-phenyladamantane-1 -carboxylic acid.

3-Bromo-5,7-dimethyladamantane-1 -carboxylic acid (0.033 g, 0.1 1 mmol) was added to a mixture of AICI 3 (0.018 g, 0.14 mmol) in 1 mL anhydrous benzene. The reaction mixture was heated to reflux for 6 h, then diluted with EtOAc. The organic phase was washed with 0.5 M aq. HCI and brine, dried over Na 2 S0 4 , filtered, and concentrated in vacuo to give 0.03 g (91 %) of the product as white solid, which was used in the next step without further purification.

3-Ethyl-5-phenyladamantane-1 -carboxylic acid.

The title compound was prepared from 3-bromo-5-ethyladamantane-1 -carboxylic acid and benzene in the same manner as described for 3,5-dimethyl-7-phenyladamantane-1 -carboxylic acid. enyl)adamantane-1 -carboxylic acid.

The title compound was prepared from 3-bromoadamantane-1 -carboxylic acid and chlorobenzene in the same manner as described for 3,5-dimethyl-7-phenyladamantane-1 -carboxylic acid. enyl)adamantane-1 -carboxylic acid.

The title compound was prepared from 3-bromoadamantane-1 -carboxylic acid and fluorobenzene i the same manner as described for 3,5-dimethyl-7-phenyladamantane-1 -carboxylic acid.

1 -Phenyltricyclo[3.3.1 .0 3,7 ]nonane-3-carboxylic acid. The title compound was prepared following the procedure described in patent application US2013/0345127, published Dec 26, 2013.

5-Phenyladamantane-2-carboxylic acid.

Step 1 : 5-phenyladamantan-2-one

To 5-hydroxyadamantan-2-one (5 g, 150 mmol) was added benzene (20 ml_) and triflic acid (3 ml_). The heterogenous mixture was heated to reflux, at which time all starting materials dissolved, and reflux was continued overnight. The mixture was cooled to room temperature and partitioned between water and fe/f-butylmethyl ether (50 ml_), and the organic phase was washed 3 times with water, followed by saturated NaHC0 3 and brine. The organics were dried over Na 2 S0 4 and evaporated to give 4.7g of crude product. Purification with flash chromatography on silica gel using 8:2

hexanes:ethyl acetate eluent gave 3.7 g of pure product as a white solid. H NMR (300MHz, CDCI 3 ) δ: 7.41 -7.32 (m, 4H), 7.27-7.21 (m, 1 H), 2.70 (br:s, 2H), 2.37-2.03 (m, 1 1 H).

Step 2: c/ ' s-5-phenyladamantane-2-carbonitrile and frans-5-phenyladamantane-2-carbonitrile

To 5-phenyladamantan-2-one (1 .1 g, 4.87 mmol) in a flask with strong stirrer was added

dimethoxyethane (20 ml_), ethanol (7 ml_), and p-toluenesulfonylmethylisocyanide (1 .23 g, 6.33 mmol). The mixture was cooled to 0 °C, and potassium fe/f-butoxide (1 .31 g, 1 1 .7 mmol) was added in 3 portions (mild exotherm). The reaction mixture was heated to 35 °C overnight and then cooled to room temperature and filtered to remove potassium tosylate. The filtrate was partitioned between water and fe/f-butylmethyl ether and washed three times with water, then brine, dried (Na 2 S0 4 ), and evaporated. The crude residue was purified via flash chromatography on silica gel using 95:5 hexanes:ethyl acetate eluent. Purification gave 200 mg and 234 mg of the less and more polar isomers, respectively, and 410 mg of mixed fractions containing co-eluted meso-isomers A and B. Top isomer on TLC, configuration unknown: H NMR (500MHz, CDCI 3 ) δ: 7.38-7.30 (m, 4H), 7.20 (t, 1 H), 2.93 (s, 1 H), 2.39 (s, 2H), 2.25-2.15 (m, 3H), 2.07 (d, 2H), 1 .94 (t, 4H), 1 .77 (d, 2H). Bottom isomer on TLC, configuration unknown: H NMR (500MHz, CDCI 3 ) δ: 7.38-7.31 (m, 4H), 7.20 (t, 1 H), 2.94 (s, 1 H), 2.39 (s, 2H), 2.35 (d, 2H), 2.15 (s, 1 H), 1 .97 (s, 1 H), 1 .90 (d, 4H), 1 .78 (d, 2H).

Step 3: 5-phenyladamantane-2-carboxylic acid In two separate reactions, 140 mg of the top nitrile on TLC (absolute configuration unknown) and 230 mg of the bottom nitrile on TLC (absolute configuration unknown) were combined with 5 mL of 33% HBr/glacial acetic acid solution. Each reaction was brought to reflux until TLC indicated that the nitriles had hydrolyzed to the primary amide, and the maximum amount of water that does not cause the amide to precipitate was titrated into the reaction mixtures through their condensers. The separate reactions were refluxed overnight and then cooled to room temperature. On cooling, both reactions produced precipitates that were filtered off into sintered glass funnels, washed 3 times with 1 M HCI, and dried on a rotary evaporator to yield fine white crystals. The top nitrile on TLC yielded 105 mg, while the bottom nitrile produced 185 mg. 3 C NMR of both reaction products confirms that both nitriles hydrolyze to identical carboxylic acids, presumably the most thermodynamically stable product. The absolute cis / trans configuration of the single carboxylic acid product is unknown. HPLC and TLC also indicate the formation of a single, identical carboxylic acid isomer from both reactions. 3 C NMR (125 MHz, CDCI 3 ) δ: 180.35, 150.64, 128.45, 125.96, 125.01 , 49.04, 48.76, 43.76, 43.33, 39.16, 37.43, 35.83, 32.96, 30.40, 28.41

3-(3-Methylphenyl)adamantane-1 -carboxylic acid

Step 1 : 3-(2-hydroxy-5-methylphenyl)adamantane-1 -carboxylic acid

4-Methylphenol (0.25 g, 2.3 mmol) was added to a solution of 3-hydroxyadamantane-1 -carboxylic acid (0.5 g, 2.55 mmol) in methanesulfonic acid (2.3 mL) heated to 40 °C under N 2 atmosphere (G. Ogawa, M. A. Tius, H. Zhou, S. P. Nikas, A. Halikhedkar, S. Mallipeddi, and A. Makriyannis, J. Med. Chem. 2015, 58, 3104 - 31 16). The reaction mixture was stirred at 50 °C for 3 h, then cooled to rt, diluted with CH 2 CI 2 , poured onto ice-water. The organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The organic layers were combined, washed with brine, dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by Si0 2 column chromatography (hexanes / EtOAc from 7:3 to 1 :1) to give 0.34 g (47%) of the product as a colorless oil. H NMR (500 MHz, CDCI 3 ) δ 6.99 (s, 1 H), 6.86 (dd, 1 H), 6.53 (d, 1 H), 2.28 (br. s, 2H), 2.26 (s, 3H), 2.23 - 2.19 (m, 2H), 2.13 - 2.06 (m, 4H), 1 .97 - 1 .91 (m, 4H), 1 .77 - 1 .70 (m, 2H).

Step 2: methyl 3-(2-hydroxy-5-methylphenyl)adamantane-1 -carboxylate

TMSCHN 2 (0.51 mL of 2 M in hexanes, 1 .02 mmol) was added to a mixture of 3-(2-hydroxy-5- methylphenyl)adamantane-1 -carboxylic acid (0.18 g, 0.63 mmol) in benzene (2.5 mL) and MeOH (0.6 mL) at 10 °C. The reaction mixture was allowed to warm to rt for 1 h, then concentrated in vacuo to give the product as a colorless oil (0.19g, 100%), which was used in the next step without further purification.

Step 3: methyl 3-{5-methyl-2-[(trifluoromethane)sulfonyloxy]phenyl}adamanta ne-1 -carboxylate

To a mixture of methyl 3-(2-hydroxy-5-methylphenyl)adamantane-1 -carboxylate (0.15 g, 0.5 mmol), pyridine (0.12 g, 1 .5 mmol) and DMAP (0.6 mg, 0.005 mmol) in CH 2 CI 2 (2 mL) cooled to 0 °C was added Tf 2 0 (0.17 g, 0.6 mmol) in CH 2 CI 2 (0.12mL) dropwise. The reaction mixture was brought to rt gradually and stirred for 3 h, then diluted with CH 2 CI 2 and washed quickly with water. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by flash chromatography on Si0 2 (hexanes / EtOAc from 8:1 to 7:1) to give 0.1 1 g (52%) of the title compound as a colorless oil.

Step 4: methyl 3-(3-methylphenyl)adamantane-1 -carboxylate

To a solution of methyl 3-{5-methyl-2-[(trifluoromethane)sulfonyloxy]phenyl}adamanta ne-1 - carboxylate (0.1 1 g, 0.25 mmol) in dry DMF (1 mL) were added PdCI 2 (PPh 3 ) 2 (9 mg, 0.013 mmol), dppp (10 mg, 0.024 mmol), n-Bu 3 N (0.23 g, 1 .27 mmol), HCOOH (0.03 g, 0.63 mmol), and catalytic polymethylhydrosiloxane (PMHS) under N 2 atmosphere. The reaction mixture was stirred at 95 °C overnight, then cooled to rt, diluted with MTBE and 1 N HCI, stirred for 10 min, filtered through a plug of Celite. The organic layer was separated, and the aqueous layer was extracted with MTBE. The organic layers were combined, dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by flash chromatography on Si0 2 (hexanes / EtOAc from 9:1 to 8:1) to give 54 mg (75%) of the title compound as a colorless oil. H NMR (500 MHz, CDCI 3 ) δ 7.20 (t, 1 H), 7.18 - 7.14 (m, 2H), 7.00 (d, 1 H), 3.66 (s, 3H), 2.34 (s, 3H), 2.24 - 2.20 (m, 2H), 2.03 (br. s, 2H), 1 .95 - 1 .87 (m, 8H), 1 .75 - 1 .71 (m, 2H). Step 5: 3-(3-methylphenyl)adamantane-1 -carboxylic acid

To a solution of methyl 3-(3-methylphenyl)adamantane-1 -carboxylate (30 mg, 0.105 mmol) in CH 2 CI 2 (1 mL) cooled to 0 °C was added BBr 3 (0.2 mL of 1 M in CH 2 CI 2 , 0.2 mmol) dropwise under N 2 atmosphere. The reaction mixture was gradually warmed to rt over 3 h, diluted with CH 2 CI 2 , then quenched with ice. The organic layer was separated, and the aqueous layer was extracted with EtOAc. The organic layers were combined, dried over Na 2 S0 4 , filtered, and concentrated in vacuo to give 25 mg (90%) of the title compound as a white solid, which was used in the next step without further purification.

3-(Propan-2-yl)adamantane-1 -carboxylic acid

The title compound was prepared by isomerization of 2-(adamantan-1 -yl)propan-2-ol into 3-(propan-2- yl)adamantan-1 -ol in trifluoroacetic acid, followed by carboxylation of the formed alcohol using Koch- Haaf conditions (V. V. Kovalev, A. K. Rozov, and E. A. Shokova, Tetrahedron 1996, 52, 3983-3990).

3-Hydroxy-5-(propan-2-yl)adamantane-1 -carboxylic acid

To a suspension of 3-(propan-2-yl)adamantane-1 -carboxylic acid (90 mg, 0.4 mmol) in fuming nitric acid (0.25mL) cooled to 0 °C was added sulfuric acid (0.41 mL) (L. Wanka, C. Cabrele, M. Vanejews and P. R. Schreiner, Eur. J. Org. Chem. 2007, 1474-1490). The reaction mixture was stirred at 0 °C for 10 min. Then, oleum (0.14 mL, 25% S0 3 ) was added, and the reaction mixture was stirred for 1 h at 0 °C and 3 h at rt, poured onto ice. The aqueous layer was extracted with EtOAc. The organic layer was dried over Na 2 S0 4 , filtered, and concentrated in vacuo to give 90 mg (94%) of the crude product as a white solid, which was used in the next step without further purification.

3-phenyl-5-(propan-2-yl)adamantane-1 -carboxylic acid

The title compound was prepared from 3-hydroxy-5-(propan-2-yl)adamantane-1 -carboxylic acid and benzene in the same manner as described above for 5-phenyladamantan-2-one. H NMR (500 MHz, CDCI 3 ) δ 7.39 - 7.35 (m, 2H), 7.34 - 7.29 (m, 2H), 7.19 (t, 1 H), 2.34 - 2.30 (m, 1 H), 2.01 (br. s, 2H), 1 .89 - 1 .81 (m, 4H), 1 .72 - 1 .65 (m, 2H), 1 .63 - 1 .58 (m, 2H), 1 .54 - 1 .48 (m, 2H), 1 .39 - 1 .34 (m, 1 H), 0.86 (d, 6H).

3-Ethoxyadamantane-1 -carboxylic acid

To a solution of 3-bromoadamantane-1 -carboxylic acid (0.2 g, 0.77 mmol) in dry ethanol (2 mL) was added silver (I) oxide (0.18 g, 0.77 mmol) (H. Stetter and J. Mayer, Chem. Ber. 1962, 95, 667-672). The reaction mixture was refluxed for 3 h, then cooled to rt, diluted with EtOAc. Solids were filtered off and washed with 5% AcOH in EtOAc. Volatiles were concentrated in vacuo to give 0.13 g (76%) of the title compound as a colorless oil, which was used in the next step without further purification.

1 ,3-Dimethyl 5-phenyladamantane-1 ,3-dicarboxylate

The title compound was prepared from 1 ,3-dimethyl 5-hydroxyadamantane-1 ,3-dicarboxylate and benzene in the same manner as described above for 5-phenyladamantan-2-one. H NMR (500 MHz, CDCI 3 ) δ 7.37 - 7.30 (m, 4H), 7.20 (t, 1 H), 3.67 (s, 6H), 2.38 - 2.35 (m, 1 H), 2.07 (br. s, 2H), 2.05 - 1 .99 (m, 4H), 1.92 - 1 .85 (m, 6H).

