WO2013016160A1 - NOVEL IMIDAZO[1,2-A]PYRAZINE DERIVATIVES AS mTOR INHIBITORS - Google Patents

NOVEL IMIDAZO[1,2-A]PYRAZINE DERIVATIVES AS mTOR INHIBITORS Download PDF

Info

Publication number
WO2013016160A1
WO2013016160A1 PCT/US2012/047522 US2012047522W WO2013016160A1 WO 2013016160 A1 WO2013016160 A1 WO 2013016160A1 US 2012047522 W US2012047522 W US 2012047522W WO 2013016160 A1 WO2013016160 A1 WO 2013016160A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
membered
compound
cycloalkyl
Prior art date
Application number
PCT/US2012/047522
Other languages
French (fr)
Inventor
Zhaoyang Meng
M. Arshad Siddiqui
Panduranga Adulla P. Reddy
Original Assignee
Merck Sharp & Dohme Corp.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Sharp & Dohme Corp. filed Critical Merck Sharp & Dohme Corp.
Publication of WO2013016160A1 publication Critical patent/WO2013016160A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to certain Imidazopyrazine compounds of Formula (I) as inhibitors of mammalian Target Of Rapamycin (mTOR) kinase, which is also known as FRAP, RAFT, RAPT or SEP. The compounds may be used in the treatment of cancer and other disorders where mTOR is deregulated. The present invention further provides pharmaceutical compositions comprising the Imidazopyrazine compounds.

Description

TITLE OF THE INVENTION
NOVEL IMIDAZO[l ,2-A]PYRAZINE DERIVATIVES AS mTOR INHIBITORS
FIELD OF THE INVENTION
This invention is directed to certain Imidazopyrazine compounds of Formula
(I) as inhibitors of mammalian Target Of Rapamycin (mTOR) kinase, which is also known as FRAP, RAFT, RAPT or SEP. The compounds may be useful in the treatment of cancer and other disorders where mTOR is deregulated. BACKGROUND OF THE INVENTION
The mammalian target of rapamycin (mTOR) is a central regulator of cell growth and proliferation and plays a gatekeeper role in the control of cell cycle progression. The mTOR signaling pathway, which integrates both extracellular and intracellular signals, is activated in certain cellular processes such as tumor formation, angiogenesis, insulin resistance, adipogenesis, and T-lymphocyte activation. In addition, the mTOR signaling pathway is deregulated in diseases such as cancer and type 2 diabetes. See Laplante et al., J. Cell Science 122, pp 3589-3593 (2009).
mTOR mediates mitogenic signals from PI3K/AKT through to the downstream targets S6K1 (ribosomal S6 kinase 1), 4E-BP1 (eukaryotic translation initiation factor 4E- binding protein) and AKT. Recently, it has been shown that mTOR exists in two complexes.
Raptor-mTOR complex (mTORCl) is a rapamycin- sensitive complex that phosphorylates S6K1 and 4E-BP1. Rictor-mTOR complex (mTORC2) is a rapamycin-insensitive complex that phosphorylates AKT at Ser473. Although the precise mechanism by which rapamycin inhibits mTOR function is not well understood, rapamycin partially inhibits mTOR function through mTORCl . Since mTORC2 is involved in the regulation of cell survival, metabolism, proliferation, and cytoskeletal organization in a rapamycin-independent manner, complete inhibition of mTOR function through inhibition of both mTORCl and mTORC2 may lead to a broader spectrum antitumor activity in the treatment of cancer or better efficacy. In addition, inhibition of both mTORCl and mTORC2 may lead to better efficacy in treating other diseases than through inhibition of mTORCl alone.
There exists a need in the art for small-molecule compounds having desirable physicochemical properties that are useful for treating cancer and other disorders associated with deregulated mTOR activity. Specifically, there exists a need for small molecule inhibitors of mTOR kinase that block signaling through mTORCl and mTORC2 for treating cancer and other disorders.
SUMMARY OF THE INVENTION
The present invention relates to certain imidazopyrazine compounds of
Formula (I) as inhibitors of mammalian Target Of Rapamycin (mTOR) kinase, which is also known as FRAP, RAFT, RAPT or SEP. The compounds may be used in the treatment of cancer and other disorders where mTOR is deregulated. The present invention further provides pharmaceutical compositions comprising the imidazopyrazine compounds.
The present invention thus relates to compounds of Formula I and pharmaceutically acceptable salts thereof, as detailed herein:
Figure imgf000003_0001
I
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides Imidazopyrazine Compounds, pharmaceutical compositions comprising an Imidazopyrazine Compound, and methods of using the Imidazopyrazine Compounds for treating cancer in a patient. In addition, the present invention provides methods of using the Imidazopyrazine Compounds for treating a disease or disorder associated with deregulated mTOR activity in a patient.
COMPOUNDS
The present invention provides compounds of Formula I
Figure imgf000004_0001
I
U is N or CH; W is absent, or W is selected from the group consisting of C(O), S(O), S(0)2, C 1 -C4 alkylene,
C3-C8 cycloalkyl, phenyl, 5- to 6-membered heteroaryl, and 3- to 8-membered heterocyclyl;
V is absent, or V is selected from the group consisting of C(O), O, S, N(H), N(Ci-C3 alkyl), N(C3-C8 cycloalkyl), S(O), S(0)2, and C C4 alkylene;
or W and V together form a C3-C8 cycloalkyl, phenyl, 5- to 6-membered heteroaryl, or
3 to 8-membered heterocyclyl ring;
R1 is selected from the group consisting of
(i) Cj-C6 alkyl or C3-Cio cycloalkyl, wherein said alkyl or cycloalkyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of hydroxy, C!-C6 alkoxy, halo, trifluoromethyl, carboxy, 5- to 6-membered heteroaryl, -S02H, Ci-C6 alkyl-C(0)-NH-, Cj-C6 alkyl- S02-NH-, and Ci-C6 alkyl-SO-NH-;
(ii) 3- to 8-membered heterocyclyl wherein said heterocyclyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of halo, Ci-C6 alkyl, Ci-C6 alkoxy, cyano, hydroxy, amino, Cj-C6 alkylamino, and C)-C6 dialkylamino;
(iii) C6-Cio aryl or 5- to 10-membered heteroaryl, wherein said aryl or heteroaryl of R1 is unsubstituted or is substituted with one to three moieties independently selected from the group consisting of halo, trifluoromethyl, trifluoromethoxy, C]-C6 alkyl, C)-C6 alkoxy, cyano, hydroxy, amino, Ci-C6 alkylamino, and C]-C6 dialkylamino; and
(iv) -N(H)OH or -N(H)-C,-C3 alkoxy; L and Z are bonded to any two carbons of the ring and are independently selected from the group consisting of CH2, C(H)(R10),C(R10)(Rn), N(R10), C(O), 0, S, S(O) and S(0)2; T is not present such that L is bonded directly to Z, or T is selected from the group consisting of CH2, C(H)(R10),C(R,0)(R11), N(R10), C(O), O, S, S(O) and S(0)2 and d-C4 alkylene, wherein said alkylene of T is unsubstituted or substituted with 1 to two substituents selected from the group consisting of C1-C3 alkyl, halo, hydroxyl, C1-C3 alkoxy, amino, C1-C3 alkylamino and Cj-C3 dialkylamino;
Ra and Rb are independently selected from H, halogen and Ci-C6 alkyl;
R3 is selected from the group consisting of H, halogen, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halo-C]-C6alkyl, -CF3, -C(0)R9, C6-Ci0aryl, C3-C8cycloalkyl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclyl, 5- to 10-membered heterocyclenyl, C6-CioarylCi-
C6alkyl, C3-C8cycloalkylC]-C6alkyl, 5- to 10-membered heteroarylCi-C6alkyl, 5- to 10- membered heterocyclylC i-C6alkyl and 5- to 10-membered heterocyclenylC]-C6alkyl, wherein each of said aryl, cycloalkyl, heteroaryl, heterocyclyl, heterocyclenyl, arylalkyl,
cycloalkylalkyl, heteroarylalkyl, heterocyclylalkyl and heterocyclenylalkyl is unsubstituted or substituted with one to three moieties which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, C3-Cgcycloalkyl, -CF3, -CN, - C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -(CRaRb)nOR9, -(CRaRb)nC(0)R9, -(CRaRb)nNR8R9, - (CRaRb)nNR8,
-NR8R9, -(CRaRb)nC(0)0-C1-C6alkyl, -O-halod-Qalkyl, -(CRaRb)nC(0)NR8R9, -(CRaRb)n C(0)NR8S(0)2R9, -(CRaRb)nNR8C(0)R9, -(CRaRb)nNR8C(0)OR9,
-(CRaRb)nNR8C(0)NR8R9, -(CRaRb)nS(02)NR8R9, -(CRaRb)nS(02)NR8C(0)R9, -(CRaRb)n NR8S(02)R9, -(CRaRb)nSR9, -(CRaRb)nS(02)R9, C6-C,0aryl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclenyl, 5- to 10-membered heterocyclyl, C6-C]0arylalkyl, 5- to 10- membered heteroarylalkyl, 5- to 10-membered heterocyclenylalkyl and 5- to 10-membered heterocyclylalkyl wherein each of said alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, heterocyclenyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocyclylalkyl and
heterocyclenylalkyl is unsubstituted or substituted with one to five moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Ci- C6alkyl, C3-C8cycloalkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O-haloC,- Cealkyl, -OR9, -C(0)R9, -NR8R9, -C(0)0-C1-C6alkyl, -C(0)NR8R9, -NR8C(0)R9, - S(02)NR8R9, -NR8S(02)R9, -SR9, and -S(02)R9;
R6 is independently selected from the group consisting of H, halogen, Ci-C6alkyl, C3- Cscycloalkyl, -CF3, -CN, -(CRaRb)nC(0)OH, -OCF3, -OR9, -C(0)R9, -NR8R9, -C(0)0-Ci-
C6alkyl, -CRaRb, -ORa, -S(0)Ra, -C(0)ORa, -S(02)NRaRb, -NRaC(0)Rb, -NRaS(02)Rb,- C(0)NR8R9, -SR9, and -S(02)R9;
R8 and R9 are independently selected from the group consisting of H, OH, Ci-C6alkyl, C3- C8cycloalkyl, C6-Ci0aryl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclenyl,
5- to 10-membered heterocyclyl, CrCscycloalkyld-Cealkyl, Ce-CjoarylCi-Qalkyl, 5- to 10- membered heteroarylCi-C6alkyl, 5- to 10-membered heterocyclylCi-C6alkyl, 5- to 10- membered heterocyclenylCi-C6alkyl, and said alkyl, cycloalkyl, aryl, heteroaryl,
heterocyclenyl, heterocyclyl, cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclenylalkyl or heterocyclylalkyl is optionally substituted with halogen, Ci-C6alkyl, C3-C cycloalkyl, -CF3,
-CN, -(CRaRb)nC(0)OH, -OCF3, -ORa, -C(O), amino, -C(0)0-Ci-C6alkyl, -C(0)NRaRb,-SRa, and -S(02)Ra; or R8 and R9 together with the nitrogen atom to which they are attached form a 3- to 6-membered heterocyclyl ring; R10 and Rn are independently selected from the group consisting of H, Ci-C3alkyl, halo, hydroxyl, Ci-C3alkoxy, amino, C[-C3alkylamino and Ci-C3dialkylamino; n is independently 0, 1, 2, 3 or 4;
m is independently 0, 1 , 2, 3 or 4;
or a pharmaceutically acceptable salt thereof.