3-(Methoxycarbonyl)-5-phenyladamantane-1 -carboxylic acid

To a solution of 1 ,3-dimethyl 5-phenyladamantane-1 ,3-dicarboxylate (0.12 g, 0.37 mmol) in MeOH (1 mL) was added NaOH (15 mg, 0.37 mmol). The reaction mixture was stirred at 50 °C overnight, then concentrated in vacuo without heating. The residue was dissolved in water, acidified with 1 N HCI, and extracted with EtOAc. The organic layer was dried over Na 2 S0 4 , filtered, and concentrated in vacuo to give 80mg (68%) of the crude product as a colorless oil, which was used in the next step without further purification. (pyrrolidin-1 -yl)carbonyl]adamantane-1 -carboxylate

The title compound was prepared from 3-(methoxycarbonyl)-5-phenyladamantane-1 -carboxylic acid and pyrrolidine in the same manner as described for 2-benzyl-7-[(3-phenyladamantan-1 -yl)carbonyl]- 2,7-diazaspiro[4.4]nonane (example A1 , Step 2). LC/MS m/z: 368.32 (M+H) + , 735.54 (2M+H) + .

3-Phenyl-5-[(pyrrolidin-1 -yl)carbonyl]adamantane-1 -carboxylic acid.

The title compound was prepared from methyl 3-phenyl-5-[(pyrrolidin-1 -yl)carbonyl]adamantane-1 - carboxylate using excess of NaOH in the same manner as described above for 3-(methoxycarbonyl)- 5-phenyladamantane-1 -carboxylic acid.

Example A1 : 2-benzyl-7-[(3-phenyladamantan-1 -yl)carbonyl]-2,7-diazaspiro[4.4]nonane

Stepl : 2-benzyl-2,7-diazaspiro[4.4]nonanedihydrochloride

To a solution of fe/f-butyl 7-benzyl-2,7-diazaspiro[4.4]nonane-2-carboxylate (0.08 g, 0.25 mmol) in 1 .5 mL CH 3 OH was added 0.75 ml_ cone. HCI. The reaction mixture was stirred at rt for 12 h, then concentrated in vacuoto give 0.073 g (100%) of the product as a viscous oil, which was used in the next step without further purification. LC/MS m/z: 258.09 (M+H+CH 3 CN) + .

Step 2: 2-benzyl-7-[(3-phenyladamantan-1 -yl)carbonyl]-2,7-diazaspiro[4.4]nonane

To a mixture of 2-benzyl-2,7-diazaspiro[4.4]nonane dihydrochloride(12.5 mg, 0.043 mmol) in 1 ml_ anhydrous CH 2 CI 2 was added /V,/V-diisopropylethylamine (25 mg, 0.19 mmol), 3-phenyl-adamantane- 1 -carboxylic acid (1 1 mg, 0.043 mmol), EDC hydrochloride (1 1 mg, 0.057 mmol), and 1 -hydroxy-7- azabenzotriazole (HOAt, 6.5 mg, 0.048 mmol). The reaction mixture was stirred at rt for 12 h, then diluted with CH 2 CI 2 and washed with saturated aq. NaHC0 3 solution. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by Si0 2 column chromatography (EtOAc, then CH 2 CI 2 / CH 3 OH from 40:1 to 30:1) to give 10mg (51 %) of the product as a colorless oil. LC/MS m/z: 455.40 (M+H) + , 496.43 (M+H+CH 3 CN) + .

Examples A2 to A14 were prepared in the same manner as described for example A1 , Steps 1 and 2 starting from the appropriate Boc-protected amine and the appropriate carboxylic acid.

Examples A15 to A48 were prepared in the same manner as described for example A1 , Step 2 using the appropriate commercially available amine and carboxylic acid as starting materials.

Ex. Starting materials Product/Name Analytical Data

A2 ferf-butyl A/-(2-{7-benzyl- LC/MS m/z: 512.42 (M+H) + , 2,7-diazaspiro 553.47 (M+H+CH 3 CN) +

[4.4]nonan-2-yl}-2- 0

oxoethyl) carbamate A/-(2-{7-benzyl-2,7-diazaspiro

3-phenyl-adamantane-1 - [4.4] nonan-2-yl}-2-oxoethyl)- carboxylic acid 3-phenyladamantane-1 - carboxamide

A3 methyl 4-(2-{[4-(2-{[(ferf- LC/MS m/z: 636.48 (M+H) + butoxy)carbonyl]

amino}acetyl)piperazin- 1 yl]methyl}phenoxymeth

methyl 4-{2-[(4-{2-[(3-phenyl

yl) benzoate

adamantan-1 -yl)formamido]

3-phenyl-adamantane-1 - acetyl}piperazin-1 -yl)methyl]

carboxylic acid phenoxy methyl}benzoate

A4 methyl 4-(2-{[4-(2-{[(ferf- LC/MS m/z: 650.44 (M+H) + butoxy)carbonyl] S jp y U T

amino}acetyl)piperazin-

1 -yl]methyl}phenoxy methyl 4-{2-[(4-{2-[2-(3-phenyl

methyl) benzoate adamantan-1 -yl)acetamido]

2-(3-phenyladamantan- acetyl} piperazin-1 -yl)methyl]

1 -yl)acetic acid phenoxymethyl} benzoate A10 ferf-butyl 6-benzyl-2,6- LC/MS m/z: 441 .31 (M+H) + , diazaspiro[3.4]octane-2- 482.36 (M+H+CH 3 CN) + carboxylate

3-phenyl-adamantane-1 -

carboxylic acid 6-benzyl-2-[(3-phenyl

adamantan-1 -yl)carbonyl]-2, 6- diazaspiro [3.4]octane

A1 1 te/ -butyl A/-(2-{7-benzyl- LC/MS m/z: 526.45 (M+H) + 2J-diazaspiro [4.4]

nonan-2-yl}-2-oxoethyl)

carbamate V-(2-{7-benzyl-2,7-

2-(3-phenyladamantan- diazaspiro[4.4] nonan-2-yl}-2- 1 -yl)acetic acid oxoethyl)-2-(3-phenyl

adamantan-1 -yl)acetamide

A12 ferf-butyl Λ/-{[(1 -benzyl LC/MS m/z: 500.46 (M+H) piperidin-4-yl)carbamoyl]

methyl}carbamate

2-(3-phenyladamantan- Λ/-{[(1 -benzylpiperidin-4-yl)

1 -yl)acetic acid carbamoyl] methyl}-2-(3- phenyladamantan-1 -yl)

acetamide

A13 terf-butyl A/-({[(3R)-1 - LC/MS m/z: 486.43 (M+H) benzyl pyrrolidin-3- yl]carbamoyl}

methyl)carbamate A/-({[(3f?)-1 -benzylpyrrolidin-3-

2-(3-phenyladamantan- yl]carbamoyl}methyl)-2-(3- 1 -yl)acetic acid phenyladamantan-1 -yl)

acetamide

A14 I te/ -butyl A/-(2-{5-benzyl- LC/MS m/z: 498.39 (M+H) 2,5-diazabicyclo

[2.2.1 ]heptan-2-yl}-2-

oxoeth y I) ca rba m ate A/-(2-{5-benzyl-2,5-diaza

2-(3-phenyladamantan- bicycio [2.2.1 ] heptan-2-yl}-2- 1 -yl)acetic acid oxoethyl)-2-(3-phenyl

adamantan-1 -yl)acetamide A15 (3S)-1 -benzylpyrrolidin- LC/MS m/z: 415.33 (M+H) + 3-amine 456.41 (M+H+CH 3 CN) +

3-phenyl-adamantane-1 -

carboxylic acid A/-[(3S)-1 -benzylpyrrolidin-3- yl]-3-phenyladamantane-1 - carboxamide

A16 2-amino-1 -(4-benzyl LC/MS m/z: 472.33 (M+H) piperazin-1 -yl)ethan-1 - one dihydrochloride

A/-[2-(4-benzylpiperazin-1 -yl)-

3-phenyl-adamantane-1 - 2-oxoethyl]-3-phenyl

carboxylic acid

adamantane-1 -carboxamide

A17 2-amino-1 -(4-benzyl LC/MS m/z: 486.33 (M+H) piperazin-1 -yl)ethan-1 - one dihydrochloride

A/-[2-(4-benzylpiperazin-1 -yl)-

2-(3-phenyladamantan- 2-oxoethyl]-2-(3-phenyl

1 -yl)acetic acid

adamantan-1 -yl)acetamide

A18 2-amino-1 -(4-benzyl LC/MS m/z: 382.33 (M+H) piperazin-1 -yl)ethan-1 - pi

one dihydrochloride

A/-[2-(4-benzylpiperazin-1 -yl)- tricyclo[3.3.1 .0 ' Jnonane 2- oxoethyl]tricyclo [3.3.1 .0 3,7 ]

-3-carboxylic acid nonane-3-carboxamide

A19 (3R)-1 -benzylpyrrolidin- LC/MS m/z: 415.44 (M+H) 3-amine

3-phenyl-adamantane-1

carboxylic acid A/-[(3f?)-1 -benzylpyrrolidin-3- yl]-3-phenyladamantane-1 - carboxamide

2-benzyl-2,5-diaza LC/MS m/z: 427.36 (M+H) bicyclo [2.2.1 Jheptane 468.36 (M+H+CH 3 CN) + dihydrobromide

3-phenyl-adamantane-1

2-benzyl-5-[(3-phenyl

carboxylic acid

adamantan-1 -yl)carbonyl]-2, 5- diazabicyclo [2.2.1 ]heptane A21 1 -benzylpiperidin-4- LC/MS m/z: 339.38 (M+H) amine

tricyclo[3.3.1 .0 ' Jnonane Λ/-(1 -benzylpiperidin-4-yl)

-3-carboxylic acid tricyclo [3.3.1 .0 3,7 ]nonane-3- carboxamide

A22 benzyl(methyl)[2- LC/MS m/z: 417.35 (M+H)

(methylamino)ethyl]

amine

3-phenyl-adamantane-1 - A/-{2-[benzyl(methyl)amino]

carboxylic acid ethyl}- A/-methyl-3-phenyl

adamantane-1 -carboxamide

A23 1 -benzyl-/V-methyl LC/MS m/z: 443.39 (M+H) piperidin-4-amine

3-phenyl-adamantane-1

carboxylic acid Λ/-(1 -benzylpiperidin-4-yl)-/V- methyl-3-phenyl adamantane- 1 -carboxamide

A24 (3R)-1 -benzylpyrrolidin- LC/MS m/z: 429.26 (M+H) 3-amine

2-(3-phenyladamantan- 1 -yl)acetic acid

A/-[(3f?)-1 -benzylpyrrolidin-3- yl]-2-(3-phenyladamantan-1 -yl)

acetamide

A25 1 -methylpiperazine LC/MS m/z: 339.27 (M+H)

3-phenyl-adamantane-1

carboxylic acid

1 -methyl-4-[(3-phenyl

adamantan-1 -yl)carbonyl]

piperazine A26 1-methylpiperidin-4- H NMR (300 MHz, CDCI 3 ) δ amine 7.26- 7.38 (m, 4H) 7.14 -7.21

3-phenyl-adamantane-1 - (m, 1H) 5.67 (d, 1H) 3.83-4.01

Λ/-(1 -methylpiperidin-4-yl)-3- carboxylic acid (m, 1H) 3.22 (d,2H) 2.57 (s, phenyladamantane-1 - 4H) 2.25 (br. s., 2H) 1.98 (br. carboxamide s., 3H) 1.79- 1.95 (m, 10H)

1.72 (br. s.,2H); LC/MS m/z: 353.37 (M+H) +

A27 Λ/,1 -dimethylpiperidin-4- LC/MS m/z: 367.36 (M+H) + amine

3-phenyl-adamantane-1

A/-methyl-/V-(1 -methylpiperidin- carboxylic acid

4-yl)-3-phenyl adamantane-1 - carboxamide

A28 1-benzylpiperazine LC/MS m/z: 325.35 (M+H) tricyclo[3.3.1.(T'] o 366.41 (M+H+CH 3 CN) + nonane-3-carboxylic 1-benzyl-4-({tricyclo

acid [3.3.1.0 37 ]nonan-3- yl}carbonyl)piperazine

A29 1-benzylpiperazine LC/MS m/z: 415.34 (M+H)

3-phenyl-adamantane-1

carboxylic acid

1 -benzyl-4-[(3-phenyl

adamantan-1- yl)carbonyl]piperazine

A30 1-benzylpiperazine LC/MS m/z: 429.30 (M+H)

2-(3-phenyladamantan- 470.49 (M+H+CH 3 CN) + 1-yl)acetic acid

1 -(4-benzylpiperazin-1 -yl)-2-

(3-phenyladamantan-1- yl)ethan-1-one

A31 1-benzylpiperazine LC/MS m/z: 339.33 (M+H) adamantane-2- 380.28

carboxylic acid 1 -[(adamantan-2-yl)carbonyl]- 4-benzylpiperazine A38 (3R)-1 -methylpiperidin- LC/MS m/z: 353.33 (M+H) 3-amine

3-phenyl-adamantane-1 -

carboxylic acid

A/-[(3f?)-1 -methylpiperidin-3- yl]-3-phenyladamantane-1 - carboxamide

A39 1-methylazetidin-3- LC/MS m/z: 339.38 (M+H)

amine

2-(3-phenyladamantan- 1-yl)acetic acid

Λ/-(1 -methylazetidin-3-yl)-2-

(3-phenyladamantan-1- yl)acetamide

A40 (3R)-1 -methylpyrrolidin- LC/MS m/z: 353.34 (M+H)

3-amine

2-(3-phenyladamantan- 1-yl)acetic acid

A/-[(3f?)-1 -methylpyrrolidin-3- yl]-2-(3-phenyladamantan-1 - yl)acetamide

A41 (3R)-1 -methylpyrrolidin- 1 H NMR (300 MHz, CDCI 3 ) δ 7.38 3-amine -7.28 (m, 4H), 7.20-7.15 (m,

3-methyl-5-phenyl 1H), 6.66-6.56 (m, 1H), 4.68-

adamantane-1 - 4.58 (m, 1H), 3.22-3.17 (m, 1H),

3-methyl-/V-[(3R)-1 -methyl

carboxylic acid 2.89 (d, 1H), 2.64 (dd, 1H), 2.50 pyrrolidin-3-yl]-5-phenyl (s, 3H), 2.43 - 2.35 (m, 2H), 2.33 adamantane-1 -carboxamide -2.26 (m, 1H), 1.95-1.86 (m,

2H), 1.85- 1.72 (m, 5H), 1.64- 1.55 (m, 4H), 1.46 (br. s, 2H), 0.92 (s, 3H). LC/MS m/z: 353.3 (M+H) + , 394.43(M+H+CH 3 CN) +

1-methylazetidin-3- LC/MS m/z: 339.4 (M+H) + amine

3-methyl-5-

phenyladamantane-1 3-methyl-/V-(1 -methyl

carboxylic acid azetidin-3-yl)-5-phenyl

adamantane-1 -carboxamide A43 1 -[3-(pyrrolidin-1 - LC/MS m/z: 464.43 (M+H) yl)propyl]-1 ,4-diazepane

3-methyl-5-

phenyladamantane-1 -

1 -[(3-methyl-5-phenyl

carboxylic acid

adamantan-1 -yl) carbonyl]- 4-[3-(pyrrolidin-1 -yl)propyl]- 1 ,4-diazepane

A44 (3S)-1 -azabicyclo H NMR (500 MHz, MeOH-d4) δ

[2.2.2]octan-3-amine 7.39 (d, 2H), 7.30 (t, 2H), 7.15 (t,

3-methyl-5- 1 H), 4.07 (m, 1 H), 3.45 (t, 1 H), phenyladamantane-1 - 3.2-3.12 (m, 1 H), 3.12-2.98 (m, carboxylic acid A/-[(3S)-1 - 3H), 2.95-2.88 (m, 1 H), 2.31 -2.29 azabicyclo[2.2.2]octan-3-yl]- (m, 1 H), 2.1 -2.05 (m, 1 H), 1 .99- 3-methyl-5-phenyl 1 .78 (m, 9H), 1 .70-1 .58 (m, 5H), adamantane-1 -carboxamide 1 .53 (br:s, 2H), 0.97 (s, 3H).

LC/MS m/z: 379.42 (M+H) + , 420.30 (M+H+CH 3 CN)

A45 (3R)-1 -azabicyclo Ή NMR (300 MHz, CDCI 3 ) δ

[2.2.2]octan-3-amine 7.41 (d, 2H), 7.33 (t, 2H), 7.20 (t,

3-methyl-5- 1 H), 4.33 (br:s. 1 H), 3.85-3.66

phenyladamantane-1 - (m, 2H), 3.36 (t, 1 H), 3.26-2.88

A/-[(3f?)-1 -azabicyclo

carboxylic acid (m, 4H), 2.31 (br:s, 2H), 2.09- [2.2.2]octan-3-yl]-3-methyl- 1 .40 (m, 15H), 0.95 (s, 3H). 5-phenyl adamantane-1 - LC/MS m/z: 379.33 (M+H) + , carboxamide

420.35 (M+H+CH 3 CN)

A46 (2-aminoethyl) LC/MS m/z: 341 .37 (M+H) + dimethylamine

3-methyl-5-

phenyladamantane-1

A/-[2-(dimethylamino)ethyl]- carboxylic acid

3-methyl-5-phenyl

adamantane-1 -carboxamide

4-methylpiperazin-1 - LC/MS m/z: 368.40 (M+H) amine

3-methyl-5-

phenyladamantane-1

3-methyl-/V-(4-methyl

carboxylic acid

piperazin-1 -yl)-5-phenyl

adamantane-1 -carboxamide A48 2-(morpholin-4-yl)ethan- LC/MS m/z: 383.39 (M+H) + 1 -amine

3-methyl-5- phenyladamantane-1 - 3- umethyl-/V-[2-(morpholin-4- carboxylic acid yl)ethyl]-5-phenyl

adamantane-1 -carboxamide

Example A49: A/-Benzyl-1 -[(3-phenyladamantan-1 -yl)carbonyl]piperidin-4-amine

Step 1 : fe/f-butyl A/-{1 -[(3-phenyladamantan-1 -yl)carbonyl]piperidin-4-yl}carbamate

The title compound was prepared in the same manner as described for example A1 , Step 2 using tert- butyl A/-(piperidin-4-yl)carbamate instead of 2-benzyl-2,7-diazaspiro[4.4]nonane. LC/MS m/z: 383.31 (M-tBu+H) + , 439.34 (M+H) + , 877.74 (2M+H) + -benzyl-1 -[(3-phenyladamantan-1 -yl)carbonyl]piperidin-4-amine

To a solution of fe/f-butyl A/-{1 -[(3-phenyladamantan-1 -yl)carbonyl]piperidin-4-yl}carbamate (74mg, 0.17 mmol) in 1 mL CH 3 OH was added 0.5 mL cone. HCI. The reaction mixture was stirred at rt for 12 h, then concentrated in vacuoto give 64 mg (100%) of HCI salt as a viscous oil. To a mixture of the obtained HCI salt (24 mg, 0.064 mmohin 1 mL CH 2 CI 2 was added /V,/V-diisopropylethylamine (8.2 mg, 0.064 mmol) followed by benzaldehyde (6.8 mg, 0.064 mmol). The resulting mixture was stirred at rt for 1 h. Sodium triacetoxyborohydride (34 mg, 0.16mmol) was then added and the reaction mixture was stirred at rt for 12 h. The reaction mixture was then diluted with CH 2 CI 2 and washed with saturated aq. NaHC0 3 solution. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by Si0 2 column chromatography (EtOAc, then CH 2 CI 2 / CH 3 OH from 40:1 to 20:1) to give 6 mg (22%) of the title compound as a colorless oil. LC/MS m/z: 429.28 (M+H) + . Example A50: 2-(Adamantan-1 -yl)-1 -{7-benzyl-2,7-diazaspiro[4.4]nonan-2-yl}ethan-1 -one joc *2rHCIo

To a mixture of 2-benzyl-2,7-diazaspiro[4.4]nonane dihydrochloride (21 mg, 0.073 mmol) in 0.75 mL CH 2 CI 2 was added /V,/V-diisopropylethylamine (37 mg, 0.29 mmol), followed by 1 -adamantaneacetyl chloride (15 mg, 0.073 mmol). The reaction mixture was stirred at rt for 3 h, then diluted with CH 2 CI 2 and washed with saturated aq. NaHC0 3 solution. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by preparative HPLC. The obtained TFA salt was dissolved in methanol and filtered through an Agilent Stratopheres PL-HCO 3 ion exchange resin. The filtrate was concentrated to afford 16 mg (56%) of the product as a colorless oil. LC/MS m/z: 393.42 (M+H) + , 434.42 (M+H+CH 3 CN) + .

Examples A51 to A53 were prepared in the same manner as described above for 2-(adamantan-1 -yl)- 1 -{7-benzyl-2,7-diazaspiro[4.4]nonan-2-yl}ethan-1 -one using the appropriate commercially available amine and 1 -adamantaneacetyl chloride as starting materials.

Starting amines in Examples A54 to A59 were prepared from the appropriate Boc-protected amines in the same manner as described above for 2-benzyl-2,7-diazaspiro[4.4]nonane dihydrochloride (Example A1 , Step 1), and used in the next step without further purification.

Examples A54 to A59 were prepared in the same manner as described above for 2-(adamantan-1 -yl)- 1 -{7-benzyl-2,7-diazaspiro[4.4]nonan-2-yl}ethan-1 -one using the appropriate amine and 1 - adamantaneacetyl chloride as starting materials.

Ex. Starting amine Product/Name Analytical Data

A51 1 -benzylpiperidin-4- LC/MS m/z: amine 367.36 (M+H) +

2-(adamantan-1 -yl)-/V-(1 -benzylpiperidin-4- yl)acetamide

A52 (3R)-1 -benzylpyrrolidin-3- LC/MS m/z: amine 353.25 (M+H) +

2-(adamantan-1 -yl)-/V-[(3f?)-1 -benzyl

pyrrolidin-3-yl]acetamide

A53 2-benzyl-2,5-diazabicyclo LC/MS m/z:

[2.2.1 ]heptane 365.35 (M+H) + , dihydrobromide 406.35

2-(adamantan-1 -yl)-1 -{5-benzyl-2,5-diaza

(M+H+CH 3 CN) + bicyclo[2.2.1 ]heptan-2-yl}ethan-1 -one

A54 2-amino-/V-(1 -benzyl LC/MS m/z: piperidin-4-yl)acetamide 424.29 (M+H) + dihydrochloride 2-(adamantan-1 -yl)-/V-{[(1 -benzyl

piperidin-4-yl)carbamoyl] methyl}acetamide

A55 methyl 4-(2-{[4-(2-amino LC/MS m/z: acetamido)piperidin-1 - 588.45 (M+H) + yl]methyl}phenoxymethyl)

benzoate hydrochloride methyl 4-{2-[(4-{2-[2-(adamantan-1 - yl)acetamido]acetamido}piperidin-1 - yl)methyl]phenoxymethyl}benzoate

benzoate

Examples A60 to A1 15 were prepared in the same manner as described for example A1 , Steps 1 and 2 starting from the appropriate Boc-protected amine (Step 2) and the appropriate carboxylic acid followed by Boc-deprotecting as decribed for Step 1 .

Ex. Starting materials Product/Name Analytical Data

A60 fe/ -butyl piperazine-1 - LC/MS m/z: 325.24 (M+H) +

carboxylate

3-phenyl-adamantane- 1 -carboxylic acid

1 0 -[(3-phe ° nyladamantan-1 - yl)carbonyl]piperazine

7.37

(d, (br. -

A74 fe/f-butyl 4- LC/MS m/z: 355.31 (M+H) + aminopiperidine-1 - 396.30 (M+H+CH 3 CN) + carboxylate

3-(4-hydroxyphi

adamantane-1 -

3-(4-hydroxyphenyl)-/V- carboxylic acid

(piperidin-4-yl)adamantane-

1 -carboxamide

A75 fe/f-butyl 4-amino LC/MS m/z: 357.29 (M+H)

piperidine-1 - 398.45 (M+H+CH 3 CN) + carboxylate

3-(4-fluorophenyl)

adamantane-1 -

3-(4-fluorophenyl)-/V- carboxylic acid

(piperidin-4-yl)adamantane-

1 -carboxamide

A76 fe/f-butyl 4- LC/MS m/z: 384.30 (M+H) + , aminopiperidine-1 - 425.32 (M+H+CH 3 CN) + carboxylate

3-(4-nitrophenyl)

adamantane-1 - 3-(4-nitrophenyl)-/V- carboxylic acid (piperidin-4-yl)adamantane-

1 -carboxamide

I c/s-fe/f-butyl Λ/-(4- Ή NMR (500 MHz, MeOH-d 4 ) δ aminocyclohexyl) 7.37 (d, 2H), 7.28 (t, 2H), 7.16 (t, carbamate 1 H), 3.80 (m, 1 H), 3.04 (m, 1 H),

3-methyl-5-phenyl 2.29 (m 1 H), 1 .90 (t, 3H), 1 .82 (t,

adamantane-1 - 3H), 1 .75-1 .67 (m, 4H), 1 .66-1 .58 c/ ' s-/V-(4-aminocyclohexyl)-3- carboxylic acid (m, 8H), 1 .51 (br:s, 2H), 0.96 (s, methyl-5-phenyl

3H). LC/MS m/z: 367.43 (M+H) + , adamantane-1 -carboxamide

408.30 (M+H+CH 3 CN) + δ

7.40 δ

δ

.70 1.48

A1 14 trans-tert-buty\ Λ/-(4- LC/MS m/z: 450.42 (M+H) + aminocyclohexyl)carb

a mate

3-phenyl-5-[(pyrrolidin- irar)s-/V-(4-aminocyclo

1 -yl)carbonyl]

hexyl)-3-phenyl-5[(pyrrolidin- adamantane-1 - 1 -yl)carbonyl] adamantane- carboxylic acid

1 -carboxamide

A1 15 trans-tert-buty\ N-(4- LC/MS m/z: 41 1 .27 (M+H) + ,

aminocyclohexyl)carb 452.2 (M+H+CH 3 CN) + , 821 .63 a mate (2M+H) +

3-(methoxycarbonyl)- methyl 3-[frans-(4-amino

5-phenyladamantane- cyclohexyl)carbamoyl]-5- 1 -carboxylic acid

phenyladamantane-1 - carboxylate

Example A1 16: irar)s-/V-(4-aminocyclohexyl)-3-(hydroxymethyl)-5-phenyladam antane-1 -carboxamide

Step 1 : methyl 3-[frar)s-(4-{[(fe/ -butoxy)carbonyl]amino}cyclohexyl)carbamoyl]-5-phenyladamant ane- 1 -carboxylate

The title compound was prepared from 3-(methoxycarbonyl)-5-phenyladamantane-1 -carboxylic acid and trans-tert-buty\ A/-(4-aminocyclohexyl)carbamate in the same manner as described above for 2- benzyl-7-[(3-phenyladamantan-1 -yl)carbonyl]-2,7-diazaspiro[4.4]nonane (example A1 , Step 2). LC/MS m/z: 455.32 (M-iBu+H) + , 496.35 (M-Me+H) + , 51 1 .41 (M+H) + , 552.44 (M+H+CH 3 CN) + .