In one embodiment of the invention,
L and Z are both CH2> and T is not present; Ra and Rb are independently selected from H and Ci-C6 alkyl;
R is selected from the group consisting of C6-Ci0aryl, 5- to 10-membered heteroaryl, wherein each of said aryl or heteroaryl is unsubstituted or substituted with one to three moieties which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, C6-C10aryl, 5- to 10-membered heteroaryl, -CF3, -CN,
-C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloCi-C6alkyl, -OR8, -C(0)R8, -NR8R9,
-C(0)0-Ci-C6alkyl, -C(0)NR8R9, -NR8C(0)R9, -S(02)NR8R9, -NR8S(02)R9, -SR8, and -S(02)R8, wherein each of said heteroaryl or aryl is unsubstituted or substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, d-dalkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O- haloCi-Qalkyl, -ORa, -C(0)Ra, -NRaRb, -C(0)0-C C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, - S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra;
R6 is independently selected from the group consisting of H, -ORa, -NRaRb, -SRa, -S(0)Ra, - S(02)Ra, -C(0)Ci-C6alkyl, -C(0)NRaRb, -C(0)ORa, -S(02)NRaRb, -NRaC(0)Rb, - NRaS(02)Rb, d-C6alkyl, d-C6alkoxy, halogen, hydroxyl, amino and -CN; R8 and R9 are independently selected from the group consisting of H, OH, Ci-C6alkyl, C3-
Cgcycloalkyl, C6-Cioaryl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclenyl, 5- to 10-membered heterocyclyl, C3-C8cycloalkylCi-C6alkyl, C6-Ci0arylCi-C6alkyl, 5- to 10- membered heteroarylCi-C6alkyl, 5- to 10-membered heterocyclylCi-Cealkyl, 5- to 10- membered heterocyclenylC]-C6alkyl, and said alkyl, cycloalkyl, aryl, heteroaryl,
heterocyclenyl, heterocyclyl, cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclenylalkyl or heterocyclylalkyl is optionally substituted with halogen, Ci-C6alkyl, C3-C8cycloalkyl, -CF3, -CN, -(CRaRb)nC(0)OH, -OCF3, -ORa, -C(O), amino, -C(0)0-C,-C6alkyl, -C(0)NRaRb,-SRa, and -S(02)Ra; n is independently 0, 1 or 2;
m is 1 ; or a pharmaceutically acceptable salt thereof. In another aspect of the foregoing embodiments, W is C(O).
In another aspect of the foregoing embodiments, V is absent. In yet another aspect of the foregoing embodiments, R1 is selected from the group consisting of :
(i) Ci-C6 alkyl or C3-C10 cycloalkyl, wherein said alkyl or cycloalkyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of hydroxy, Ci-C6 alkoxy, fluoro, trifluoromethyl, carboxy, tetrazolyl, -S02H, Ci-
C6 alkyl-C(0)-NH-, CrC6 alkyl-S02-NH-, and CrC6 alkyl-SO-NH-; and
(ii) 5- to 6-menibered heterocyclyl containing 1 to 3 heteroatoms selected from the group consisting of N, O, S, and S(0)2 wherein said heterocyclyl of R1 is unsubstituted or substituted with one to two moieties independently selected from the group consisting of halo, Ci-C6 alkyl, Ci-C6 alkoxy, cyano, hydroxy, amino, Ci-C6 alkylamino, and Ci-C6
dialkylamino.
In one embodiment, R1 is selected from the group consisting of :
(i) Ci-C6 alkyl, wherein said alkyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of hydroxy, C C6 alkoxy, fluoro,; and
(ii) triazol unsubstituted or substituted with one to two moieties independently selected from the group consisting of halo, C C6 alkyl, Q-Q alkoxy, cyano, hydroxy, amino, Ci-C6 alkylamino, and Ci-C6 dialkylamino.
In another aspect of the foregoing embodiments,
R is a 5- to 6-membered heteroaryl or phenyl unsubstituted or substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, C]-C6alkyl, phenyl, 5- to 6-membered heteroaryl, -CF3, -CN,
-C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O-halod-Cealkyl, -ORa, -C(0)Ra, -NRaRb,
-C(0)0-C!-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra, wherein the alkyl, phenyl or heteroaryl is optionally substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O- haloCrC6alkyl, -ORa, -C(0)Ra, -NRaRb, -C(0)0-Ci-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, - S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra;
Wherein all other substituents are as defined in any one of claims 1 to 7.
In one embodiment, R3 is pyrazolyl, isoquinolinyl, pyrimidinyl, phenyl or pyridyl, unsubstituted or substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, phenyl, 5- to 6-membered heteroaryl, -CF3, -CN,
-C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloC,-C6alkyl, -ORa, -C(0)Ra, -NRaRb,
-C(0)0-C,-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra, wherein the alkyl, phenyl or heteroaryl is optionally substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, d-Qalkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O- haloCrCgalkyl, -ORa, -C(0)Ra, -NRaRb, -C(0)0-C1-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, - S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra.
In another embodiment, R3 is
Figure imgf000009_0001
Ar is phenyl or a 5- to 6-membered heteroaryl optionally substituted with one to three of R , which can be the same or different, each R12 being selected from the group consisting of halogen, C C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloC]-C6alkyl, -
ORa, -C(0)Ra, -NRaRb, -C(0)0-Ci-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -
NRaS(02)Rb, -SRa, and -S(02)Ra.
In one embodiment, Ar1 is phenyl, pyridyl, pyrimidinyl,
imidazolyl, pyrazinyl, pyrazolyl, or thiazolyl, optionally substituted with one to three of R12, which can be the same or different, each R being selected from the group consisting of halogen, Ci-C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloCrC6alkyl, -
ORa, -C(0)Ra, -NRaRb, -C(0)0-C,-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -
NRaS(02)Rb, -SRa, and -S(02)Ra.
In another embodiment, R is selected from the group consisting of halogen, Ci-C6alkyl, -
CF3, and -OCF3.
In one embodiment, R3 is pyrazolyl, isoquinolinyl, pyrimidinyl, phenyl or pyridyl, unsubstituted or substituted with one to three moieties as defined above.
In one embodiment, R3 is unsubstituted or substituted pyridyl or pyrazolyl as defined above.
In another embodiment, R is
Figure imgf000010_0001
Ar1 is C6-Ci0aryl or a 5- to 10-membered heteroaryl optionally substituted with one to three of R12, which can be the same or different, each R12 being selected from the group consisting of halogen, Ci-C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O-halod-Cealkyl, - ORa, -C(0)Ra, -NRaRb, -C(0)0-Ci-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -
NRaS(02)Rb, -SRa, and -S(02)Ra. In one embodiment, Ar1 is phenyl or a 5- to 6-membered heteroaryl optionally substituted.
In a another embodiment, R3 is
Figure imgf000010_0002
, wherein R8 and R9 are as defined above. In one embodiment, R8 is H and R9 is cyclopropyl.
In a further embodiment, Ar1 is phenyl, pyrazolyl, pyrimidinyl, pyridyl, imidazolyl, pyrazinyl or thiazolyl optionally substituted with one to three of R12. In a another
embodiment, Ar1 is phenyl, pyridyl or imidazolyl optionally substituted with one to three of
12
R
In another embodiment, R3 is
Figure imgf000010_0003
Ar is is phenyl , pyridyl, pyrazinyl or imidazolyl optionally substituted with one to three of R as defined above.
In another embodiment, R3 is
Figure imgf000010_0004
Ar is is phenyl , pyridyl, pyrazinyl or imidazolyl optionally substituted with one to three of R as defined above. In another embodiment, R is selected from the group consisting
Figure imgf000011_0001
Figure imgf000011_0002
is defined above.
In one embodiment, in the foregoing embodiments, R is selected from the group
consisting of halogen, Ci-C6alkyl, -CF3, and -OCF3. In another embodiment, R12 is selected from the group consisting of F and methyl.
In one embodiment, R6 is independently selected from the group consisting of
-C(0)Ci-C6alkyl, -S(0)2Ci-C6alkyl and CN; and all other substituents are as defined above. In another aspect of the foregoing embodiments, R6 is H or amino.
Specific embodiments depicting non-limiting Examples of of the above Formulas are provided in the Experimental Section hereinbelow.
Specific examples of the compounds of the instant invention include:
(( 1 R,3 s,5 S)-3 -(8-amino-3 -(6-phenylpyridin-3 -yl)imidazo [ 1 ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octan-8-yl)(4H-l ,2,4-triazol-3-yl)methanone;
((lR,3r,5S)-3-(8-amino-3-(6-phenylpyridin-3-yl)imidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2. l]octan-8-yl)(4H-l ,2,4-triazol-3-yl)methanone;
l-((lR,3s,5S)-3-(7-amino-6-fluoro-3-(6-phenylpyridin-3-yl)pyrazolo[l ,5-a]pyrimidin-5-yl)-8- azabicyclo[3.2.1]octan-8-yl)-2-hydroxyethanone; (( 1 R,3 s, 5 S)-3 -(8-amino-3 -(6-( 1 -methyl- 1 H-pyrazol-3 -yl)pyridin-3 -yl)imidazo [ 1 ,2-a]pyrazin- 6-yl)-8-azabicyclo[3.2.1]octan-8-yl)(4H-l ,2,4-triazol-3-yl)methanone;
((lR,3s,5S)-3-(8-amino-3-(l-phenyl-lH-pyrazol-4-yl)imidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octan-8-yl)(4H-l,2,4-triazol-3-yl)methanone;
1 -(( 1 R,3 s,5 S)-3-(3 -(6-( 1 H-imidazol-2-yl)pyridin-3-yl)-8-aminoimidazo[ 1 ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1 ]octan-8-yl)-2-hydroxyethanone; and
((lR,3s,5S)-3-(3-(6-(lH-imidazol-2-yl)pyridin-3-yl)-8-aminoimidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octan-8-yl)(4H-l,2,4-triazol-3-yl)methanone; Or a stereoisomer thereof;
Or a pharmaceutically acceptable salt thereof;
Or a pharmaceutically acceptable salt of the stereoisomer thereof.
Chemical Definitions
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, Ci-Cio, as in "Ci-Cio alkyl" is defined to include groups having 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbons in a linear or branched arrangement. For example, "Ci -Cio alkyl" specifically includes methyl, ethyl, ^-propyl, /-propyl, «-butyl, t-butyl, /-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, and so on.
When used in the phrases "alkylaryl", "alkylcycloalkyl" and
"alkylheterocyclyl" the term "alkyl" refers to the alkyl portion of the moiety and does not describe the number of atoms in the heterocyclyl portion of the moiety. In an embodiment, if the number of carbon atoms is not specified, the "alkyl" of "alkylaryl", "alkylcycloalkyl" and "alkylheterocyclyl" refers to C1-C12 alkyl and in a further embodiment, refers to C1-C6 alkyl.
The term "cycloalkyl" means a monocyclic saturated or unsaturated aliphatic hydrocarbon group having the specified number of carbon atoms. The cycloalkyl is optionally bridged (i.e., forming a bicyclic moiety), for example with a methylene, ethylene or propylene bridge. The cycloalkyl may be fused with an aryl group such as phenyl, and it is understood that the cycloalkyl substituent is attached via the cycloalkyl group. For example, "cycloalkyl" includes cyclopropyl, methyl-cyclopropyl, 2,2-dimethyl-cyclobutyl, 2-ethyl-cyclopentyl, cyclohexyl, cyclopentenyl, cyclobutenyl and so on. In an embodiment, if the number of carbon atoms is not specified, "alkyl" refers to C1-C12 alkyl and in a further embodiment, "alkyl" refers to C1-C6 alkyl. In an embodiment, if the number of carbon atoms is not specified, "cycloalkyl" refers to C3-C10 cycloalkyl and in a further embodiment, "cycloalkyl" refers to C3-C7 cycloalkyl. In an embodiment, examples of "alkyl" include methyl, ethyl, n-propyl, /-propyl, «-butyl, t-butyl and -butyl.