Step 2: fe/f-butyl frans-A/-{4-[3-(hydroxymethyl)-5-phenyladamantane-1 -amido]cyclohexyl}carbamate

To a solution of methyl 3-[frar)s-(4-{[(fe/ -butoxy)carbonyl]amino}cyclohexyl)carbamoyl]-5- phenyladamantane-1 -carboxylate (15 mg, 0.03 mmol) in THF (0.5 ml_) was added dropwise 2.4 M solution of UAIH 4 in THF (0.012 ml_, 0.03 mmol) at 0 °C. The reaction mixture was stirred at ambient temperature for 2h, then cooled in an ice-bath, and quenched with saturated aqueous Na 2 S0 4 solution. After stirring for 0.5 h, the mixture was diluted with THF, filtered through Celite, and concentrated in vacuo to give 1 1 mg (78%) of the title compound as a white solid, which was used in the next step without further purification. LC/MS m/z: 427.34 (M-fBu+H) + , 483.38 (M+H) + , 965.75 (2M+H) + .

Step 3: frar)s-/V-(4-aminocyclohexyl)-3-(hydroxymethyl)-5-phenyladam antane-1 -carboxamide

The title compound was prepared from fe/f-butyl frans-A/-{4-[3-(hydroxymethyl)-5-phenyladamantane- 1 -amido]cyclohexyl}carbamate in the same manner as described above for 2-benzyl-2,7- diazaspiro[4.4]nonanedihydrochloride (example A1 , Step 1). LC/MS m/z: 366.35 (M-OH+H) + , 383.38 (M+H) + , 424.3 (M+H+CH 3 CN) + .

Examples A201 to A203 were prepared in the same manner as described for example A1 , Step 2 using the appropriate commercially available amine and carboxylic acid as starting materials.

phenyladamantane-1 -carboxamide

Example A204: A/-(piperidin-4-yl)adamantane-1 -sulfinamide

Step 1 : tert-butyl 4-(((-adamantan-1 -yl)sulfinyl)amino) -carboxylate

To a solution of adamantane-1 -sulfinyl chloride (50 mg, 0.23 mmol) in DCM (1 mL) was added triethylamine (64 uL, 0.46 mmol) and 4-dimethylaminopyridine (3 mg), followed by tert-butyl 4- aminopiperidine-1 -carboxylate (56 mg, 0.28 mmol). The mixture was stirred overnight, diluted with DCM, washed 1X with 1 M HCI, dried with brine and sodium sulfate, and evaporated. The crude product was purified via silica gel flash column using 5% MeOH in DCM as eluent to afford 49 mg of the title compound as a clear, viscous oil. LC/MS m/z: 383.35 (M+H) + , 765.68 (2M+H) +

Step 2: N-(piperidin-4-yl)adamantane-1 -sulfinamide

To a solution of fe/f-butyl 4-(((-adamantan-1 -yl)sulfinyl)amino)piperidine-1 -carboxylate (20 mg) in DCM (0.5 mL) was added TFA (0.5 mL), and the mixture was stirred for 2 hours at room temperature. All volatiles were evaporated and the residue dissolved in methanol and passed through an Agilent PL-HC03 ion exchange column. The filtrate was evaporated to afford 15 mg of the title compound as a clear oil which requires no further purification. LC/MS m/z: 283.33 (M+H) + eridin-4-yl)adamantane-1 -sulfonamide

Step 1 ; fe/f-butyl 4-(adamantane-1 -sulfonamido)piperidine-1 -carboxylate

To a solution of tert-butyl 4-(((-adamantan-1 -yl)sulfinyl)amino)piperidine-1 -carboxylate (30 mg, 0.08 mmol) in DCM (1 mL) was added m-CPBA (20 mg, 0.1 mmol), and the mixture was stirred at room temperature for 3 hours. After completion, the reaction was diluted with DCM and washed 2X with saturated NaHC03, dried over sodium sulfate, and evaporated. The crude product was purified via silica gel flash column using 50:50 hexanes:ethyl acetate as eluent to afford 23 mg of the title compound as a white solid. LC/MS m/z: 399.33 (M+H) + , 343.27 (M+H-(f-butyl)J + tep 2: N-(piperidin-4-yl)adamantane-1 -sulfonamide

The title compound was prepared from tert-butyl 4-(adamantane-1 -sulfonamido)piperidine-1 - carboxylate using the procedure described for example A204, Step 2. LC/MS m/z: 299.26 (M+H) + , 597.48 (2M+H) + -Methyl-4-(3-phenyladamantan-1 -yl)methyl)piperazine

To a suspension of LAH (0.030 g, 0.78 mmol) in THF (1 mL) cooled to 0 °C was added a solution of (4-fe/ -butoxycarbonylpiperazin-1 -yl)(3-phenyladamantan-1 -yl)methanone (0.060 g, 0.14 mmol) in THF (1 mL) dropwise. The resulting reaction mixture was brought to room temperature and then refluxed for 6 h, cooled to 0 °C and quenched with sat. aq. Na 2 S0 4 solution, filtered and the solvent was evaporated to give a crude residue that was purified using preparative HPLC to give the title compound as pale yellow syrup. Yield : 0.012 g (28%). LC/MS m/z: 325.28 (M+H) + H NMR (300

MHz, CDCI 3 ) δ 7.26 - 7.38 (m, 4 H) 7.17 (d, 1 H) 2.63 (br. s., 7 H) 2.39 (s, 3 H) 2.14 (br. s., 2 H) 2.06 (s, 2 H) 1 .83 (br. s., 4 H) 1 .66 (d, 2 H) 1 .60 (s, 2 H) 1 .45 - 1 .52 (m, 3 H)

The following examples were prepared from the appropriate amide using the same procedure as for example C1 .

Example C4: 1 -(3-(4-(adamantan-1 -yl)phenoxy)propyl)-4-methylpiperidine

-(4-(adamantan-1 -yl)phenoxy)propan-1 -ol

To a solution of 4-(adamantan-1 -yl)phenol (0.456 g, 2 mmol) in DMF (2 mL) was added 1 - bromopropanol (0.305 g, 2.2 mmol) followed by K 2 C0 3 (0.414 g, 3 mmol) and resulting reaction mixture was stirred at 80 °C for 6 h. Solvent was evaporated, and residue was extracted with ethyl acetate (2 x 10 mL) and washed with water (1 x 10 mL). Dried (Na 2 S0 4 ), filtered and evaporation of solvent under vacuum gave crude residue, which was purified by Si0 2 column chromatography to give the title compound as a white solid. Yield : 0.253 g (44%).

Step 2: 3-(4-(adamantan-1 -yl)phenoxy)propyl trifluoromethanesulfonate

To a solution of 3-(4-(adamantan-1 -yl)phenoxy)propan-1 -ol (0.143 g, 0.5 mmol) in DCM (5 mL) was added Et 3 N (0.101 g, 1 mmol) and cooled to 0 °C. Then Tf z O (0.225 g, 0.8 mmol) was added dropwise. Brought to room temperature gradually and stirred overnight. Reaction mixture was washed quickly with water (2 mL). Dried (Na 2 S0 4 ), filtered and evaporation of solvent gave crude product as syrup, which was used as such in the next step. Yield : 0.208 g (100%). Step 3: 1 -(3-(4-(adamantan-1 -yl)phenoxy)propyl)-4-methylpiperidine

To a solution of 3-(4-(adamantan-1 -yl)phenoxy)propyl trifluoromethanesulfonate (0.021 g, 0.05mmol) in THF (0.5 mL) was added 4-methylpiperidine (0.010 g, 1 mmol) followed by Et 3 N (0.010 g, 1 mmol) and resulting reaction mixture was stirred at room temperature for 12h. Solvent was evaporated, and residue was purified using preparative HPLC to give the title compound as pale yellow syrup. Yield :

0.007 g (39%). LC/MS m/z: 368.36 (M+H) + H NMR (300 MHz, CDCI 3 ) δ 7.19 - 7.28 (m, 2 H) 6.78 - 6.85 (m, 2 H) 3.98 (t, 2 H) 3.03 (d, 2 H) 2.62 (t, 2 H) 2.06 (br. s., 6 H) 1 .86 (d, 6 H) 1 .59 - 1 .80 (m, 8 H) 1 .39 (br. s., 2 H) 0.93 (d, 3 H). The following examples were prepared as decribed for examples C4 using the appropriate amines.

Example C7: 4-(4-(adamantan-1 -yl)phenoxy)-1 -(4-methylpiperidin-1 -yl)butan-2-ol

Step 1 : 2-(2-(4-(adamantan-1 -yl)phenoxy)ethyl)oxirane To a solution of 4-(adamantan-1 -yl)phenol (0.228 g, 1 mmol) in DMF (2 mL) was added K 2 C0 3 (0.276 g, 2 mmol) followed by 2-(2-bromoethyl)oxirane (0.151 g, 1 mmol) and reaction mixture was stirred at 80 °C for 12h. Solvent was evaporated, and residue was extracted with ethyl acetate (2 x 5 mL) and washed with water (1 x 5 mL). Dried (Na 2 S0 4 ), filtered and evaporation of solvent under vacuum gave crude residue, which was purified by Si0 2 column chromatography to give the title compound as a white solid. Yield : 0.224 g (75%). Step 2: 4-(4-(adamantan-1 -yl)phenoxy)-1 -(4-methylpiperidin-1 -yl)butan-2-ol

To a solution of 2-(2-(4-(adamantan-1 -yl)phenoxy)ethyl)oxirane (0.015 g, 0.05mmol) in 'PrOH (0.5 mL) was added 4-methylpiperidine (0.010 g, 1 mmol) and resulting reaction mixture was stirred under reflux for 12h. Solvent was evaporated, and residue was purified using preparative HPLC to give the title compound as an off-white solid. Yield : 0.007 g (36%). LC/MS m/z: 398.43 (M+H) +

The following examples were prepared as decribed for examples C7, step 2 using the appropriate amines.

(3-Phenyladamantan-1 -yl)methanol

The title compound was prepared by LAH reduction of 3-phenyladamantane-1 -carboxylic acid (A. Koperniku, I. Papanastasiou, G. B. Foscolos, A. Tsotinis, M. C. Taylor, and J. M. Kelly, Med. Chem. Commun. 2013, 4, 856-859).

2-(3-Phenyladamantan-1 -yl)ethan-1 -ol

The title compound was prepared from 2-(3-phenyladamantan-1 -yl)acetic acid in the same manner as described for (3-phenyladamantan-1 -yl)methanol.

(3-Phenyladamantan-1 -yl)methyltrifluoromethanesulfonate

The title compound was prepared from (3-phenyladamantan-1 -yl)methanol in the same manner as described for Example C4, Step 2.

2-(3-Phenyladamantan-1 -yl)ethyltrifluoromethanesulfonate

The title compound was prepared from 2-(3-phenyladamantan-1 -yl)ethan-1 -ol in the same manner as described for Example C4, Step 2.

(2S)-2-{[(3-phenyladamantan-1 -yl)methoxy]methyl}oxirane

To a solution of (2S)-oxiran-2-ylmethanol (5.7 mg , 0.061 mmol) in 0.5 ml_ anhydrous DMF, under N 2 was added NaH (60% w/w in mineral oil, 4 mg, 0.092 mmol) at 0 °C, and the resulting mixture was stirred for 1 h. Then (3-phenyladamantan-1 -yl)methyltrifluoromethanesulfonate (23 mg , 0.061 mmol) in 0.3 mL anhydrous DMF was added at 0 °C. The reaction mixture was stirred at 50 °C overnight, then diluted with EtOAc and washed with water. The organic phase was dried over Na 2 S0 4 , filtered, and concentrated in vacuo. The residue was purified by Si0 2 column chromatography (hexanes / EtOAc from 6:1 to 4:1) to give 5 mg (28%) of the product as a colorless oil.

(2R)-2-{[(3-phenyladamantan-1 -yl)methoxy]methyl}oxirane

The title compound was prepared from (3-phenyladamantan-1 -yl)methyltrifluoromethanesulfonate and (2R)-oxiran-2-ylmethanol in the same manner as described above for (2S)-2-{[(3-phenyladamantan-1 - yl)methoxy]methyl}oxirane. (2S)-2-{[2-(3-phenyladamantan-1 -yl)ethoxy]methyl}oxirane

The title compound was prepared from 2-(3-phenyladamantan-1 -yl)ethyltrifluoromethanesulfonate and (2S)-oxiran-2-ylmethanol in the same manner as described above for (2S)-2-{[(3-phenyladamantan-1 - yl)methoxy]methyl}oxirane.

(2R)-2-{[2-(3-phenyladamantan-1 -yl)ethoxy]methyl}oxirane

The title compound was prepared from 2-(3-phenyladamantan-1 -yl)ethyltrifluoromethanesulfonate and (2R)-oxiran-2-ylmethanol in the same manner as described above for (2S)-2-{[(3-phenyladamantan-1 - yl)methoxy]methyl}oxirane.

Example C10: (2S)-1 -(4-methylpiperidin-1 -yl)-3-[(3-phenyladamantan-1 -yl)methoxy]propan-2-ol

The title compound was prepared from (2S)-2-{[(3-phenyladamantan-1 -yl)methoxy]methyl}oxirane and 4-methylpiperidine in the same manner as described for Example C7, Step 2. LC/MS m/z: 398.36 (M+H) + .

Example C1 1 : (2R)-1 -(4-methylpiperidin-1 -yl)-3-[(3-phenyladamantan-1 -yl)methoxy]propan-2-ol

The title compound was prepared from (2R)-2-{[(3-phenyladamantan-1 -yl)methoxy]methyl}oxirane and 4-methylpiperidine in the same manner as described for Example C7, Step 2. LC/MS m/z: 398.38 (M+H) + .

Example C12: (2S)-1 -(4-methylpiperidin-1 -yl)-3-[2-(3-phenyladamantan-1 -yl)ethoxy]propan-2-ol

The title compound was prepared from (2S)-2-{[2-(3-phenyladamantan-1 -yl)ethoxy]methyl}oxirane and 4-methylpiperidine in the same manner as described for Example C7, Step 2. LC/MS m/z: 412.32 (M+H) + . Example C13: (2R)-1 -(4-methylpiperidin-1 -yl)-3-[2-(3-phenyladamantan-1 -yl)ethoxy]propan-2-ol

The title compound was prepared from (2R)-2-{[2-(3-phenyladamantan-1 -yl)ethoxy]methyl}oxirane and 4-methylpiperidine in the same manner as described for Example C7, Step 2. LC/MS m/z: 412.29 (M+H) + .

Example C14: 6-{[(3-phenyladamantan-1 -yl)methyl]amino}-/V-(piperidin-4-yl)pyridine-3-carboxamide

Step 1 : 3-phenyladamantane-1 -carboxamide

To a solution of 3-phenyladamantane-1 -carboxylic acid (300 mg, 1 .17 mmol) in dichloromethane (5 mL) is added diisopropylethylamine (610 mg, 3.51 mmol), HOAT (206mg, 1 .52 mmol), and EDCI*HCI (290mg, 1 .52 mmol). Finally, 2.8 mL of a 0.5 M solution of ammonia (1 .4 mmol) in dioxane is added, and the mixture is stirred overnight. The mixture is then diluted with DCM, washed 3X with 1 M HCI, dried over sodium sulfate, and evaporated. The crude product is purified via flash column using ethyl acetate as the eluent to give 180 mg of the title compound as a white solid. LC/MS m/z: 256.18 (M+H) + , 297.27 (M+H+CH3CN) + , 51 1 .39 (2M+H) +

Step 2: (3-phenyladamantan-1 -yl)methanamine

To a solution of 3-phenyladamantane-1 -carboxamide (180 mg, 0.71 mmol) in THF (5 mL) was slowly added 0.9 mL of a 2.4M solution of lithium aluminum hydride in THF. The mixture was heated to reflux and stirred overnight. After completion, the reaction was cooled in an ice bath and 80 uL water, followed by 80 uL 15% NaOH, and finally 0.24 mL water were added successively, and stirring was continued for one hour at room temperature. The formed precipitate was filtered off and the filtrate wasevaporated, giving 165 mg of the title compound as clear oil that was not purified further. LC/MS m/z: 242.22 (M+H) + , 283.26 (M+H+CH3CN) +

Step 3: methyl 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinate

To a solution of (3-phenyladamantan-1 -yl)methanamine (30 mg, 0.12 mmol) in THF (1 mL) was added 23 mg (0.15 mmol) of methyl 6-fluoronicotinate and 0.021 mL (0.18 mmol) of 2,6-lutidine. The mixture was heated to reflux and stirred overnight, then diluted with ethyl acetate and washed 2X with water, dried with brine and sodium sulfate, and evaporated to give the crude product. Purification on flash column using 1 :1 hexanes:ethyl acetate eluent gave 41 mg of the title compound as a white solid. LC/MS m/z: 377.33 (M+H) + , 418.29 (M+H+CH 3 CN) +

Step 4: 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinic acid

To 40 mg of methyl 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinate was added 1 mL of solvent THF:MeOH:water (2:1 :1) mixture, followed by 10 mg of LiOH. Stirring was continued for 2 hours and all solvents are then evaporated. The residue was dissolved in 1 M HCI and extracted 4X with ethyl acetate. The organics were dried with sodium sulfate and evaporated to give 35 mg of the title compound as a white solid, requiring no further purification. LC/MS m/z: 363.27 (M+H) + , 404.35 (M+H+CH 3 CN) + Step 5: fe/f-butyl 4-(6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinamido)piperidine-1 -carboxylate

To a solution of 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinic acid (15 mg, 0.04 mmol) in dichloromethane (1 mL) was added diisopropylethylamine (0.022 mL, 0.13 mmol), HOAT (7 mg, 0.052 mmol), and EDCI*HCI (10 mg, 0.052 mmol). Finally, fe/ -butyl 4-aminopiperidine-1 -carboxylate (10 mg, 0.052 mmol) was added, and the mixture was stirred overnight. The reaction was then diluted with DCM, washed 2X with water, dried over sodium sulfate, and evaporated. The crude product was purified via flash column using 1 :1 hexanes:ethyl acetate as eluent, giving 18 mg of the title compound as a white solid. LC/MS m/z: 545.48 (M+H) + , 586.47 (M+H+CH3CN) +

Step 6: 6-(((-3-phenyladamantan-1 -yl)methyl)amino)-N-(piperidin-4-yl)nicotinamide

To 18 mg of the protected amine was added 0.25 mL of dichloromethane and 0.25 mL of TFA. The mixture was stirred for 2 h and all volatiles are evaporated. The residue was dissolved in methanol, passed through an Agilent PL-HC03 ion exchange column to remove remaining TFA, and evaporated to give 10.5 mg of the title compound requiring no further purification. LC/MS m/z: 445.39 (M+H) +

Example C15: A/-[(3-phenyladamantan-1 -yl)methyl]-5-(piperidin-1 -ylmethyl)pyridin-2-amine

Step 1 : (6-(((3-phenyladamantan-1 -yl)methyl)amino)pyridin-3-yl)(piperidin-1 -yl)methanone

The title compound was prepared from 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinic acid and piperidine using the procedure from example C14, Step 5. LC/MS m/z: 430.37 (M+H) + , 471 .49 (M+H+CH3CN) + , 859.75 (2M+H) +

Step 2: A/-[(3-phenyladamantan-1 -yl)methyl]-5-(piperidin-1 -ylmethyl)pyridin-2-amine

The title compound was prepared from (6-(((3-phenyladamantan-1 -yl)methyl)amino)pyridin-3- yl)(piperidin-1 -yl)methanone using the procedure from example C14, Step 2. LC/MS m/z: 416.31 (M+H) + Example C16: 5-(aminomethyl)-N-[(3-phenyladamantan-1 -yl)methyl]pyridin-2-amine

Step 1 : 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinamide

The title compound was prepared from 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinic acid and ammonia using the procedure for example C14, Step 1 . LC/MS m/z: 362.17 (M+H) + , 403.39 (M+H+CH 3 CN) + ,

Step 2: 5-(aminomethyl)-N-((3-phenyladamantan-1 -yl)methyl)pyridin-2-amine

The title compound was prepared from 6-(((-3-phenyladamantan-1 -yl)methyl)amino)nicotinamide using the procedure from example C14, Step 2. LC/MS m/z: 348.26 (M+H) +

Example C17: {4-[(3-phenyladamantan-1 -yl)methoxy]phenyl}methanamine

Step 1 : methyl 4-((-3-phenyladamantan-1 -yl)methoxy)benzoate

To a flame dried flask was added toluene (1 ml_), (3-phenyladamantan-1 -yl)methanol (30 mg, 0.12 mmol), methyl-4-hydroxybenzoate (20 mg, 0.12 mmol), and 0.5 ml_ of a 0.5M (cyanomethylene) trimethyl phosphorane solution in THF. The mixture was brought to reflux and stirred overnight, then diluted with ethyl acetate, washed 2X with water, dried with brine and sodium sulfate, and evaporated. The crude residue was purified by flash column using 9:1 hexanes:ethyl acetate as eluent to give 22 mg of the title compound as a clear oil. H NMR (500MHz, CDCI 3 ) δ: 7.98 (d, 2H), 7.39 (d, 2H), 7.33 (t, 2H), 7.19 (t, 1 H), 6.91 (d, 2H), 3.88 (s, 3H), 3.64 (s, 2H), 2.25 (br:s, 1 H), 1 .93 (q, 4H), 1 .83 (br:s, 2H), 1 .76 (d, 5H) Step 3: 4-((-3-phenyladamantan-1 -yl)methoxy)benzoic acid

The title compound was prepared from methyl 4-((-3-phenyladamantan-1 -yl)methoxy)benzoate using the procedure from example C14, Step 4.

Step 4: 4-((-3-phenyladamantan-1 -yl)methoxy)benzamide

The title compound was prepared from 4-((-3-phenyladamantan-1 -yl)methoxy)benzoic acid using the procedure from example C14, Step 1 . LC/MS m/z: 362.07 (M+H) + , 403.30 (M+H+CH3CN) +

Step 5: {4-[(3-phenyladamantan-1 -yl)methoxy]phenyl}methanamine

The title compound was prepared from 4-((-3-phenyladamantan-1 -yl)methoxy)benzamide using the procedure from example C14, Step 2. LC/MS m/z: 348.30 (M+H) + , 331 .31 (M-NH2) +

Example C18: (2S)-1 -(4-methylpiperidin-1 -yl)-3-[4-(3-phenyladamantan-1 -yl)phenoxy]propan-2-ol

Step 1 : 1 -bromo-3-phenyladamantane

To a solution of phenyladamantane (100 mg, 0.47 mmol) in fluorobenzene (1 .5 ml_) wass added carbon tetrabromide (240 mg) and tetrabutylammonium bromide (10 mg). The mixture is stirred until homogenous, and 1 ml_ of a 50% (w/w) aqueous NaOH solution is added. The mixture is heated to 80°C overnight, cooled to rt, and diluted with water. The mixture is extracted 3X with DCM, and the organic layer dried with brine and sodium sulfate. The crude product is purified by flash column with pure hexanes as eluent to give 48 mg of the title compound as an off-brown solid. 3 C NMR (125 MHz, CDCI 3 ) δ: 148.93, 128.57, 126.37, 124.90, 66.21 , 54.57, 48.66, 41 .50, 34.96, 32.91

Step 2: 4-(3-phenyladamantan-1 -yl)phenol

To 50 mg (0.17 mmol) of 1 -bromo-3-phenyladamantane was added phenol (160 mg, 1 .7 mmol). The reaction was then heated to 120°C with vigorous stirring for 14 hours and allowed to cool to room temperature. The solid residue was dissolved in methanol, cooled to 0°C, and water was added until a precipitate forms. The formed solid is filtered off, washed with ice cold methanol and water, and dried by air suction giving 20 mg of the title compound as an off-brown solid. H NMR (500 MHz, CDCI 3 ) δ 7.39 (d, 2H), 7.327 (t, 2H), 7.27 (d, 2H), 7.19 (t, 1 H) 6.79 (d, 2H), 4.62 (br:s, 1 H), 2.3 (br:s, 2H), 2.0 (s, 2H), 1 .94 (dd, 9H), 1 .78 (br:s, 2H)

Step 3: (2S)-2-[4-(3-phenyladamantan-1 -yl)phenoxymethyl]oxirane

To a solution of 4-(3-phenyladamantan-1 -yl)phenol (32 mg, 0.1 mmol) in THF (1 ml_) is added NaHMDS (22 mg, 0.12 mmol). The mixture is brought to reflux for 10 minutes, and (S)-epichlorohydrin (50 mg, 0.50 mmol) is added in one portion. Reflux is continued overnight, and then all volatiles are evaporated. The residue was partitioned between ethyl acetate and water, washed 2X with water, then brine, dried over sodium sulfate, and evaporated. The crude product was purified by silica gel flash column with 90:10 hexanes:ethyl acetate as eluent, giving 20 mg of the title compound as a yellow oil. H NMR (500MHz, CDCI 3 ) δ: 7.40 (d, 2H), 7.32 (t, 4H), 7.19 (t, 1 H), 6.88 (d, 2H), 4.18 (dd, 1 H), 3.99-3.95 (m, 1 H), 3.34 (m, 1 H), 2.90 (t, 1 H), 2.76-2.74 (m, 1 H), 2.31 (br:s, 2H), 2.01 (s, 2H), 1 .94 (dd, 9H), 1 .78 (br:s. 2H)

Step 4: (2S)-1 -(4-methylpiperidin-1 -yl)-3-[4-(3-phenyladamantan-1 -yl)phenoxy]propan-2-ol

To a solution of (2S)-2-[4-(3-phenyladamantan-1 -yl)phenoxymethyl]oxirane (20 mg, 0.056 mmol) in isopropanol (1 ml_) is added triethylamine (0.012 ml_, 0.083 mmol) and 4-methylpiperidine (10 uL, 0.083 mmol). The mixture was brought to reflux and stirred overnight. After cooling, all volatiles are evaporated and the crude residue is added directly to a small silica flash column using first ethyl acetate and then 95:5 DCM:MeOH as eluents, giving 10.8 mg of the title compound as a viscous oil LC/MS m/z: 460.41 (M+H) + Example C19: (S)-1 -(4-(-3-ethyladamantan-1 -yl)phenoxy)-3-(4-methylpiperidin-1 -yl)propan-2-ol

Step 1 : 1 -bromo-3-ethyladamantane

80°C

The title compound was prepared from ethyladamantane using the procedure from example C18, Step 1 .

Step 2: 4-(3-ethyladamantan-1 -yl)phenol

The title compound was prepared from 1 -bromo-3-ethyladamantane and phenol using the procedure from example C18, Step 2.

Step 3: (S)-2-((4-(3-ethyladamantan-1 -yl)phenoxy)methyl)oxirane

The title compound was prepared from 4-(3-ethyladamantan-1 -yl)phenol and (s)-epichlorohydrin using the procedure from example C18, Step 3.