The term "alkylene" means a hydrocarbon diradical group having the specified number of carbon atoms. For example, "alkylene" includes -CH2-, -CH2CH2- and the like. In an embodiment, if the number of carbon atoms is not specified, "alkylene" refers to C1-C12 alkylene and in a further embodiment, "alkylene" refers to C1-C6 alkylene.
If no number of carbon atoms is specified, the term "alkenyl" refers to a non- aromatic hydrocarbon radical, straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon double bond. Preferably one carbon to carbon double bond is present, and up to four non-aromatic carbon-carbon double bonds may be present. Thus, "C2-C6 alkenyl" means an alkenyl radical having from 2 to 6 carbon atoms. Alkenyl groups include ethenyl, propenyl, butenyl, 2-methylbutenyl and cyclohexenyl. The straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated.
The term "alkynyl" refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 10 carbon atoms and at least one carbon to carbon triple bond. Up to three carbon-carbon triple bonds may be present. Thus, "C2-C6 alkynyl" means an alkynyl radical having from 2 to 6 carbon atoms. Alkynyl groups include ethynyl, propynyl, butynyl, 3-methylbutynyl and so on. The straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
In certain instances, substituents may be defined with a range of carbons that includes zero, such as (Co-C6)alkylene-aryl. If aryl is taken to be phenyl, this definition would include phenyl itself as well as -CH2PI1, -CH2CH2PI1, CH(CH3)CH2CH(CH3)Ph, and so on.
"Aryl" is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 7 atoms in each ring, wherein at least one ring is aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl and biphenyl. In cases where the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring. In one embodiment, "aryl" is an aromatic ring of 6 to 14 carbons atoms, and includes a carbocyclic aromatic group fused with a 5-or 6-membered cycloalkyl group such as indan. Examples of carbocyclic aromatic groups include, but are not limited to, phenyl, naphthyl, e.g. 1-naphthyl and 2-naphthyl; anthracenyl, e.g. 1 -anthracenyl, 2-anthracenyl; phenanthrenyl; fluorenonyl, e.g. 9-fluorenonyl, indanyl and the like.
The term heteroaryl, as used herein, represents a stable monocyclic, bicyclic or tricyclic ring of up to 7 atoms in each ring, wherein at least one ring is aromatic and contains carbon and from 1 to 4 heteroatoms selected from the group consisting of O, N and S. In another embodiment, the term heteroaryl refers to a monocyclic, bicyclic or tricyclic aromatic ring of 5- to 14-ring atoms of carbon and from one to four heteroatoms selected from O, N, or
S. As with the definition of heterocycle below, "heteroaryl" is also understood to include the N-oxide derivative of any nitrogen-containing heteroaryl. In cases where the heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively.
Heteroaryl groups within the scope of this definition include but are not limited to acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, benzotriazolyl, furanyl, thienyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline. Additional examples of heteroaryl include, but are not limited to pyridyl, e.g., 2-pyridyl (also referred to as a-pyridyl), 3 -pyridyl (also referred to as β -pyridyl) and 4-pyridyl (also referred to as (γ- pyridyl); thienyl, e.g., 2-thienyl and 3-thienyl; furanyl, e.g., 2-furanyl and 3-furanyl;
pyrimidyl, e.g., 2-pyrimidyl and 4-pyrimidyl; imidazolyl, e.g., 2-imidazolyl; pyranyl, e.g., 2- pyranyl and 3-pyranyl; pyrazolyl, e.g., 4-pyrazolyl and 5-pyrazolyl; thiazolyl, e.g., 2-thiazolyl, 4-thiazolyl and 5-thiazolyl; thiadiazolyl; isothiazolyl; oxazolyl, e.g., 2-oxazoyl, 4-oxazoyl and 5-oxazoyl; isoxazoyl; pyrrolyl; pyridazinyl; pyrazinyl and the like.
In an embodiment, "heteroaryl" may also include a "fused polycyclic aromatic", which is a heteroaryl fused with one or more other heteroaryl or nonaromatic heterocyclic ring. Examples include, quinolinyl and isoquinolinyl, e.g. 2-quinolinyl, 3- quinolinyl, 4-quinolinyl, 5-quinolinyl, 6-quinolinyl, 7-quinolinyl and 8-quinolinyl, 1- isoquinolinyl, 3-quinolinyl, 4-isoquinolinyl, 5-isoquinolinyl, 6-isoquinolinyl, 7-isoquinolinyl and 8-isoquinolinyl; benzofuranyl, e.g. 2 -benzofuranyl and 3 -benzofuranyl; dibenzofuranyl, e.g. 2,3-dihydrobenzofuranyl; dibenzothiophenyl; benzothienyl, e.g. 2-benzothienyl and 3- benzothienyl; indolyl, e.g. 2-indolyl and 3-indolyl; benzothiazolyl, e.g., 2-benzothiazolyl; benzooxazolyl, e.g., 2-benzooxazolyl; benzimidazolyl, e.g. 2-benzoimidazolyl; isoindolyl, e.g.
1-isoindolyl and 3-isoindolyl; benzotriazolyl; purinyl; thianaphthenyl, pyrazinyland the like.
"Heterocyclyl" means a non-aromatic saturated monocyclic, bicyclic, tricyclic or spirocyclic ring system comprising up to 7 atoms in each ring. Preferably, the heterocyclyl contains 3 to 14, or 5 to 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example, nitrogen, oxygen, phosphor or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system.
Preferred heterocyclyls contain about 5 to about 6 ring atoms. The heterocycle may be fused with an aromatic aryl group such as phenyl or heterocyclenyl. The prefix aza, oxa or thia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom, respectively, is present as a ring atom. The nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide. Non-limiting examples of suitable monocyclic heterocyclyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, lactam, lactone, and the like. "Heterocyclyl" also includes heterocyclyl rings as described above wherein =0 replaces two available hydrogens on the same ring carbon atom. An example of such a moiety is pyrrolidone:
Figure imgf000015_0001
In describing the heteroatoms contained in a specified heterocyclyl group, the expression, "having one to x heteroatoms selected from the group of N, O, P and S" (wherein x is an a specified integer), for example, means that each heteroatom in the specified heterocyclyl is independently selected from the specified selection of heteroatoms.
Attachment of a heterocyclyl substituent can occur via a carbon atom or via a heteroatom.
"Heterocyclenyl" means a non-aromatic monocyclic, bicyclic, tricyclic or spirocyclic ring system comprising up to 7 atoms in each ring. Preferably, the heterocyclenyl contains 3 to 14, or 5 to 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur atom, alone or in combination, and which contains at least one carbon-carbon double bond or carbon-nitrogen double bond. There are no adjacent oxygen and/or sulfur atoms present in the ring system. Preferred heterocyclenyl rings contain about 5 to about 6 ring atoms. The prefix aza, oxa or thia before the heterocyclenyl root name means that at least a nitrogen, oxygen, phosphor or sulfur atom respectively is present as a ring atom. The nitrogen or sulfur atom of the heterocyclenyl can be optionally oxidized to the corresponding N-oxide, S -oxide or S,S- dioxide. Non-limiting examples of suitable heterocyclenyl groups include 1,2,3,4- tetrahydropyridinyl, 1 ,2-dihydropyridinyl, 1 ,4-dihydropyridinyl, 1 ,2,3,6-tetrahydropyridinyl,
1,4,5,6-tetrahydropyrimidinyl, 2-pyrrolinyl, 3-pyrrolinyl, 2-imidazolinyl, 2-pyrazolinyl, dihydroimidazolyl, dihydrooxazolyl, dihydrooxadiazolyl, dihydrothiazolyl, 3,4-dihydro-2H- pyranyl, dihydrofuranyl, fluorodihydrofuranyl, 7-oxabicyclo[2.2.1]heptenyl,
dihydrothiophenyl, dihydrothiopyranyl, and the like. "Heterocyclenyl" also includes heterocyclenyl rings as described above wherein =0 replaces two available hydrogens on the same ring carbon atom. An example of such a moiety is pyrrolidinone:
Figure imgf000016_0001
In describing the heteroatoms contained in a specified heterocyclenyl group, the expression, "having one to x heteroatoms selected from the group of N, O, P and S" (wherein x is an a specified integer), for example, means that each heteroatom in the specified heterocyclenyl is independently selected from the specified selection of heteroatoms.
It should also be noted that tautomeric forms such as, for example, the moieties:
Figure imgf000016_0002
are considered equivalent in certain embodiments of this invention.
An "alkylaryl group" is an alkyl group substituted with an aryl group, for example, a phenyl group. Suitable aryl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the aryl group.
An "alkylheteroaryl group" is an alkyl group substituted with a heteroaryl group. Suitable heteroaryl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the heteroaryl group. An "alkylheterocyclyl group" is an alkyl group substituted with a heterocyclyl group. Suitable heterocyclyl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the heterocyclyl group.
An "alkylheterocyclenyl group" is an alkyl group substituted with a heterocyclenyl group. Suitable heterocyclenyl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the heterocyclenyl group.
An "alkylcycloalkyl group" is an alkyl group substituted with a cycloalkyl group. Suitable cycloalkyl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the cycloalkyl group.
An "arylalkyl group" is an aryl group substituted with an alkyl group, for example, a phenyl group. Suitable aryl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the alkyl group.
A "heteroarylalkyl group" is a heteroaryl group substituted with an alkyl group. Suitable heteroaryl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the alkyl group.
A "heterocyclylalkyl group" is a heterocyclyl group substituted with an alkyl group. Suitable heterocyclyl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the alkyl group.
A "heterocyclenylalkyl group" is a heterocyclenyl group substituted with an alkyl group. Suitable heterocyclenyl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the alkyl group.
A "cycloalkylalkyl group" is a cycloalkyl group substituted with an alkyl group. Suitable cycloalkyl groups are described herein and suitable alkyl groups are described herein. The bond to the parent moiety is through the alkyl group.
An "aryloxy group" is an aryl group that is attached to a compound via an oxygen (e.g., phenoxy).
An "alkoxy group" (alkyloxy), as used herein, is a straight chain or branched C]-Ci2 or cyclic C3-C12 alkyl group that is connected to a compound via an oxygen atom. Examples of alkoxy groups include but are not limited to methoxy, ethoxy and propoxy.
An "arylalkoxy group" (arylalkyloxy) is an arylalkyl group that is attached to a compound via an oxygen on the alkyl portion of the arylalkyl (e.g., phenylmethoxy).
An "arylamino group" as used herein, is an aryl group that is attached to a compound via a nitrogen. An "alkylamino group" as used herein, is an alkyl group that is attached to a compound via a nitrogen.
As used herein, an "arylalkylamino group" is an arylalkyl group that is attached to a compound via a nitrogen on the alkyl portion of the arylalkyl.
An "alkylsulfonyl group" as used herein, is an alkyl group that is attached to a compound via the sulfur of a sulfonyl group.