Step 4: (S)-1 -(4-(-3-ethyladamantan-1 -yl)phenoxy)-3-(4-methylpiperidin-1 -yl)propan-2-ol

The title compound was prepared from (S)-2-((4-(3-ethyladamantan-1 -yl)phenoxy)methyl)oxirane and 4-methylpiperidine using the procedure from example C18, step 4. LC/MS m/z:412.36 (M+H) +

Example E1 : 3-(3-phenyladamantan-1 -yl)-1 -(piperidin-4-yl)urea

To a solution of 3-phenyladamantane-1 -carboxylic acid (0.064 g, 0.25mmol) in toluene (0.75 mL) was added Et 3 N (0.04 ml, 0.29 mmol) followed by diphenylphosphorylazide (0.060 ml, 0.28 mmol) and the resulting reaction mixture was stirred at 70°C for 2 h. It was cooled to room temperature, then tert- butyl-4-aminopiperidine-1 -carboxylate (0.050 g, 0.25 mmol) was added and the mixture was stirred at room temperature overnight. The solvent was evaporated, and the residue was dissolved in DCM (0.5 mL). TFA (0.2 mL) was added and the mixture was stirred for 2h. The Ssolvent was evaporated and the crude residue was purified by preparative HPLC to give the title compound as an off- white solid. Yield: 9.1 mg (10.3%). LC/MS m/z: 354.36 (M+H) +

Example E2: 1 -(4-aminocyclohexyl)-3-(3-phenyladamantan-1 -yl)urea

The title compound was prepared following the same procedure as decribed for example E1 , using the appropriate amine.Yield : 10.1 mg (1 1 %). LC/MS m/z: 409.19 (M+H+CH 3 CN) +

Example E3: A/-(3-phenyladamantan-1 -yl)piperidine-4-carboxamide Stepl : 3-phenyladamantan-1 -amine

To a solution of 3-phenyladamantane-1 -carboxylic acid (0.064 g, 0.25 mmol) in toluene (0.75 mL) was added Et 3 N (0.04 ml, 0.29 mmol) followed by diphenylphosphorylazide (0.060 ml, 0.28 mmol) and the resulting reaction mixture was stirred at 70 °C for 2 h. After cooling rt, aq. NaHC0 3 solution was added and the mixture was stirred for 0.5 h, and extracted with ethyl acetate (2 mL). The organic extract was dried (Na 2 S0 4 ), filtered and concentrated to give the title compound which was used without purification in the next step.

Step 2: A/-(3-phenyladamantan-1 -yl)piperidine-4-carboxamide

To a solution of A/-BOC-piperidine-4-carboxylic acid (0.057 g, 0.25 mmol) in DCM (1 mL) was added EDCI.HCI (0.059 g, 0.30 mmol) followed by DIEA (0.06 ml, 0.29 mmol). The mixture was stirred for 0.5 h, then 3-phenyl-adamantane-1 -amine obtained from step 1 was added and the resulting mixture was stirred for 3 h, then TFA was added and stirring was continued for 2 h. The solvent was

evaporated and the crude residue was purified by preparative HPLC to give the title compound as a syrup. Yield: 1 .5 mg (1 .8%). LC/MS m/z: 339.40 (M+H) +

Examples E4 to E6 were prepared as described for example E3 using the appropriate amine and carboxylic acid.

Ex. Starting materials Product Analytical Data

E4 3-phenyladamantan-1 - H NMR (300 MHz, CDCI 3 ) δ 7.26 - amine 7.37 (m, 4 H) 7.13 - 7.21 (m, 1 H)

2-{1 -[(fe/ -butoxy) 5.16 (br. s., 1 H) 4.29 (d, 1 H) 3.47 (s, carbonyl]piperidin-4- 0.5 H) 3.05 (s, 0.5 H) 2.84 (d, 1 H) yl}acetic acid A/-(3-phenyladamantan-1 - 2.27 (br. s., 2 H) 2.13 (s, 2 H) 2.02 (d, yl)-2-(piperidin-4-yl) 4 H) 1 .72 - 1 .96 (m, 6 H) 1 .68 (br. s., acetamide 3 H) 1 .59 (s, 4 H). LC/MS m/z:

353.49 (M+H) + E5 3-phenyladamantan-1 - Γ H NMR (300 MHz, CDCI 3 ) δ 8.22 (br. amine I s., 2 H) 7.25 - 7.36 (m, 4 H) 7.12 - trans-4-{[(tert- 7.20 (m, 1 H) 3.05 - 3.41 (m, 1 H) butoxy)carbonyl]amino 2.15 - 2.36 (m, 3 H) 1 .96 - 2.15 (m,

}cyclohexane-1 - irans-4-amino-/V-(3-phenyl 10 H) 1 .71 - 1 .96 (m, 3 H) 1 .34 - 1 .71 carboxylic acid adamantan-1 -yhcyclo (m, 6 H). LC/MS m/z: 394.48

hexane-1 -carboxamide (M+H+CH 3 CN) +

E6 (3-phenyladamantan- LC/MS m/z: 353.37 (M+H) +

1 -yl)methanamine

1 -[(fe/ -butoxy)

carbonyl]piperidine-4-

A/-[(3-phenyladamantan-1 -yl)

carboxylic

methyl]piperidine-4- acid

carboxamide

In some embodiments, the invention provides for methods of treating infection by members of the Filoviridae family, which includes without limitation Ebolavirus, Marburgvirus, Cuevavirus, or any newly emerging filovirus genera. Five species of Ebolavirus have been identified: Zaire (EBOV),

Bundibugyo (BDBV), Tai Forest (TAFV), Sudan (SUDV), and Reston (RESTV). Two species of

Marburgvirus have been identified: (MARV) and Ravn (RAW). One species of Cuervavirus has currently been identified: Lloviu virus (LLOV).

In some embodiments, the compounds of the invention can selectively inhibit Ebolavirus infection. Infection by Ebolavirus in humans leads to Ebola Hemorrhagic Fever (EHF), the clinical manifestations of which are severe and/or fatal. The incubation period varies between four and sixteen days. The initial symptoms are generally a severe frontal and temporal headache, generalized aches and pains, malaise, and by the second day the victim will often have a fever. Later symptoms include watery diarrhea, abdominal pain, nausea, vomiting, a dry sore throat, and anorexia. By day seven of the symptoms, the patient will often have a maculopapular (small slightly raised spots) rash. At the same time the person may develop thrombocytopenia and hemorrhagic manifestations, particularly in the gastrointestinal tract, and the lungs, but it can occur from any orifice, mucous membrane or skin site. Ebolavirus infections may cause lesions in almost every organ, although the liver and spleen are the most noticeably affected. Both are darkened and enlarged with signs of necrosis. The cause of death (>75% in most outbreaks) is normally shock, associated with fluid and blood loss into the tissues. The hemorrhagic and connective tissue complications of the disease are not well understood, but may be related to onset of disseminated intra-vascular coagulation.

Infectious virus may linger in some tissues of some infected individuals for weeks and months after the intial infection.

In some embodiments, the compounds of the invention may inhibit Marburgvirus infection.

Marburg hemorrhagic fever (MHF) is a severe type of hemorrhagic fever associated with Marburgvirus infection, which affects both humans and non-human primates. The case-fatality rate for MHF was approximately 70% in a recent Angola outbreak. After an incubation period of 5-10 days, the onset of the disease is sudden and is marked by fever, chills, headache, and myalgia. Around the fifth day after the onset of symptoms, a maculopapular rash, most prominent on the trunk (chest, back, stomach), may occur. Nausea, vomiting, chest pain, a sore throat, abdominal pain, and diarrhea then may appear. Symptoms become increasingly severe and may include jaundice, inflammation of the pancreas, severe weight loss, delirium, shock, liver failure, massive hemorrhaging, and multi-organ dysfunction.

In some embodiments, the compounds of the invention may inhibit Cuervavirus infection or infections with any newly emerging filovirus.

In some embodiments, the compounds of the invention may inhibit infection by any virus, whether native or engineered, whose cell entry process is mediated by filovirus or hybrid filovirus glycoproteins.

Exemplary kits

The invention also includes kits. The kit has a container housing an inhibitor of the invention and optionally additional containers with other therapeutics such as antiviral agents or viral vaccines. The kit also includes instructions for administering the component(s) to a subject who has or is at risk of having an enveloped viral infection.

In some aspects of the invention, the kit can include a pharmaceutical preparation vial, a pharmaceutical preparation diluent vial, and inhibitor. The vial containing the diluent for the pharmaceutical preparation is optional. The diluent vial contains a diluent such as physiological saline for diluting what could be a concentrated solution or lyophilized powder of inhibitor. The instructions can include instructions for mixing a particular amount of the diluent with a particular amount of the concentrated pharmaceutical preparation, whereby a final formulation for injection or infusion is prepared. The instructions may include instructions for use in an oral formulation, inhaler, intravenous injection or any other device useful according to the invention. The instructions can include instructions for treating a patient with an effective amount of inhibitor. It also will be understood that the containers containing the preparations, whether the container is a bottle, a vial with a septum, an ampoule with a septum, an infusion bag, and the like, can contain indicia such as conventional markings which change color when the preparation has been autoclaved or otherwise sterilized.

INCORPORATION BY REFERENCE

All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definintions herein, will control.

Protocol A for pseudotvpe inhibitory testing of compounds. Utilizing a VSV pseudotype system, EBOV or BDBV mucin-domain-deleted glycoproteins were expressed along with the Renilla luciferease reporter gene to screen a library collection of small molecule compounds to identify individual compounds that inhibit infectivity of these VSV filovirus-GP pseudotyped viruses and not VSV expressing the native VSV glycoprotein [Cote, M.; Misasi, J.; Ren, T.; Bruchez, A.; Lee, K.; Filone, C. M.; Hensley, L; Li, Q.; Ory, D.; Chandran, K.; Cunningham, J. Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection, Nature (201 1) 477: 344-348; Chandran, K.; Sullivan, N. J.; Felbor, U.; Whelan, S.P.; Cunningham, J.M. Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection , Science 2005 308:1643-1645]. For select compounds, similar pseudotyped viruses expressing full-length EBOV [Genbank:

AAB81004], SUDV [Genbank: YP_138523.1 ], MARV [Genbank: AAC40460] or Lassa virus [Genbank: NP_694870] (a member of the Arenaviridae family of viruses) glycoproteins were also tested to further determine the specificity of action of our compounds against full length glycoproteins within and between different virus families. Vero cells (ATCC: CCL-81) were grown in clear 384 well plates (3000 cells/well) in DMEM media with 10% FBS, 1X Pen-Strep, non-essential amino acids and L-glutamine. After incubating overnight at 37°C and 5% C0 2 , cells were treated with compounds at desired concentrations and pseudotyped virus in assay media. VSV viruses expressing the full-length VSV glycoprotein, as well as all pseudotyped VSV viruses expressing the other viral glycoproteins, were generated in cultured HEK-293T cells (ATCC CRL-3216) grown in 10 cm dishes in DMEM

supplemented with 10% FBS, 1X Pen-Strep, non-essential amino acids, L-glutamine and 500 μg/mL G418 antibiotic. When cells reached approximately 80% confluency, they were transfected with a mixture of 15 μg of the pCAGGS plasmid encoding one of the desired glycoproteins and 45 μΙ of PEI (polyethylenimine) transfection reagent. The cells were incubated with the solution for 5 hours at 37°C at 5% C0 2 . The cells were then washed and the mixture replaced with supplemented DMEM and incubated at 37°C at 5% C0 2 for approximately 16-18 hours. Subsequently cells were infected with approximately 50 μΙ of VSV parent pseudotype virus lacking VSV glycoprotein and containing the gene for luciferase. The cells were infected for 1 hour, then washed 1X with PBS and incubated in supplemented media. 24 hours post-infection, supernatant was collected, aliquoted and stored at - 80°C. For VSV-Luciferase pseudotypes, one aliquot was thawed and tested in a serial dilution for luminescence activity in Vero cells as described in the Luciferase assay protocol (below). Each of the viral supernatants generated was diluted (from 1 :100 to 1 :2000) to give similar luminescence signal / background values of > 200 and stored at -80°C as aliquots for later use. Assay media consisted of 50% Opti-MEM, 50% DMEM, with 1 % FBS, Pen-Strep, non-essential amino acids and L-glutamine. Final DMSO concentration in the compound testing wells was kept < 1 % and control wells were treated with assay media and 1 % DMSO. Cells were incubated for 24 hours at 37°C and 5% C0 2 . The compound-virus mixture was aspirated off the cells 24 hours post-infection and washed 1X with PBS. Cells were lysed using 20 μΙ of lysis buffer from a Luciferase kit diluted according to manufacturer's (Thermo Scientific) instructions. After incubating for approximately 20 minutes, 5 μΙ of cell lysate was transferred to an opaque white plate and mixed with 12.5 μΙ of Coelenterazine diluted in buffer. This mixture was incubated at room temperature for 10 minutes on a plate shaker, and then the luminescence was read using a plate reader (Beckman Coulter DTX 880 multimode detector with an emission of 535 nm) Luminescence signals were obtained for compound containing and control wells to determine % activity (inhibition of luciferase signal) for each compound.

To identify filovirus inhibitors a library collection of commercial compounds was initially tested at a 5 or 10μΜ concentration against the EBOV pseudotype virus and selected compounds were tested against BDBV pseudotyped viruses to facilitiate identification of broader-spectrum filovirus inhibitors. In order to distinguish between inhibitory activities of the compounds on filovirus cell entry from those potentially resulting from cellular cytoxicity and/or inhibiton of the replication of VSV virusesor luciferase reporter, selected compounds were tested in cytotoxicity assays (see cytotoxicity assay methods below) and/or against parent VSV virus, which expresses the native VSV glycoprotein (rather than a filovirus glycoprotein). Compounds exhibiting inhibitory activity against the EBOV and BDBV pseudotype viruses at concentrations (≥ 5-10 fold) below those observed in cytotoxicity and/or parental VSV assays were thereby identified as filovirus cell entry inhibitors. A number of these compounds were selected for prosecution in dose-response experiements and/or additional functional assays (see below) to further characterize their potency and antiviral spectrum of activity.

Tables 5 - 9. Examples. Example (Ex.) compounds from chemical series A, B, C, D, and E including their inhibitory activities (EC 50 values; the concentration at half-maximal inhibition) are shown against the indicated pseudotyped viruses (EBOV and BDBV) or parental VSV and/or for cytotoxicity, which was evaluated as the % cell viability (% CV) at 3, 5 and/or 10 μΜ compound or as the CC 50 (the concentration inducing half-maximal cell death). Vera cells were infected with dilutions of the viral stocks of the luciferase-expressing EBOV or BDBV pseudotyped viruses, or VSV native viruses (to rule out inhibitors of VSV non- glycoprotein gene products or the luciferase reporter) and incubated for 24 h with or without the example compounds; cells were lysed and luciferase activity read on a Beckman Coulter DTX 880 detector. Compound cytotoxicity was determined to rule out potential false-positive inhibitory activity on filovirus pseudotyped virus cell entry. Compounds exhibiting potential activity against the EBOV and BDBV pseudotyped viruses were more fully characterized in cytotoxicity and/or dose response experiments to confirm activity and determine EC 50 values. Compounds exhibiting activity against one or more pseudotyped filoviruses without comparable VSV activity or cytotoxicity, indicates they are of potential therapeutic interest to treat filovirus infection.

Compounds were serially diluted and added to Vera cells (4000 cells/well) with final DMSO concentration maintained at 1 % in growth media consisting of minimal essential media (MEM) with 2% FBS. The plates were incubated at 37 °C for 7 days, and then dead cells were removed by washing with Phosphate buffered saline (PBS). Cells were stained with neutral red vital dye for 1 hour and then de-stained with a solution of 50% ethanol/ 1 % acetic acid solution. Absorbance was read at 540 nm and 690 nm on a Spectramax Plus 384 spectrophotometer. Data were analyzed as (540 nm - 690 nm) and then compared to untreated controls to obtain % cell viability. Table 5

B8 A/-[2-(morpholin-4-yl)-2- 3.6 4.6 ND 103 ND

(thiophen-2-yl)ethyl]-3- phenyladamantane-1 - carboxamide

B9 Λ/-(1 -benzylpiperidin-4- 3.8 3.1 ND 1 12 ND yl)-3-chloro

adamantane-1 - carboxamide

B10 A/-cyclopropyl-2-{4-[(3- <5 ND <20 ND ND phenyladamantan-1 -yl)

carbonyl]piperazin-1 - yl}acetamide

B1 1 A/-{1 -azabicyclo [2.2.2] <5 ND <20 ND 92 octan-3-yl}-3-(4-methyl

phenyl)adamantane-1 - carboxamide

B12 3,5-dimethyl-/V-[2- 5.0 ND <20 ND 90

(piperidin-1 -yl)ethyl]

adamantane-1 -

carboxamide

B13 1 -benzyl-4-{[3-(4- >5 >5 <20 ND ND methylphenyl)

adamantan-1 -yl]

carbonyl} piperazine

B14 1 -methyl-4-{[3-(4- 5.6 ND <20 ND 95 methylphenyl)

adamantan-1 - yl]carbonyl} piperazine

B15 Λ/-(1 -benzylpiperidin-4- 5.9 3.9 ND 99 ND yl)adamantane-1 - carboxamide

B16 1 -ethyl-4-{[3-(4- 1 .0 0.9 ND ND ND methylphenyl)adamanta

n-1 -yl]carbonyl}

piperazine B17 2-(adamantan-1 - >5 <5 ND ND ND ND

9 ylformamido)-/V-(1 - benzylpiperidin-4-yl)-3- methylbutanamide

B18 2-(4-{[3-(4-methyl >5 >5 ND ND ND ND phenyl) adamantan-1 -yl]

carbonyl}piperazin-1 - yl)ethan-1 -ol

B19 N-(piperidin-4- 1 .1 >1 ND ND ND ND yl)adamantane-1 -

0 I^NH carboxamide

Table 6

EC 50 (μΜ) cc 50 EC 50 (μΜ) cc 50 EC 50 (μΜ) cc 50

Ex. Ex. Ex.

EBOV BDBV (μΜ) EBOV BDBV (μΜ) EBOV BDBV (μΜ)

A1 0.22 0.25 5.8 A50 2.9 3.3 54.9 A99 0.69 0.19 26.7

A2 0.30 0.39 8.5 A51 9.0 >10 10 A100 0.22 0.18 6.3

A3 0.03 1 .24 >3 A52 4.9 4.0 >10 A101 >1 >1 ND

A4 0.19 1 .24 >3 A53 9.2 9.0 ND A102 0.12 0.21 29

A5 1 .4 3.9 >3 A54 4.0 10.0 >10 A103 0.07 0.28 7.9

A6 0.21 0.25 16.8 A55 2.9 2.6 >10 A104 0.06 0.22 66

A7 0.48 0.84 >10 A56 3.1 10.0 >10 A105 0.14 1 .05 35

A8 0.19 0.22 >10 A57 2.9 >10 ND A106 >1 >1 ND

A9 0.23 0.29 10.8 A58 0.65 8.0 >10 A107 0.046 0.30 14.7

A10 0.24 0.21 6.9 A59 1 .1 1 .5 ND A108 0.025 0.21 16.5

A1 1 0.2 0.23 ND A60 0.21 0.17 ND A109 0.080 0.93 37

A12 0.36 0.43 2.5 A61 0.14 0.08 >10 A1 10 0.021 0.19 17.5

A13 0.32 0.32 2.3 A62 0.16 0.1 1 30 A1 1 1 0.018 0.15 10.7

A14 0.2 0.23 6.2 A63 0.08 0.08 >10 A1 12 0.037 0.26 7.2

A15 1 .7 2.6 >10 A64 0.71 0.18 >10 A1 13 >1 >1 ND

A16 0.1 1 0.35 12.9 A65 0.09 0.09 >10 A1 14 >1 >1 ND

A17 0.93 0.48 >10 A66 0.16 0.10 >10 A1 15 0.98 >1 59

A18 >10 >10 ND A67 0.39 0.41 >10 A1 16 >1 >1 >100

A19 0.29 0.44 3.9 A68 0.035 0.16 >10 A201 1 .1 >1 16.4

A20 0.49 0.41 10.6 A69 0.22 0.24 22.4 A202 0.59 0.96 41 .5

A21 >10 >10 ND A70 0.49 0.19 >50 A203 0.91 1 .02 3.9 A22 0.26 0.22 ND A71 0.13 0.13 4.6 A204 >1 >1 >100

A23 0.21 0.17 ND A72 0.28 0.29 17.4 A205 >1 >1 37

A24 0.29 0.29 7.2 A73 0.32 0.37 13.0 C1 0.18 0.206 5.8

A25 0.45 0.19 >10 A74 1.72 >1 14.0 C2 0.25 0.2 3.6

A26 0.12 0.09 34 A75 0.3 0.3 17.7 C3 0.234 0.21 1.8

A27 0.28 0.14 >10 A76 1.05 1.06 ND C4 0.14 0.4 7.2

A28 >10 >10 >10 A77 0.12 0.22 8.7 C5 0.14 0.55 14.3

A29 0.34 0.27 5.8 A78 0.02 0.09 ND C6 0.12 0.64 3.8

A30 0.34 0.22 7.5 A79 0.26 0.29 4.2 C7 0.47 0.77 4.1

A31 >10 >10 >10 A80 0.01 0.05 10.0 C8 0.14 0.29 1.3

A32 0.2 0.26 1.6 A81 0.18 0.11 13.9 C9 0.66 0.99 8.3

A33 0.32 0.3 2.2 A82 0.13 0.16 4.5 C10 0.19 0.17 3.6

A34 0.17 0.22 1.1 A83 0.1 0.18 12.9 C11 0.18 0.11 3.6

A35 0.28 0.22 >10 A84 0.146 0.11 9.0 C12 0.27 0.19 1.2

A36 0.51 0.3 4.3 A85 0.013 0.096 12.1 C13 0.17 0.11 3.6

A37 0.23 0.24 >10 A86 0.155 0.18 3.9 C14 >1 1 3.9

A38 0.25 0.19 >10 A87 0.14 0.19 1.4 C15 >1 0.98 3.9

A39 0.51 0.21 19.1 A88 0.36 0.50 12.5 C16 0.98 0.75 8.0

A40 0.31 0.16 ND A89 0.25 0.88 40.7 C17 >1 1.2 ND

A41 0.103 0.18 13.6 A90 0.05 0.20 59.0 C18 0.08 0.26 2.7

A42 0.121 0.19 13.9 A91 0.12 0.19 17.4 C19 0.11 0.20 >100

A43 0.14 0.08 1.2 A92 0.19 0.7 10.8 E1 >1 0.77 18.3

A44 0.09 0.19 11.0 A93 0.21 0.36 12.2 E2 0.87 0.61 12.6

A45 0.06 0.14 7.2 A94 1.0 0.88 5.1 E3 >1 0.27 36.3

A46 0.18 0.21 13.5 A95 0.49 0.97 23.1 E4 1.08 0.79 23.1

A47 0.07 0.17 36.2 A96 0.03 0.08 11.6 E5 0.68 0.23 13.3

A48 1.04 2.0 54.9 A97 0.34 0.25 100 E6 0.71 0.20 11.9

A49 0.21 0.29 10 A98 1.16 0.58 11.3 Table 7

D37 1 -{[2-(adamantan-1 -yl)imidazo[1 ,2- 8.9 3.2 ND a]pyridin-3-yl]methyl}piperidine

D38 N-[5-(adamantan-1 -yl)-1 H-pyrazol-3-yl]- 3.7 6.8 ND

1 -(prop-2-yn-1 -yl)piperidine-4-

0 carboxamide

D39 (2S)-1 -[4-(3,5-dimethyladamantan-1 - 0.12 0.25 3.2 yl)phenoxy]-3-(piperidin-1 -yl)propan-2-ol

D40 (2S)-1 -[4-(3,5-dimethyladamantan-1 - 0.19 0.46 2.5 yl)phenoxy]-3-(4-methylpiperidin-1 - yl)propan-2-ol

D41 A/-[2-(adamantan-1 -yl)ethyl]-4- 2.3 9.7 >3

(morpholin-4-ylmethyl) benzamide

D42 Λ/-[1 -(adamantan-1 -yl)ethyl]-2-[5-(2- 3.2 3.9 >10 methylphenyl)-2H-1 ,2,3,4-tetrazol-2-yl]

acetamide

D43 (2f?)-/V-[1 -(adamantan-1 -yl)ethyl] 4.6 2.5 ND pyrrolidine-2-carboxamide

D44 Λ/-[1 -(adamantan-1 -yl)ethyl]-2-[3- 5.8 2.7 >10

(trifluoromethyl)-l H-pyrazol-1 -yl]

acetamide

D45 1 -[1 -(adamantan-1 -yl)ethyl]-3-[2-methyl- 6.5 4.6 >10

2-(morpholin-4-yl)propyl] urea

D46 A/-(adamantan-1 -ylmethyl)-3- 6.7 4.3 >10

(dimethylamino)piperidine-l -

0

carboxamide

D47 Λ/-[1 -(adamantan-1 -yl)ethyl]-2-[5-(furan- 9.3 5.4 >10

2-yl)-2H-1 ,2,3,4-tetrazol-2-yl]acetamide

As mentioned above, the EBOV and BDBV glycoproteins expressed in pseudotyped virus assays were missing the mucin domain given that it can contribute to cellular toxicity [Francica, J.R.; Matukonis, M.K.; Bates, P. Requirements for cell rounding and surface protein down-regulation by Ebola virus glycoprotein, Virology (2009), 383:237-247]. However, because native Ebolaviruses express glycoproteins that include the mucin domain, we further tested the compounds against native full-length EBOV glycoproteins in pseudotyped viruses (also expressing the Renilla luciferase gene). A representative subset of the compounds exhibiting potent inhibition of the EBOV and BDBV pseudotyped viruses relative to parental VSV (and/or cytotoxicity) were therefore tested against pseudotyped viruses expressing full-length EBOV and SUDV GP to confirm activity against these full length filovirus glycoproteins. In addition, these compounds were also tested against a VSV pseudotyped virus expressing full-length LASV glycoprotein to further define the antiviral activity spectrum of the compounds against both distinct filovirus species as well as other virus families expressing other class I fusion glycoproteins (LASV). As shown in Table 8, compounds of the invention exhibited inhibition of Ebolavirus glycoprotein pseudotyped viruses but did not exhibit similar inhibition of the parental VSV or pseudotyped LASV viruses (data are a average +/- sd , n = 3 or 4). However, the compound inhibition EC 50 values obtained against full-length EBOV and SUDV pseudotyped viruses are comparable to those found for the mucin-deleted EBOV and BDBV pseudotyped viruses. These data further indicate that compounds of the series described herein are inhibitors of filovirus cell entry and as such are inhibitors of filovirus infection.