When a moiety is referred to as "unsubstituted" or not referred to as
"substituted" or "optionally substituted", it means that the moiety does not have any substituents. When a moiety is referred to as substituted, it denotes that any portion of the moiety that is known to one skilled in the art as being available for substitution can be substituted. The phrase "optionally substituted with one or more substituents" means, in one embodiment, one substituent, two substituents, three substituents, four substituents or five substituents. For example, the substitutable group can be a hydrogen atom that is replaced with a group other than hydrogen (i.e., a substituent group). Multiple substituent groups can be present. When multiple substituents are present, the substituents can be the same or different and substitution can be at any of the substitutable sites. Such means for substitution are well known in the art. For purposes of exemplification, which should not be construed as limiting the scope of this invention, some examples of groups that are substituents are: alkyl, alkenyl or alkynyl groups (which can also be substituted, with one or more substituents), alkoxy groups (which can be substituted), a halogen or halo group (F, CI, Br, I), hydroxy, nitro, oxo, -CN, -COH, -COOH, amino, azido, N-alkylamino or Ν,Ν-dialkylamino (in which the alkyl groups can also be substituted), N-arylamino or Ν,Ν-diarylamino (in which the aryl groups can also be substituted), esters (-C(O)-OR, where R can be a group such as alkyl, aryl, etc., which can be substituted), ureas (-NHC(O)-NHR, where R can be a group such as alkyl, aryl, etc., which can be substituted), carbamates (-NHC(O)-OR, where R can be a group such as alkyl, aryl, etc., which can be substituted), sulfonamides (-NHS(0)2R, where R can be a group such as alkyl, aryl, etc., which can be substituted), alkylsulfonyl (which can be substituted), aryl (which can be substituted), cycloalkyl (which can be substituted) alkylaryl (which can be substituted), alkylheterocyclyl (which can be substituted), alkylcycloalkyl (which can be substituted), and aryloxy.
In the compounds of generic Formula I, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of generic Formula I. For example, different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds within generic Formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
Certain isotopically-labelled compounds of Formula (I) (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon- 14 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Certain isotopically-labelled compounds of Formula (I) can be useful for medical imaging purposes. For instance those compounds labeled with positron- emitting isotopes like nC or 18F can be useful for application in Positron Emission
Tomography (PET) and those labeled with gamma ray emitting isotopes like 123I can be useful for application in Single Photon Emission Computed Tomography (SPECT). Additionally, isotopic substitution of a compound at a site where epimerization occurs may slow or reduce the epimerization process and thereby retain the more active or efficacious form of the compound for a longer period of time.
It is also possible that the compounds of Formula (I) may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. Also, for example, all keto-enol and imine-enamine forms of the compounds are included in the invention.
Stereochemistry
When structures of the same constitution differ in respect to the spatial arrangement of certain atoms or groups, they are stereoisomers, and the considerations that are significant in analyzing their interrelationships are topological. If the relationship between two stereoisomers is that of an object and its nonsuperimposable mirror image, the two structures are enantiomeric, and each structure is said to be chiral. Stereoisomers also include diastereomers, cis-trans isomers and conformational isomers. Diastereoisomers can be chiral or achiral, and are not mirror images of one another. Cis-trans isomers differ only in the positions of atoms relative to a specified planes in cases where these atoms are, or are considered as if they were, parts of a rigid structure. Conformational isomers are isomers that can be interconverted by rotations about formally single bonds. Examples of such
conformational isomers include cyclohexane conformations with chair and boat conformers, carbohydrates, linear alkane conformations with staggered, eclipsed and gauche confomers, etc. See J. Org. Chem. 35, 2849 (1970)
Many organic compounds exist in optically active forms having the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, enantiomers are identical except that they are non-superimposable mirror images of one another. A mixture of enantiomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture. Many of the compounds described herein can have one or more chiral centers and therefore can exist in different enantiomeric forms. If desired, a chiral carbon can be designated with an asterisk (*). When bonds to the chiral carbon are depicted as straight lines in the Formulas of the invention, it is understood that both the (R) and (S) configurations of the chiral carbon, and hence both enantiomers and mixtures thereof, are embraced within the Formula. As is used in the art, when it is desired to specify the absolute configuration about a chiral carbon, one of the bonds to the chiral carbon can be depicted as a wedge (bonds to atoms above the plane) and the other can be depicted as a series or wedge of short parallel lines is (bonds to atoms below the plane). The Cahn-Inglod-Prelog system can be used to assign the (R) or (S) configuration to a chiral carbon.
When the compounds of the present invention contain one chiral center, the compounds exist in two enantiomeric forms and the present invention includes both enantiomers and mixtures of enantiomers, such as the specific 50:50 mixture referred to as a racemic mixtures. The enantiomers can be resolved by methods known to those skilled in the art, such as formation of diastereoisomeric salts which may be separated, for example, by crystallization (see, CRC Handbook of Optical Resolutions via Diastereomeric Salt Formation by David Kozma (CRC Press, 2001)); formation of diastereoisomeric derivatives or complexes which may be separated, for example, by crystallization, gas-liquid or liquid chromatography; selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic esterification; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support for example silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where the desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step is required to liberate the desired enantiomeric form. Alternatively, specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
Designation of a specific absolute configuration at a chiral carbon of the compounds of the invention is understood to mean that the designated enantiomeric form of the compounds is in enantiomeric excess (ee) or in other words is substantially free from the other enantiomer. For example, the "R" forms of the compounds are substantially free from the "S" forms of the compounds and are, thus, in enantiomeric excess of the "S" forms.
Conversely, "S" forms of the compounds are substantially free of "R" forms of the compounds and are, thus, in enantiomeric excess of the "R" forms. Enantiomeric excess, as used herein, is the presence of a particular enantiomer at greater than 50%. In a particular embodiment when a specific absolute configuration is designated, the enantiomeric excess of depicted compounds is at least about 90%.
When a compound of the present invention has two or more chiral carbons it can have more than two optical isomers and can exist in diastereoisomeric forms. For example, when there are two chiral carbons, the compound can have up to 4 optical isomers and 2 pairs of enantiomers ((S,S)/(R,R) and (R,S)/(S,R)). The pairs of enantiomers (e.g., (S,S)/(R,R)) are mirror image stereoisomers of one another. The stereoisomers that are not mirror-images (e.g., (S,S) and (R,S)) are diastereomers. The diastereoisomeric pairs may be separated by methods known to those skilled in the art, for example chromatography or crystallization and the individual enantiomers within each pair may be separated as described above. The present invention includes each diastereoisomer of such compounds and mixtures thereof.
As used herein, "a," an" and "the" include singular and plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an active agent" or "a pharmacologically active agent" includes a single active agent as well a two or more different active agents in combination, reference to "a carrier" includes mixtures of two or more carriers as well as a single carrier, and the like. This invention is also intended to encompass pro-drugs of the imidazopyrazine compounds disclosed herein. A prodrug of any of the compounds can be made using well- known pharmacological techniques. Pharmaceutically acceptable salts
The imidazopyrazine compounds described herein can, as noted above, be prepared in the form of their pharmaceutically acceptable salts. Pharmaceutically acceptable salts are salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects. Examples of such salts are (a) acid addition salts organic and inorganic acids, for example, acid addition salts which may, for example, be hydrochloric acid, sulphuric acid, methanesulphonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, oxalic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid, trifluoroacetic acid, formic acid and the like. Pharmaceutically acceptable salts can also be prepared from by treatment with inorganic bases, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like. Pharmaceutically acceptable salts can also be formed from elemental anions such as chlorine, bromine and iodine.
The active compounds disclosed can, as noted above, also be prepared in the form of their hydrates. The term "hydrate" includes but is not limited to hemihydrate, monohydrate, dihydrate, trihydrate, tetrahydrate and the like.
The active compounds disclosed can, as noted above, also be prepared in the form of a solvate with any organic or inorganic solvent, for example alcohols such as methanol, ethanol, propanol and isopropanol, ketones such as acetone, aromatic solvents and the like.
The active compounds disclosed can also be prepared in any solid or liquid physical form. For example, the compound can be in a crystalline form, in amorphous form, and have any particle size. Furthermore, the compound particles may be micronized, or may be agglomerated, particulate granules, powders, oils, oily suspensions or any other form of solid or liquid physical form.
The compounds of the present invention may also exhibit polymorphism. This invention further includes different polymorphs of the compounds of the present invention. The term "polymorph" refers to a particular crystalline state of a substance, having particular physical properties such as X-ray diffraction, IR spectra, melting point, and the like. As used herein, "a," an" and "the" include singular and plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an active agent" or "a pharmacologically active agent" includes a single active agent as well a two or more different active agents in combination, reference to "a carrier" includes mixtures of two or more carriers as well as a single carrier, and the like.
METHODS OF TREATMENT
The Imidazopyrazine Compounds may be useful in human and veterinary medicine in the therapy of proliferative diseases such as cancer, autoimmune diseases, viral diseases, fungal diseases, neurological/neurodegenerative disorders, arthritis, inflammation, anti-proliferative (e.g., ocular retinopathy), neuronal, alopecia and cardiovascular disease.
Many of these diseases and disorders are listed in U.S. 6,413,974, which is hereby
incorporated by reference.
While not being bound by any specific theory it is believed that the
Imidazopyrazine Compounds may be useful in the treatment of proliferative diseases such as cancer, autoimmune diseases, viral diseases, fungal diseases, neurological/neurodegenerative disorders, arthritis, inflammation, anti-proliferative (e.g., ocular retinopathy), neuronal, alopecia and cardiovascular disease because of their mTOR inhibitory activity.
The general value of the compounds of the invention in inhibiting mTOR can be determined, for example, using the assay described in Example 3. In addition, the general value in inhibiting mTORCl or mTORC2 function can be evaluated using the assays described in Example 4.
More specifically, the Imidazopyrazine Compounds can be useful in the treatment of a variety of cancers, including (but not limited to) the following:
tumor of the bladder, breast (including BRCA-mutated breast cancer), colorectal, colon, kidney, liver, lung, small cell lung cancer, non-small cell lung cancer, head and neck, esophagus, bladder, gall bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin, including squamous cell carcinoma;
leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T- cell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, mantle cell lymphoma, myeloma and Burkett's lymphoma;
chronic lymphocytic leukemia ("CLL"),
acute and chronic myelogenous leukemia, myelodysplastic syndrome and promyelocytic leukemia; fibrosarcoma, rhabdomyosarcoma;
head and neck, mantle cell lymphoma, myeloma;
astrocytoma, neuroblastoma, glioma, glioblastoma, malignant glial tumors, astrocytoma, hepatocellular carcinoma, gastrointestinal stromal tumors ("GIST") and schwannomas;
melanoma, multiple myeloma, seminoma, teratocarcinoma, osteosarcoma, xenoderma pigmentosum, keratoctanthoma, thyroid follicular cancer, endometrial cancer, gastrointestinal tract cancer and Kaposi's sarcoma.
While not being bound by any specific theory, due to the key role of kinases in the regulation of cellular proliferation in general, inhibitors of kinases could act as reversible cytostatic agents which may be useful in the treatment of any disease process which features abnormal cellular proliferation, e.g., benign prostate hyperplasia, familial adenomatosis polyposis, neuro-fibromatosis, atherosclerosis, pulmonary fibrosis, arthritis, psoriasis, glomerulonephritis, restenosis following angioplasty or vascular surgery, hypertrophic scar formation, inflammatory bowel disease, transplantation rejection, endotoxic shock, and fungal infections.
The Imidazopyrazine Compounds may induce or inhibit apoptosis. The apoptotic response is aberrant in a variety of human diseases. The Imidazopyrazine .
Compounds, as modulators of apoptosis, can be useful in the treatment of cancer (including but not limited to those types mentioned hereinabove), viral infections (including, but not limited to, herpeviras, poxvirus, Epstein- Barr virus, Sindbis virus and adenovirus), prevention of AIDS development in HIV-infected individuals, autoimmune diseases (including but not limited to systemic lupus, erythematosus, autoimmune mediated glomerulonephritis, rheumatoid arthritis, psoriasis, inflammatory bowel disease, and autoimmune diabetes mellitus), neurodegenerative disorders (including but not limited to Alzheimer's disease, AIDS-related dementia, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, spinal muscular atrophy and cerebellar degeneration), myelodysplastic syndromes, aplastic anemia, ischemic injury associated with myocardial infarctions, stroke and reperfusion injury, arrhythmia, atherosclerosis, toxin-induced or alcohol related liver diseases, hematological diseases (including but not limited to chronic anemia and aplastic anemia), degenerative diseases of the musculoskeletal system (including but not limited to osteoporosis and arthritis) aspirin-sensitive rhinosinusitis, cystic fibrosis, multiple sclerosis, kidney diseases and cancer pain.
While not being bound by any specific theory, the Imidazopyrazine
Compounds, as inhibitors of kinases, can modulate the level of cellular RNA and DNA synthesis. These compounds would therefore be useful in the treatment of viral infections (including but not limited to HIV, human papilloma virus, herpesvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus).
In particular embodiments of the invention, Imidazopyrazine Compounds, as inhibitors of mTOR kinase could act in diseases or disorders other than cancer that are associated with dysregulated mTOR activity such as viral infections (including, but not limited to, herpevirus, poxvirus, Epstein- Barr virus, Sindbis virus and adenovirus), prevention of AIDS development in HIV-infected individuals, autoimmune diseases (including but not limited to systemic lupus, erythematosus, autoimmune mediated glomerulonephritis, rheumatoid arthritis, psoriasis, inflammatory bowel disease, and autoimmune diabetes mellitus), neurodegenerative disorders (including but not limited to Alzheimer's disease, AIDS-related dementia, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, spinal muscular atrophy and cerebellar degeneration), myelodysplastic syndromes, aplastic anemia, ischemic injury associated with myocardial infarctions, stroke and reperfusion injury, arrhythmia, atherosclerosis, toxin-induced or alcohol related liver diseases, hematological diseases (including but not limited to chronic anemia and aplastic anemia), degenerative diseases of the musculoskeletal system (including but not limited to osteoporosis and arthritis) aspirin-sensitive rhinosinusitis, cystic fibrosis, multiple sclerosis, kidney diseases and cancer pain. .
The Imidazopyrazine Compounds may also be useful in the chemoprevention of cancer. Chemoprevention is defined as inhibiting the development of invasive cancer by either blocking the initiating mutagenic event or by blocking the progression of pre-malignant cells that have already suffered an insult or inhibiting tumor relapse.
The Imidazopyrazine Compounds may also be useful in inhibiting tumor angiogenesis and metastasis.
Another aspect of this invention is a method of treating a patient (e.g., human) having a disease or condition associated with mTOR kinases by administering a
therapeutically effective amount of an Imidazopyrazine Compound, or a pharmaceutically acceptable salt of said compound to the patient.
The present invention provides a method of treating cancer comprising the step of administering to a subject a therapeutically effective amount of the Imidazopyrazine Compounds. The present invention also provides the Use of the Imidazopyrazine Compounds for the preparation of a medicament for the treatment of cancer. The invention also provides the Imidazopyrazine Compounds for use in the treatment of cancer. In the therapies described above, an example dosage for administration to a patient is about 0.001 to 1000 mg/kg of body weight/day of the Imidazopyrazine Compound. Another example dosage is about 0.01 to 25 mg/kg of body weight/day of the Imidazopyrazine Compound, or a pharmaceutically acceptable salt of said compound.
The dosage regimen utilizing the compounds of the present invention can be selected in accordance with a variety of factors including type, species, age, weight, sex and the type of cancer being treated; the severity (i.e., stage) of the disease to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to treat, for example, to prevent, inhibit (fully or partially) or arrest the progress of the disease.
Definitions:
As used herein, the term "therapeutically effective amount" means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. The therapeutic effect is dependent upon the disease or disorder being treated or the biological effect desired. As such, the therapeutic effect can be a decrease in the severity of symptoms associated with the disease or disorder and/or inhibition (partial or complete) of progression of the disease. The amount needed to elicit the therapeutic response can be determined based on the age, health, size and sex of the subject. Optimal amounts can also be determined based on monitoring of the subject's response to treatment.
Further, a therapeutically effective amount, can be an amount that selectively induces terminal differentiation, cell growth arrest and/or apoptosis of neoplastic cells, or an amount that induces terminal differentiation of tumor cells.
The method of the present invention is intended for the treatment or
chemoprevention of human patients with cancer. However, it is also likely that the method would be effective in the treatment of cancer in other subjects. "Subject", as used herein, refers to animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, pigs, dogs, cats, rabbits, guinea pigs, rats, mice or other bovine, ovine, equine, canine, feline, rodent or murine species.
The term "administration" and variants thereof (e.g., "administering" a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a cytotoxic agent, etc.), "administration" and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
COMBINATION THERAPY
The compounds of the present invention can be administered alone or in combination with other therapies suitable for the disease or disorder being treated. Where separate dosage formulations are used, the compound and the other therapeutic agent can be administered at essentially the same time (concurrently) or at separately staggered times
(sequentially). The pharmaceutical combination is understood to include all these regimens. Administration in these various ways are suitable for the present invention as long as the beneficial therapeutic effect of the compound and the other therapeutic agent are realized by the patient at substantially the same time. In an embodiment, such beneficial effect is achieved when the target blood level concentrations of each active drug are maintained at substantially the same time.
The instant compounds are also useful in combination with known therapeutic agents and anti-cancer agents. For example, instant compounds are useful in combination with known anti-cancer agents. Combinations of the presently disclosed compounds with other anti-cancer or chemotherapeutic agents are within the scope of the invention. Therefore, the present invention encompasses pharmaceutical compositions comprising a therapeutically effective amount of the compound of the invention and a pharmaceutically acceptable carrier and optionally other threrapeutic ingredients, such as an anti-cancer agent. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Such anti-cancer agents include, but are not limited to, the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl -protein transferase inhibitors, HMG-CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents, agents that interfere with cell cycle checkpoints, agents that interfere with receptor tyrosine kinases (RTKs) and cancer vaccines. The instant compounds are particularly useful when co-administered with radiation therapy. In an embodiment, the instant compounds are also useful in combination with known anti-cancer agents including the following: estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
"Estrogen receptor modulators" refers to compounds that interfere with or inhibit the binding of estrogen to the receptor, regardless of mechanism. Examples of estrogen receptor modulators include, but are not limited to, diethylstibestral, tamoxifen, raloxifene, idoxifene, LY353381, LY1 17081, toremifene, fluoxymestero, lfulvestrant, 4-[7- (2,2-dimethyl- 1 -oxopropoxy-4-methyl-2- [4- [2-( 1 -piperidinyl)ethoxy]phenyl] -2H- 1 - benzopyran-3-yl]-phenyl-2,2-dimethylpropanoate, 4,4'-dihydroxybenzophenone-2,4- dinitrophenyl-hydrazone, and SH646.
Other hormonal agents include: aromatase inhibitors (e.g., aminoglutethimide, anastrozole and tetrazole), luteinizing hormone release hormone (LHRH) analogues, ketoconazole, goserelin acetate, leuprolide, megestrol acetate and mifepristone.
"Androgen receptor modulators" refers to compounds which interfere or inhibit the binding of androgens to the receptor, regardless of mechanism. Examples of androgen receptor modulators include finasteride and other 5a-reductase inhibitors, nilutamide, flutamide, bicalutamide, liarozole, and abiraterone acetate.
"Retinoid receptor modulators" refers to compounds which interfere or inhibit the binding of retinoids to the receptor, regardless of mechanism. Examples of such retinoid receptor modulators include bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, a- difluoromethylornithine, ILX23-7553, trans-N-(4'-hydroxyphenyl) retinamide, and N-4- carboxyphenyl retinamide.
"Cytotoxic/cytostatic agents" refer to compounds which cause cell death or inhibit cell proliferation primarily by interfering directly with the cell's functioning or inhibit or interfere with cell mytosis, including alkylating agents, tumor necrosis factors,
intercalators, hypoxia activatable compounds, microtubule inhibitors/microtubule-stabilizing agents, inhibitors of mitotic kinesins, inhibitors of histone deacetylase, inhibitors of kinases involved in mitotic progression, antimetabolites; biological response modifiers;
hormonal/anti-hormonal therapeutic agents, haematopoietic growth factors, monoclonal antibody targeted therapeutic agents, topoisomerase inhibitors, proteasome inhibitors and ubiquitin ligase inhibitors. Examples of cytotoxic agents include, but are not limited to, sertenef, cachectin, chlorambucil, cyclophosphamide, ifosfamide, mechlorethamine, melphalan, uracil mustard, thiotepa, busulfan, carmustine, lomustine, streptozocin, tasonermin, lonidamine, carboplatin, altretamine, dacarbazine, procarbazine, prednimustine, dibromodulcitol, ranimustine, fotemustine, nedaplatin, oxaliplatin, temozolomide, heptaplatin, estramustine, improsulfan tosilate, trofosfamide, nimustine, dibrospidium chloride, pumitepa, lobaplatin, satraplatin, profiromycin, cisplatin, irofulven, dexifosfamide, cis-aminedichloro(2-methyl- pyridine)platinum, benzylguanine, glufosfamide, GPX100, (trans, trans, trans)-bis-mu- (hexane-l,6-diamine)-mu-[diamine-platinum(II)]bis[diamine(chloro)platinum
(II)]tetrachloride, diarizidinylspermine, arsenic tri oxide, 1-(1 l-dodecylamino-10- hydroxyundecyl)-3,7-dimethylxanthine, zorubicin, doxorubicin, daunorubicin, idarubicin, anthracenedione, bleomycin, mitomycin C, dactinomycin, plicatomycin, bisantrene, mitoxantrone, pirarubicin, pinaflde, valrubicin, amrubicin, antineoplaston, 3'-deamino-3'- morpholino-13-deoxo-10-hydroxycarminomycin, annamycin, galarubicin, elinafide, MEN 10755, and 4-demethoxy-3-deamino-3-aziridinyl-4-methylsulphonyl-daunorubicin (see
WO 00/50032).
An example of a hypoxia activatable compound is tirapazamine. Examples of proteasome inhibitors include but are not limited to lactacystin and bortezomib.
Examples of microtubule inhibitors/microtubule-stabilising agents include vincristine, vinblastine, vindesine, vinzolidine, vinorelbine, vindesine sulfate, 3',4'- didehydro-4'-deoxy-8'-norvincaleukoblastine, podophyllotoxins (e.g., etoposide (VP-16) and teniposide (VM-26)), paclitaxel, docetaxol, rhizoxin, dolastatin, mivobulin isethionate, auristatin, cemadotin, RPR109881, BMS184476, vinflunine, cryptophycin, 2,3,4,5,6- pentafluoro-N-(3-fluoro-4-methoxyphenyl) benzene sulfonamide, anhydrovinblastine, N,N- dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-prolyl-L-proline-t-butylamide, TDX258, the epothilones (see for example U.S. Pat. Nos. 6,284,781 and 6,288,237) and BMS 188797.