Table 8

*EBOV_FL: full-length Ebola Zaire GP, MARV, LASV and VSV are also full-length glycoproteins

As described above, the amino acid homology of full-length MARV and Ebolavirus glycoproteins is >30%. To better understand the potential spectrum of activity within the Filoviridae family, including the potential activity of compounds against filoviruses more distantly related than EBOV, BDBV and SUDV the compounds listed in Table 8 were tested against the full-length Marburgvirus (MARV) pseudotyped virus. In contrast to the nearly equivalent activities and EC 50 values observed against EBOV, BDBV and SUDV pseudotyped viruses, most of the compounds exhibited less inhibition against pseudotyped viruses expressing the more distantly related MARV GP. However, at least some compounds inhibited MARV pseudotyped virus with sub-micromolar EC 50 concentrations (e.g., compounds D8, D32, D29, A45 and D21). Some compounds such as D8 and D21 appeared to inhibit non-filovirus GP pseudotype viruses including VSV (classs III GP) and LASV (class I GP) but only at higher (>10 fold) cytotoxic concentrations. In addition, compounds of the invention, although containing an adamantyl or adamantyl-like group as found in influenza drugs amantadine and rimantidine [Wanka L, Iqbal K and Schreiner PR, The Lipophilic Bullet Hits the Targets: Medicinal Chemistry of Adamantane Derivatives, Chem Rev (2013), 1 13: 3516-3604] did not exhibit any activity against the influenza M2 ion channel or live influenza virus at concentrations of 10 uM or greater. The results indicate that the compounds and chemical series described herein inhibit the cell entry of a broad range of filoviruses and/or viruses expressing filovirus glycoproteins while exhibiting no similar activity against the glycoproteins from the other enveloped viruses examined.

Protocol B - Native Ebola plaque and viral yield reduction assays. Biosafety Safety Level 2 (BSL2) pseudotyped viruses expressing filovirus GPs were used (above) as surrogates to facilitiate the identification of inhibitors of wild-type Biosafety safety level 4 (BSL4) filoviruses, which may only be studied in highly specialized containment facilities. To confirm activity against native BSL4 Ebola virus example compounds were tested against EBOV (Mayinga) in both plaque forming and viral yield reduction (VYR) assay formats (Table 9) under stringent BSL4 testing requirements. In the plaque assay format confluent or near confluent (Vera) cell culture monolayers in 12-well disposable cell culture plates are prepared. Cells are maintained in MEM or DMEM supplemented with 10% FBS. For antiviral assays the same medium is used but with FBS reduced to 2% or less and supplemented with 1 % penicillin/streptomycin. The test compound is prepared at four log 0 final concentrations in 2X MEM or 2X DMEM. The virus only and cytotoxicity (compound only) controls are run in parallel with each tested compound. Further, a known active drug (favipiravir) is tested as a positive control drug with each test run. Test compounds and positive controls are tested in biological triplicates. The assay is initiated by first removing growth media from the 12-well plates of cells, and infecting cells with 0.01 MOI of virus or about 50 to 100 plaque forming units (pfu). Cells are incubated for 60 min: 100μΙ inoculum/ well, at 37°C, 5% C0 2 with constant gentle rocking. Virus inoculum is removed, cells washed and overlaid with either 1 % agarose or 1 % methylcellulose diluted 1 :1 with 2X MEM and supplemented with 2% FBS and 1 % penicillin/streptomycin and supplemented with the corresponding drug concentration. Cells are incubated at 37° C with 5% C0 2 for 10 days. The overlay is removed and plates stained with 0.05% crystal violet in 10% buffered formalin for approximately twenty minutes at room temperature. The plates are washed, dried and the number of plaques counted. The number of plaques in each set of compound dilution is converted to a percentage relative to the untreated virus control. The 50% effective (EC 50 virus-inhibitory) concentration is calculated by linear regression analysis. The cytotoxicity assay (In vitro Toxicology Assay Kit, Neutral red based; Sigma) is being performed in parallel in 96-well plates following the manufacturer's instructions. Briefly, growth medium is removed from confluent cell monolayers and replaced with fresh medium (total of 100μ I) containing the test compound with the concentrations as indicated for the primary assay. Control wells contain medium with the positive control or medium devoid of compound. A total of up to five replicates are performed for each condition. Plates are incubated for 3, 5, or 10 days at 37°C with 5% C0 2 . The plates are stained with 0.033% neutral red for approximately two hours at 37°C in a 5% C0 2 incubator. The neutral red medium is removed by complete aspiration, and the cells rinsed 1X with phosphate buffered solution (PBS) to remove residual dye. The PBS is completely removed and the incorporated neutral red eluted with 1 % acetic acid/50% ethanol for at least 30 minutes. Neutral red dye penetrates into living cells: the more intense the red color, the larger the number of viable cells present in the wells. The dye content in each well is quantified using a 96-well spectrophotometer at 540 nm wavelength and 690 nm wavelength (background reading). The 50% cytotoxic (CC 50 , cell- inhibitory) concentrations are then calculated by linear regression analysis. The quotient of CC 50 divided by EC50 gives the selectivity index (Sl 50 ) value. The viral yield reduction (VYR) assay format involves similar methodology to that described for the plaque assay format using 12-well plates of cells. Cells are infected and treated with test compound prepared at eight half-log 0 final concentration and applied in 1 ml of total volume of liquid media. In parallel compounds are added under similar conditions in the absence of virus for CC 50 determination. To quantify virus particles produced tissue culture supernatant (TCS) aliquots are collected at 72 hours, serially diluted in 10-fold increments and added to fresh monolayer of cells overlaid with 1 % agarose mixed 1 :1 with 2XMEM supplemented with 2% FBS and 1 % penicillin, and the number of plaques determined. Test compounds and a positive control (favipiravir) are tested in biological triplicates. Plotting the log 0 of the inhibitor concentration versus log 0 of virus produced at each concentration allows calculation of the 90% (one log 0 ) effective concentration or EC 90 by linear regression. The quotient of CC 50 divided by EC 90 gives the Sl 90 selectivity index.

In the plaque assay example compounds showed half-maximal activity (EC 50 ) against the native filovirus (EBOV) at sub (0.89) to low (1 .1) micromolar concentrations (Table 9). The Sl 50 selectivity index (= CC 50 /EC 50 ) is typically used to determine whether a compound is exhibiting true antiviral inhibitory in vitro. Since Sl 50 values >10 are accepted as confirmation of bona fide inhibitory activity against the virus, rather than artifactual activity reflecting cellular cytotoxicity, the data confirm the compounds as inhibitors of live filovirus. As a further test for antifilovirus inhibitory activity the compounds were then tested in a VYR assay where EC 90 values (concentration at which 90% virus inhibition is observed) were determined. Compounds exhibiting Sl 90 (= CC 50 /EC 90 ) values approaching 10 are thought to represent compounds that are sufficiently active to provide inhibition of viruses in living mammals. As shown in Table 9 the example compounds tested in the VYR assay exhibit 90% of the live BSL4 EBOV (Mayinga) virus at low micromolar (5.6 and 8.9 uM) concentrations while exhibiting Sl 90 values approaching 10. By comparison the positive control drug compound favipiravir exhibited EC 90 and Sl 90 values of 333 uM and 3, respectively. The data from the two independent live virus antiviral assays for the example compounds validate the utilization of pseudotyped virus assays to identify bona fide filovirus inhibitors compatible with administration in mammals in vivo as a method of treatment for filovirus infection. Table 9

Examples 10-12. In addition to the ability of compounds to inhibit live filoviruses in vitro, compounds must also have certain drug-like properties for them to be used to inhibit filoviruses and provide methods of treatment for filovirus infection in mammals in vivo. Such compounds may exhibit drug-like properties including but not limited to chemical stability against degradation by and lack of inhibition of liver microsomal CYP p450 enzymes, cell permeability and oral bioavailability (if the drug is to delivered orally) and lack of inhibition of the hERG ion channel, which is associated with cardiac safety [Kerns, E.H. Li, D. Drug-like Properties: Concepts, Structure Design and Methods from ADME to Toxicity Optimization, (2008) Academic Press, Burlington MA]. To characterize drug-like properties of the chemical series example compounds were evaluated for metabolic stability in human, mouse and monkey liver microsome assays (Table 10). Compounds exhibiting half-lives (T ½ ) > 60 minutes indicate attractive chemical stability. The demonstration of good microsomal stability in human and nonhuman species facilitates the ability to test and optimize compounds in preclinical animal studies. To reduce or prevent serious/!ife-ihreatening conditions caused by exposure to lethal or permanently disabling toxic agents where human efficacy trials are not feasible or ethical (such as filovirus infection) the FDA has provided an approach to test and approve drugs using the Animal Efficacy Rule; whereby the FDA can rely on evidence from animal studies to provide substantial evidence of product effectiveness. In the absence of an epidemic filovirus outbreak in humans with a sufficiently large patient population efficacy data for new methods of treatment for filovirus infection may only be obtained from relevant animal models (e.g., mouse and monkey efficacy studies). Thus the translation of drug like-properties from one species to another significantly facilitates the testing and development of filovirus inhibitor compounds.

Table 10

Microsomal stability T ½ (min)

Ex.

human Mouse Monkey

A63 258 >210 237

A68 187 >240 142

A80 103 >120 89

A78 462 267 >120

A85 >120 >120 84 To further characterize compounds of the invention for the inhibition of filoviruses in mammals in vivo additional drug-like properties of an example compound (compound A78) were determined including solubility, Caco-2 cell permeability, protein binding, Cyp-p450 inbhition and hERG ion channel inhibition (Table 1 1). The example compound was not an inhibitor of human Cyp-p450 enzymes 1A2, 2B6, 2C9, 2C19, 2D6 while for 3A4 it exhibited an IC 50 of -28 uM for one substrate and > 100 uM for another. The data indicate little if any metabolic liabilities related to Cyp-p450 inibition (formation of metabolites was measured by LC/MS). In addition, the IC 50 for compound A78 against the hERG ion channel was > 10 uM (the highest concentration tested), which indicates attractive cardiac safety properties. Compound A78 exhibits high protein binding (5 uM test concentration) in PBS, which will factor in for in vivo efficacy studies and therapeutic dosing while its solubility at neutral pH is >500 uM, which will facilitate formulation and dissolution in vivo. To help determine the potential for oral administration we evaluated the permeability of compound A78 (5 uM test concentration in PBS in absence or presence of P-gp inhibitor Verapamil) in the Caco-2 in vitro model where it demonstrated permeability but was found to be effluxed (ratio of B>A/A>B was 4.32) by P-gP; the addition of verapamil, a known P-gP pump inhibitor, inhibited the efflux to a ratio of less than 1 .

Table 11

While the P-gP efflux of compound A78 was not ideal, there remained sufficient absorption to justify exploration of an IV/PO PK (intravenous/oral pharmacokinetic) study in mice of compound A78, When administered to mice (Table 12 and Figure 1), as either 1 mg/kg IV or 10 mg/kg PO, with analysis of blood plasma samples obtained over a 24 hr period, the example compound was found to demonstrate excellent oral bioavailability (72%), clearance was less than hepatic blood flow and the compound exhibited a half-life of 7.1 hrs and volume of distribution of 14L/kg. These values are consistent with single or twice daily oral dosing. Table 12

a NA, not applicable; b NC, not calculated.

In summary, example compounds of the invention exhibit potencies of i) low nanomolar EC 50 activity against pseudotyped viruses expressing a range of filovirus glycoproteins and ii) sub to low uM EC 50 and low uM EC 90 activities against natvie BSL4 filovirus with selectivity indices that confirm them as bona fide filovirus inhibitors. In addition, drug-like property characterization of example compounds indicate: iii) attractive microsome stability in human, mouse and monkey (potential efficacy models for application of the Animal Efficacy Rule), and other drug-like properties and; iv) mouse PK properties for an example compound were characterized by a long half-life, low clearance and excellent oral bioavailability. These data indicate that the compounds of the invention have sufficient potency and drug-like properties to inhibit filoviruses in mammals in vivo as a method of treatment for filovirus infection.

This is further supported by comparisons between favipiravir and compounds of the invention. Favipiravir is a drug that has been evaluated in human clinical trials during the 2014-16 Ebola outbreak with 1 1 1 patients in Guinea as a method of treatment for Ebola virus infection. Although this study was not powered to define efficacy and tolerability the results indicated that patients with moderate levels of viremia (below 10 8 genome copies/mL) responded to the drug (3-4 log drop in viral load) while those with higher levels (>10 8 genome copies/mL) of viremia did not [Sissoko, D. Experimental Treatment with Favipiravir for Ebola Virus Disease (the JIKI Trial): A Historically Controlled, Single-Arm Proof-of-Concept Trial in Guinea. (2016) PLoS Med. 2016 Mar 1 ;13(3):e1001967]. While efficacy was limited to those in early stages of infection it was likely limited by certain specific properties including potency and chemical stability, i.e., favipiravirs weak potency against EBOV and short half-life in humans (1 - 4 hr). In this context it is useful to compare the potency and dosing characteristics of favipiravir with compounds of the current invention to gauge the potential for their efficacy in inhibiting EBOV infection in mammals including humans. The published EC 50 for favipiravir against native EBOV virus has been reported as 67 uM values [Oestereich, L. et al. Successful treatment of advanced Ebola virus infection with T-705 (favipiravir) in a small animal model. Antiviral Res. (2014) 105:17-21 ] whereas EC 50 values for example compounds of the invention are ~ 1 uM (67 times more potent than favipiravir). Furthermore, while we have not tested our compounds in humans and cannot yet compare the bioavailability and pharmacokinetic properties of the two compounds in humans to date comparisons of the half-life of favipiravir (1 .8 h with 150 mg/kg twice daily oral dosing) [Mentre, F., et al. Dose regimen of favipiravir for Ebola virus disease. Lancet Infect. Dis. (2015) 15(2):150-1 ] versus example compounds of the invention (7 hr half-life with 10 mg/kg single dosing, for compound A78) indicate that compounds of the current invention are significantly more potent than favipiravir and have greater potential to reach higher plasma concentrations in mammals. These comparisons provide compelling support for the utilization of compounds of the invention to inhibit filoviruses including in vitro in mammals and as methods of treatment for filovirus infection in humans.