Some examples of topoisomerase inhibitors are topotecan, hycaptamine, irinotecan, rubitecan, 6-ethoxypropionyl-3',4'-0-exo-benzylidene-chartreusin, 9-methoxy- N,N-dimethyl-5-nitropyrazolo[3,4,5-kl]acridine-2-(6H) propanamine, l-amino-9-ethyl-5- fluoro-2,3-dihydro-9-hydroxy-4-methyl-lH,12H-benzo[de]pyrano[3',4' :b,7]- indolizino[l ,2b]quinoline-l 0,13(9H, 15H)dione, lurtotecan, 7-[2-(N-isopropylamino)ethyl]- (20S)camptothecin, BNP1350, BNPI1 100, BN80915, BN80942, etoposide phosphate, teniposide, sobuzoxane, 2'-dimethylamino-2'-deoxy-etoposide, GL331, N-[2- (dimethylamino)ethyl]-9-hydroxy-5,6-dimethyl-6H-pyrido[4,3-b]carbazole-l-^ asulacrine, (5a, 5aB, 8aa,9b)-9-[2-[N-[2-(dimethylamino)ethyl]-N-methylarnino]ethyl]-5-[4- hydro0xy-3,5-dimethoxyphenyl]-5,5a,6,8,8a,9-hexohydrofuro(3',4':6,7)naphtho(2,3-d)-l ,3- dioxol-6-one, 2,3-(methylenedioxy)-5-methyl-7-hydroxy-8-methoxybenzo[c]- phenanthridinium, 6,9-bis[(2-aminoethyl)amino]benzo[g]isoguinoline-5,10-dione, 5-(3- aminopropylamino)-7,10-dihydroxy-2-(2-hydroxyethylaminomethyl)-6H-pyrazolo[4,5,l- de]acridin-6-one, N-[l-[2(diethylamino)ethylamino]-7-methoxy-9-oxo-9H-thioxanthen-4- ylmethyl]formamide, N-(2-(dimethylamino)ethyl)acridine-4-carboxamide, 6-[[2- (dimethylamino)ethyl]amino]-3-hydroxy-7H-indeno[2,l-c] quinolin-7-one, and dimesna.
Examples of inhibitors of mitotic kinesins, and in particular the human mitotic kinesin KSP, are described in PCT Publications WO 01/30768, WO 01/98278, WO
03/050,064, WO 03/050,122, WO 03/049,527, WO 03/049,679, WO 03/049,678, WO 03/39460 and WO2003/079973, WO2003/099211, WO2004/039774, WO2003/105855, WO2003/106417. In an embodiment inhibitors of mitotic kinesins include, but are not limited to inhibitors of KSP, inhibitors of MKLP 1 , inhibitors of CENP-E, inhibitors of MC AK, inhibitors of Kifl4, inhibitors of Mphosphl and inhibitors of Rab6-KIFL.
Examples of "histone deacetylase inhibitors" include, but are not limited to, SAHA, TSA, oxamflatin, PXD101 , MG98, valproic acid and scriptaid. Further reference to other histone deacetylase inhibitors may be found in the following manuscript; Miller, T.A. et al. J. Med. Chem. 46(24):5097-5116 (2003).
"Inhibitors of kinases involved in mitotic progression" include, but are not limited to, inhibitors of aurora kinase, inhibitors of Polo-like kinases (PLK; in particular inhibitors of PLK-1), inhibitors of bub-1 and inhibitors of bub-Rl . An example of an "aurora kinase inhibitor" is VX-680.
"Antiproliferative agents" includes antisense RNA and DNA oligonucleotides such as G3139, ODN698, RVASKRAS, GEM231, and ΓΝΧ3001, and antimetabolites such as enocitabine, carmofur, tegafur, pentostatin, doxifluridine, trimetrexate, fludarabine, capecitabine, galocitabine, cytarabine ocfosfate, fosteabine sodium hydrate, raltitrexed, paltitrexid, emitefur, tiazofurin, decitabine, nolatrexed, pemetrexed, nelzarabine, 2'-deoxy-2'- methylidenecytidine, 2'-fluoromethylene-2'-deoxycytidine, N-[5-(2,3-dihydro- benzofuryl)sulfonyl]-N'-(3,4-dichlorophenyl)urea, N6-[4-deoxy-4-[N2-[2(E),4(E)- tetradecadienoyl]glycylamino]-L-glycero-B-L-manno-heptopyranosyl]adenine, aplidine, ecteinascidin, troxacitabine, 4-[2-amino-4-oxo-4,6,7,8-tetrahydro-3H-pyrimidino[5,4- b][l ,4]thiazin-6-yl-(S)-ethyl]-2,5-thienoyl-L-glutamic acid, aminopterin, 5-flurouracil, floxuridine, methotrexate, leucovarin, hydroxyurea, thioguanine (6-TG), mercaptopurine (6- MP), cytarabine, pentostatin, fludarabine phosphate, cladribine (2-CDA), asparaginase, gemcitabine, alanosine, 1 l-acetyl-8-(carbamoyloxymethyl)-4-formyl-6-methoxy-14-oxa-l ,l 1- diazatetracyclo(7.4.1.0.0)-tetradeca-2,4,6-trien-9-yl acetic acid ester, swainsonine, lometrexol, dexrazoxane, methioninase, 2'-cyano-2'-deoxy-N4-palmitoyl-l-B-D-arabino furanosyl cytosine and 3-aminopyridine-2-carboxaldehyde thiosemicarbazone.
Examples of monoclonal antibody targeted therapeutic agents include those therapeutic agents which have cytotoxic agents or radioisotopes attached to a cancer cell specific or target cell specific monoclonal antibody. Examples include Bexxar.
"HMG-CoA reductase inhibitors" refers to inhibitors of 3-hydroxy-3- methylglutaryl-CoA reductase. Examples of HMG-CoA reductase inhibitors that may be used include but are not limited to lovastatin (MEVACOR®; see U.S. Pat. Nos. 4,231 ,938,
4,294,926 and 4,319,039), simvastatin (ZOCOR®; see U.S. Pat. Nos. 4,444,784, 4,820,850 and 4,916,239), pravastatin (PRAVACHOL®; see U.S. Pat. Nos. 4,346,227, 4,537,859, 4,410,629, 5,030,447 and 5,180,589), fluvastatin (LESCOL®; see U.S. Pat. Nos. 5,354,772,
4,911,165, 4,929,437, 5,189,164, 5,118,853, 5,290,946 and 5,356,896) and atorvastatin
(LIPITOR®; see U.S. Pat. Nos. 5,273,995, 4,681,893, 5,489,691 and 5,342,952). The structural formulas of these and additional HMG-CoA reductase inhibitors that may be used in the instant methods are described at page 87 of M. Yalpani, "Cholesterol Lowering Drugs", Chemistry & Industry, pp. 85-89 (5 February 1996) and US Patent Nos. 4,782,084 and
4,885,314. The term HMG-CoA reductase inhibitor as used herein includes all
pharmaceutically acceptable lactone and open-acid forms (i.e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds which have HMG- CoA reductase inhibitory activity, and therefor the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention.
"Prenyl -protein transferase inhibitor" refers to a compound which inhibits any one or any combination of the prenyl-protein transferase enzymes, including farnesyl -protein transferase (FPTase), geranylgeranyl-protein transferase type I (GGPTase-I), and
geranylgeranyl -protein transferase type-II (GGPTase-II, also called Rab GGPTase).
Examples of prenyl-protein transferase inhibitors can be found in the following publications and patents: WO 96/30343, WO 97/18813, WO 97/21701, WO 97/23478, WO 97/38665, WO 98/28980, WO 98/29119, WO 95/32987, U.S. Pat. No. 5,420,245, U.S. Pat. No. 5,523,430, U.S. Pat. No. 5,532,359, U.S. Pat. No. 5,510,510, U.S. Pat. No. 5,589,485, U.S. Pat. No. 5,602,098, European Patent Publ. 0 618 221, European Patent Publ. 0 675 112, European Patent Publ. 0 604 181, European Patent Publ. 0 696 593, WO 94/19357, WO 95/08542, WO 95/1 1917, WO 95/12612, WO 95/12572, WO 95/10514, U.S. Pat. No.
5,661 ,152, WO 95/10515, WO 95/10516, WO 95/24612, WO 95/34535, WO 95/25086, WO 96/05529, WO 96/06138, WO 96/06193, WO 96/16443, WO 96/21701 , WO 96/21456, WO
96/22278, WO 96/24611 , WO 96/24612, WO 96/05168, WO 96/05169, WO 96/00736, U.S. Pat. No. 5,571,792, WO 96/17861 , WO 96/33159, WO 96/34850, WO 96/34851 , WO
96/30017, WO 96/30018, WO 96/30362, WO 96/30363, WO 96/31111, WO 96/31477, WO 96/31478, WO 96/31501 , WO 97/00252, WO 97/03047, WO 97/03050, WO 97/04785, WO 97/02920, WO 97/17070, WO 97/23478, WO 97/26246, WO 97/30053, WO 97/44350,
WO 98/02436, and U.S. Pat. No. 5,532,359. For an example of the role of a prenyl-protein transferase inhibitor on angiogenesis see European J. of Cancer, Vol. 35, No. 9, pp.1394-1401 (1999).
"Angiogenesis inhibitors" refers to compounds that inhibit the formation of new blood vessels, regardless of mechanism. Examples of angiogenesis inhibitors include, but are not limited to, tyrosine kinase inhibitors, such as inhibitors of the tyrosine kinase receptors Flt-1 (VEGFR1) and Flk-l/KDR (VEGFR2), inhibitors of epidermal-derived, fibroblast-derived, or platelet derived growth factors, MMP (matrix metal loprotease) inhibitors, integrin blockers, interferon-a, interleukin-12, erythropoietin (epoietin-a), granulocyte-CSF (filgrastin), granulocyte, macrophage-CSF (sargramostim), pentosan polysulfate, cyclooxygenase inhibitors, including nonsteroidal anti-inflammatories (NSAIDs) like aspirin and ibuprofen as well as selective cyclooxy-genase-2 inhibitors like celecoxib and rofecoxib (PNAS, Vol. 89, p. 7384 (1992); JNCI, Vol. 69, p. 475 (1982); Arch. Opthalmol, Vol. 108, p.573 (1990); Anat. Rec, Vol. 238, p. 68 (1994); FEBS Letters, Vol. 372, p. 83 (1995); Clin, Orthop. Vol. 313, p. 76 (1995); J Mol. Endocrinol, Vol. 16, p.107 (1996); Jpn.
J. Pharmacol, Vol. 75, p. 105 (1997); Cancer Res., Vol. 57, p. 1625 (1997); Cell, Vol. 93, p. 705 (1998); Intl. J. Mol. Med., Vol. 2, p. 715 (1998); J Biol. Chem., Vol. 274, p. 9116 (1999)), steroidal anti-inflammatories (such as corticosteroids, mineralocorticoids, dexamethasone, prednisone, prednisolone, methylpred, betamethasone),
carboxyamidotriazole, combretastatin A-4, squalamine, 6-O-chloroacetyl-carbonyl)- fumagillol, thalidomide, angiostatin, troponin- 1, angiotensin II antagonists (see Fernandez et al., J. Lab. Clin. Med. 105: 141-145 (1985)), and antibodies to VEGF (see, Nature Biotechnology, Vol. 17, pp.963-968 (October 1999); Kim et al., Nature, 362, 841-844 (1993); WO 00/44777; and WO 00/61186).
Other therapeutic agents that modulate or inhibit angiogenesis and may also be used in combination with the compounds of the instant invention include agents that modulate or inhibit the coagulation and fibrinolysis systems (see review in Clin. Chem. La. Med.
38:679-692 (2000)). Examples of such agents that modulate or inhibit the coagulation and fibrinolysis pathways include, but are not limited to, heparin (see Thromb. Haemost. 80: 10-23 (1998)), low molecular weight heparins and carboxypeptidase U inhibitors (also known as inhibitors of active thrombin activatable fibrinolysis inhibitor [TAFIa]) (see Thrombosis Res. 101 :329-354 (2001)). TAFIa inhibitors have been described in PCT Publication WO
03/013,526 and U.S. Ser. No. 60/349,925 (filed January 18, 2002).
"Agents that interfere with cell cycle checkpoints" refer to compounds that inhibit protein kinases that transduce cell cycle checkpoint signals, thereby sensitizing the cancer cell to DNA damaging agents. Such agents include inhibitors of ATR, ATM, the Chkl and Chk2 kinases and cdk and cdc kinase inhibitors and are specifically exemplified by 7- hydroxystaurosporin, flavopiridol, CYC202 (Cyclacel) and BMS-387032.
"Agents that interfere with receptor tyrosine kinases (RTKs)" refer to compounds that inhibit RTKs and therefore mechanisms involved in oncogenesis and tumor progression. Such agents include inhibitors of c-Kit, Eph, PDGF, Flt3 and c-Met. Further agents include inhibitors of RTKs shown as described by Bume- Jensen and Hunter, Nature,
411 :355-365, 2001.
"Inhibitors of cell proliferation and survival signaling pathway" refer to pharmaceutical agents that inhibit cell surface receptors and signal transduction cascades downstream of those surface receptors. Such agents include inhibitors of inhibitors of EGFR (for example gefitinib and erlotinib), inhibitors of ERB-2 (for example trastuzumab), inhibitors of IGFR, inhibitors of CD20 (rituximab), inhibitors of cytokine receptors, inhibitors of MET, inhibitors of PI3K family kinase (for example LY294002), serine/threonine kinases (including but not limited to inhibitors of Akt such as described in (WO 03/086404, WO 03/086403, WO 03/086394, WO 03/086279, WO 02/083675, WO 02/083139, WO 02/083140 and WO 02/083138), inhibitors of Raf kinase (for example BAY-43-9006 ), inhibitors of
MEK (for example CI- 1040 and PD-098059) and inhibitors of mTOR (for example Wyeth CCI-779 and Ariad AP23573). Such agents include small molecule inhibitor compounds and antibody antagonists. Examples of mTOR inhibitors include ridaforolimus, temsirolimus, everolimus, a rapamycin-analog. Ridaforolimus, also known as AP 23573, MK-8669 and deforolimus, is a unique, non-prodrug analog of rapmycin that has antiproliferative activity in a broad range of human tumor cell lines in vitro and in murine tumor xenograft models utilizing human tumor cell lines. Ridaforolimus has been administered to patients with advanced cancer and is currently in clinical development for various advanced malignancies, including studies in patients with advanced soft tissue or bone sarcomas. Thus far, these trials have demonstrated that ridaforolimus is generally well-tolerated with a predictable and manageable adverse even profile, and possess anti-tumor activity in a broad range of cancers. A description and preparation of ridaforolimus is described in U.S. Patent No. 7,091,213 to Ariad Gene
Therapeutics, Inc.
Temsirolimus, also known as Torisel®, is currently marketed for the treatment of renal cell carcinoma. A description and preparation of temsirolimus is described in U.S. Patent No. 5,362,718 to American Home Products Corporation. Everolimus, also known as Certican® or RADOO 1 , marketed by Novartis, has greater stability and enhanced solubility in organic solvents, as well as more favorable pharmokinetics with fewer side effects than rapamycin (sirolimus). Everolimus has been used in conjunction with microemulsion cyclosporin
(Neoral®, Novartis) to increase the efficacy of the immunosuppressive regime.
"Apoptosis inducing agents" include activators of TNF receptor family members (including the TRAIL receptors).
The invention also encompasses combinations with NSAID's which are selective COX-2 inhibitors. For purposes of this specification NSAID's which are selective inhibitors of COX-2 are defined as those which possess a specificity for inhibiting COX-2 over COX-1 of at least 100 fold as measured by the ratio of IC50 f°r COX-2 over IC50 for COX-1 evaluated by cell or microsomal assays. Such compounds include, but are not limited to those disclosed in U.S. Pat. 5,474,995, U.S. Pat. 5,861,419, U.S. Pat. 6,001,843, U.S. Pat. 6,020,343, U.S. Pat. 5,409,944, U.S. Pat. 5,436,265, U.S. Pat. 5,536,752, U.S. Pat. 5,550,142, U.S. Pat. 5,604,260, U.S. 5,698,584, U.S. Pat. 5,710,140, WO 94/15932, U.S. Pat. 5,344,991, U.S. Pat. 5,134,142, U.S. Pat. 5,380,738, U.S. Pat. 5,393,790, U.S. Pat. 5,466,823, U.S. Pat. 5,633,272, and U.S. Pat. 5,932,598.
Inhibitors of COX-2 that are particularly useful in the instant method of treatment are: 3-phenyl-4-(4-(methylsulfonyl)phenyl)-2-(5H)-furanone; and 5-chloro-3-(4- methylsulfonyl)phenyl-2-(2-methyl-5-pyridinyl)pyridine; or a pharmaceutically acceptable salt thereof.
Compounds that have been described as specific inhibitors of COX-2 and are therefore useful in the present invention include, but are not limited to: parecoxib,
CELEBREX® and BEXTRA® or a pharmaceutically acceptable salt thereof.
Other examples of angiogenesis inhibitors include, but are not limited to, endostatin, ukrain, ranpirnase, IM862, 5-methoxy-4-[2-methyl-3-(3-methyl-2- butenyl)oxiranyl]- 1 -oxaspiro[2,5]oct-6-yl(chloroacetyl)carbamate, acetyldinanaline, 5-amino- l-[[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]methyl]-lH-l,2,3-triazole-4- carboxamide,CM 101, squalamine, combretastatin, RPI4610, NX31838, sulfated
mannopentaose phosphate, 7,7-(carbonyl-bis[imino-N-methyl-4,2-pyrrolocarbonylimino[N- methyl-4,2-pyrrole]-carbonylimino]-bis-( 1,3 -naphthalene disulfonate), and 3-[(2,4- dimethylpyrrol-5-yl)methylene]-2-indolinone (SU5416).
As used above, "integrin blockers" refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the αγβ3 integrin, to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ανβ5 integrin, to compounds which antagonize, inhibit or counteract binding of a physiological ligand to both the ανβ3 integrin and the νβ5 integrin, and to compounds which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the ανβ6> «νβ8> xi β χ , 2βΐ, «5β 1, 6βΐ and 6β4 integrins. The term also refers to antagonists of any
combination of ανβ3, νβ5, ανβ6> ανβ8> αΐ β ΐ' α2βΐ> α5βΐ > 6βΐ and α6β4 integrins.
Some specific examples of tyrosine kinase inhibitors include N- (trifluoromethylphenyl)-5-methylisoxazol-4-carboxamide, 3-[(2,4-dimethylpyrrol-5- yl)methylidenyl)indolin-2-one, 17-(allylamino)-17-demethoxygeldanamycin, 4-(3-chloro-4- fluorophenylamino)-7-methoxy-6- [3 -(4-morpholinyl)propoxyl] quinazoline, N-(3 - ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine, BIBX1382, 2,3,9,10,1 1,12- hexahydro- 10-(hydroxymethyl)- 10-hydroxy-9-methyl-9, 12-epoxy- 1 H-diindolo[ 1 ,2,3- fg:3',2',l '-kl]pyrrolo[3,4-i][l ,6]benzodiazocin-l-one, SH268, genistein, imatinib (STI571), CEP2563, 4-(3-chlorophenylamino)-5,6-dimethyl-7H-pyrrolo[2,3-d]pyrimidinemethane sulfonate, 4-(3-bromo-4-hydroxyphenyl)amino-6,7-dimethoxyquinazoline, 4-(4'- hydroxyphenyl)amino-6,7-dimethoxyquinazoline, SU6668, STI571A, N-4-chlorophenyl-4-(4- pyridylmethyl)-l-phthalazinamine, and EMD121974. Combinations with compounds other than anti-cancer compounds are also encompassed in the instant methods. For example, combinations of the instantly claimed compounds with PPAR-γ (i.e., PPAR-gamma) agonists and PPAR-δ (i.e., PPAR-delta) agonists are useful in the treatment of certain malingnancies. PPAR-γ and PPAR-δ are the nuclear peroxisome proliferator-activated receptors γ and δ. The expression of PPAR-γ on endothelial cells and its involvement in angiogenesis has been reported in the literature (see J. Cardiovasc. Pharmacol. 1998; 31 :909-913; J Biol. Chem. 1999; 274:9116-9121 ; Invest. Ophthalmol Vis. Sci. 2000; 41 :2309-2317). More recently, PPAR-γ agonists have been shown to inhibit the angiogenic response to VEGF in vitro; both troglitazone and rosiglitazone maleate inhibit the development of retinal neovascularization in mice. (Arch. Ophthamol.
2001 ; 119:709-717). Examples of PPAR-γ agonists and PPAR- γ/ agonists include, but are not limited to, thiazolidinediones (such as DRF2725, CS-01 1 , troglitazone, rosiglitazone, and pioglitazone), fenofibrate, gemfibrozil, clofibrate, GW2570, SB219994, AR-H039242, JTT- 501, MCC-555, GW2331, GW409544, NN2344, KRP297, NP0110, DRF4158, NN622, GI262570, PNU182716, DRF552926, 2-[(5,7-dipropyl-3-trifluoromethyl-l,2-benzisoxazol-6- yl)oxy]-2-methylpropionic acid (disclosed in USSN 09/782,856), and 2(R)-7-(3-(2-chloro-4- (4-fluorophenoxy) phenoxy)propoxy)-2-ethylchromane-2-carboxylic acid (disclosed in USSN 60/235,708 and 60/244,697).
Another embodiment of the instant invention is the use of the presently disclosed compounds in combination with gene therapy for the treatment of cancer. For an overview of genetic strategies to treating cancer see Hall et al (Am J Hum Genet 61 :785-789, 1997) and ufe et al (Cancer Medicine, 5th Ed, pp 876-889, BC Decker, Hamilton 2000). Gene therapy can be used to deliver any tumor suppressing gene. Examples of such genes include, but are not limited to, p53, which can be delivered via recombinant virus-mediated gene transfer (see U.S. Pat. No. 6,069,134, for example), Duc-4, NF-1, NF-2, RB, WT1,
BRCA1, BRCA2, a uPA/uPAR antagonist ("Adenovirus-Mediated Delivery of a uPA/uPAR Antagonist Suppresses Angiogenesis-Dependent Tumor Growth and Dissemination in Mice," Gene Therapy, August 1998; 5(8): 1 105-13), and interferon gamma (J Immunol. 2000;
164:217-222).
The compounds of the instant invention may also be administered in combination with an inhibitor of inherent multidrug resistance (MDR), in particular MDR associated with high levels of expression of transporter proteins. Such MDR inhibitors include inhibitors of p-glycoprotein (P-gp), such as LY335979, XR9576, OC144-093, R101922, VX853 and PSC833 (valspodar).
A compound of the present invention may be employed in conjunction with anti-emetic agents to treat nausea or emesis, including acute, delayed, late-phase, and anticipatory emesis, which may result from the use of a compound of the present invention, alone or with radiation therapy. For the prevention or treatment of emesis, a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin- 1 receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid,
Benecorten or others such as disclosed in U.S.Patent Nos. 2,789,118, 2,990,401 , 3,048,581, 3,126,375, 3,929,768, 3,996,359, 3,928,326 and 3,749,712, an antidopaminergic, such as the phenothiazines (for example prochlorperazine, fluphenazine, thioridazine and mesoridazine), metoclopramide or dronabinol. In an embodiment, an anti-emesis agent selected from a neurokinin- 1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is administered as an adjuvant for the treatment or prevention of emesis that may result upon administration of the instant compounds.
Neurokinin- 1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U.S. Pat. Nos. 5,162,339, 5,232,929, 5,242,930, 5,373,003, 5,387,595, 5,459,270, 5,494,926, 5,496,833, 5,637,699, 5,719,147;
European Patent Publication Nos. EP 0 360 390, 0 394 989, 0 428 434, 0 429 366, 0 430 771, 0 436 334, 0 443 132, 0 482 539, 0 498 069, 0 499 313, 0 512 901, 0 512 902, 0 514 273, 0 514 274, 0 514 275, 0 514 276, 0 515 681, 0 517 589, 0 520 555, 0 522 808, 0 528 495, 0 532 456, 0 533 280, 0 536 817, 0 545 478, 0 558 156, 0 577 394, 0 585 913,0 590 152, 0 599 538, 0 610 793, 0 634 402, 0 686 629, 0 693 489, 0 694 535, 0 699 655, 0 699 674, 0 707 006, 0 708 101, 0 709 375, 0 709 376, 0 714 891 , 0 723 959, 0 733 632 and 0 776 893; PCT International Patent Publication Nos. WO 90/05525, 90/05729, 91/09844, 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661 , 92/20676, 92/21677,
92/22569, 93/00330, 93/00331, 93/01159, 93/01 165, 93/01 169, 93/01 170, 93/06099,
93/091 16, 93/10073, 93/14084, 93/141 13, 93/18023, 93/19064, 93/21 155, 93/21 181,
93/23380, 93/24465, 94/00440, 94/01402, 94/02461, 94/02595, 94/03429, 94/03445,
94/04494, 94/04496, 94/05625, 94/07843, 94/08997, 94/10165, 94/10167, 94/10168,
94/10170, 94/11368, 94/13639, 94/13663, 94/14767, 94/15903, 94/19320, 94/19323,
94/20500, 94/26735, 94/26740, 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/11880, 95/14017, 95/1531 1 , 95/16679, 95/17382,
95/18124, 95/18129, 95/19344, 95/20575, 95/21819, 95/22525, 95/23798, 95/26338,
95/28418, 95/30674, 95/30687, 95/33744, 96/05181, 96/05193, 96/05203, 96/06094,
96/07649, 96/10562, 96/16939, 96/18643, 96/20197, 96/21661, 96/29304, 96/29317,
96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066,
97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/21702; and in British Patent Publication Nos. 2 266 529, 2 268 931, 2 269 170, 2 269 590, 2 271 774, 2 292 144, 2 293 168, 2 293 169, and 2 302 689. The preparation of such compounds is fully described in the aforementioned patents and publications.
In an embodiment, the neurokinin- 1 receptor antagonist for use in conjunction with the compounds of the present invention is selected from: 2-(R)-(l-(R)-(3,5- bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-lH,4H-l,2,4- triazolo)methyl)morpholine, or a pharmaceutically acceptable salt thereof, which is described in U.S. Pat. No. 5,719,147.
A compound of the instant invention may also be administered with an agent useful in the treatment of anemia. Such an anemia treatment agent is, for example, a continuous eythropoiesis receptor activator (such as epoetin alfa).
A compound of the instant invention may also be administered with an agent useful in the treatment of neutropenia. Such a neutropenia treatment agent is, for example, a hematopoietic growth factor which regulates the production and function of neutrophils such as a human granulocyte colony stimulating factor, (G-CSF). Examples of a G-CSF include filgrastim.
A compound of the instant invention may also be administered with an immunologic-enhancing drug, such as levamisole, bacillus Calmette-Guerin, octreotide, isoprinosine and Zadaxin.
A compound of the instant invention may also be useful for treating or preventing cancer, including bone cancer, in combination with bisphosphonates (understood to include bisphosphonates, diphosphonates, bisphosphonic acids and diphosphonic acids). Examples of bisphosphonates include but are not limited to: etidronate (Didronel), pamidronate (Aredia), alendronate (Fosamax), risedronate (Actonel), zoledronate (Zometa), ibandronate (Boniva), incadronate or cimadronate, clodronate, EB-1053, minodronate, neridronate, piridronate and tiludronate including any and all pharmaceutically acceptable salts, derivatives, hydrates and mixtures thereof. A compound of the instant invention may also be useful for treating or preventing breast cancer in combination with aromatase inhibitors. Examples of aromatase inhibitors include but are not limited to anastrozole, letrozole and exemestane.
A compound of the instant invention may also be useful for treating or preventing cancer in combination with siRNA therapeutics.
A compound of the instant invention may also be useful for treating or preventing cancer in combination withcompounds which induce terminal differentiation of the neoplastic cells. Suitable differentiation agents include the compounds disclosed in any one or more of the following references.
a) Polar compounds (Marks et al (1987); Friend, C, Scher, W., Holland, J. W., and Sato, T. (1971) Proc. Natl. Acad. Sci. (USA) 68: 378-382; Tanaka, M., Levy, J., Terada, M, Breslow, R., Rifkind, R. A., and Marks, P. A. (1975) Proc. Natl. Acad. Sci. (USA) 72: 1003-1006; Reuben, R. C, Wife, R. L., Breslow, R., Rifkind, R. A., and Marks, P. A. (1976) Proc. Natl. Acad. Sci. (USA) 73: 862-866);
b) Derivatives of vitamin D and retinoic acid (Abe, E., Miyaura, C, Sakagami,
H., Takeda, M., Konno, K., Yamazaki, T., Yoshika, S., and Suda, T. (1981) Proc. Natl. Acad. Sci. (USA) 78: 4990-4994; Schwartz, E. L., Snoddy, J. R., Kreutter, D., Rasmussen, H., and Sartorelli, A. C. (1983) Proc. Am. Assoc. Cancer Res. 24: 18; Tanenaga, K., Hozumi, M., and Sakagami, Y. (1980) Cancer Res. 40: 914-919);
c) Steroid hormones (Lotem, J. and Sachs, L. (1975) Int. J. Cancer 15: 731-
740);
d) Growth factors (Sachs, L. (1978) Nature (Lond.) 274: 535, Metcalf, D. (1985) Science, 229: 16-22);
e) Proteases (Scher, W., Scher, B. M., and Waxman, S. (1983) Exp. Hematol. 1 1 : 490-498; Scher, W., Scher, B. M., and Waxman, S. (1982) Biochem. & Biophys. Res.
Comm. 109: 348-354);
f) Tumor promoters (Huberman, E. and Callaham, M. F. (1979) Proc. Natl. Acad. Sci. (USA) 76: 1293-1297; Lottem, J. and Sachs, L. (1979) Proc. Natl. Acad. Sci.
(USA) 76: 5158-5162); and
g) inhibitors of DNA or RNA synthesis (Schwartz, E. L. and Sartorelli, A. C.
(1982) Cancer Res. 42: 2651-2655, Terada, M., Epner, E., Nudel, U., Salmon, J., Fibach, E., Rifkind, R. A., and Marks, P. A. (1978) Proc. Natl. Acad. Sci. (USA) 75: 2795-2799; Morin, M. J. and Sartorelli, A. C. (1984) Cancer Res 44: 2807-2812; Schwartz, E. L., Brown, B. J., Nierenberg, M., Marsh, J. C, and Sartorelli, A. C. (1983) Cancer Res. 43: 2725-2730; Sugano, H., Furusawa, M., Kawaguchi, T., and Ikawa, Y. (1973) Bibl. Hematol. 39: 943-954; Ebert, P. S., Wars, I., and Buell, D. N. (1976) Cancer Res. 36: 1809-1813; Hayashi, M., Okabe, J., and Hozumi, M. (1979) Gann 70: 235-238).
A compound of the instant invention may also be useful for treating or preventing cancer in combination with γ-secretase inhibitors.
Also included in the scope of the claims is a method of treating cancer that comprises administering a therapeutically effective amount of a compound of Formula I in combination with radiation therapy and/or in combination with a second compound selected from: an estrogen receptor modulator, an androgen receptor modulator, a retinoid receptor modulator, a cytotoxiccytostatic agent, an antiproliferative agent, a prenyl-protein transferase inhibitor, an HMG-CoA reductase inhibitor, an HIV protease inhibitor, a reverse transcriptase inhibitor, an angiogenesis inhibitor, PPAR-γ agonists, PPAR-δ agonists, an inhibitor of inherent multidrug resistance, an anti-emetic agent, an agent useful in the treatment of anemia, an agent useful in the treatment of neutropenia, an immunologic-enhancing drug, an inhibitor of cell proliferation and survival signaling, a bisphosphonate, an aromatase inhibitor, an siRNA therapeutic, γ-secretase inhibitors, agents that interfere with receptor tyrosine kinases (RTKs) and an agent that interferes with a cell cycle checkpoint.
The compounds of the instant invention are useful in combination with the following therapeutic agents: abarelix (Plenaxis depot®); aldesleukin (Prokine®); Aldesleukin (Proleukin®); Alemtuzumabb (Campath®); alitretinoin (Panretin®); allopurinol
(Zyloprim®); altretamine (Hexalen®); amifostine (Ethyol®); anastrozole (Arimidex®);
arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bendamustine hydrochloride (Treanda®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®);
bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib (Velcade®); busulfan intravenous (Busulfex®); busulfan oral (Myleran®); calusterone (Methosarb®); capecitabine
(Xeloda®); carboplatin (Paraplatin®); carmustine (BCNU®, BiCNU®); carmustine
(Gliadel®); carmustine with Polifeprosan 20 Implant (Gliadel Wafer®); celecoxib
(Celebrex®); cetuximab (Erbitux®); chlorambucil (Leukeran®); cisplatin (Platinol®);
cladribine (Leustatin®, 2-CdA®); clofarabine (Clolar®); cyclophosphamide (Cytoxan®, Neosar®); cyclophosphamide (Cytoxan Injection®); cyclophosphamide (Cytoxan Tablet®); cytarabine (Cytosar-U®); cytarabine liposomal (DepoCyt®); dacarbazine (DTIC-Dome®); dactinomycin, actinomycin D (Cosmegen®); dalteparin sodium injection (Fragmin®); Darbepoetin alfa (Aranesp®); dasatinib (Sprycel®); daunorubicin liposomal (DanuoXome®); daunorubicin, daunomycin (Daunorubicin®); daunorubicin, daunomycin (Cerubidine®); degarelix (Firmagon®); Denileukin diftitox (Ontak®); dexrazoxane (Zinecard®); dexrazoxane hydrochloride (Totect®); docetaxel (Taxotere®); doxorubicin (Adriamycin PFS®);
doxorubicin (Adriamycin®, Rubex®); doxorubicin (Adriamycin PFS Injection®);
doxorubicin liposomal (Doxil®); dromostanolone propionate (Dromostanolone ®);
dromostanolone propionate (Masterone Injection®); eculizumab injection (Soliris®); Elliott's B Solution (Elliott's B Solution®); eltrombopag (Promacta®); epirubicin (Ellence®); Epoetin alfa (epogen®); erlotinib (Tarceva®); estramustine (Emcyt®); etoposide phosphate
(Etopophos®); etoposide, VP- 16 (Vepesid®); everolimus tablets (Afinitor®); exemestane
(Aromasin®); ferumoxytol (Feraheme Injection®); Filgrastim (Neupogen®); floxuridine (intraarterial) (FUDR®); fludarabine (Fludara®); fluorouracil, 5-FU (Adrucil®); fulvestrant (Faslodex®); gefitinib (Iressa®); gemcitabine (Gemzar®); gemtuzumab ozogamicin
(Mylotarg®); goserelin acetate (Zoladex Implant®); goserelin acetate (Zoladex®); histrelin acetate (Histrelin implant®); hydroxyurea (Hydrea®); Ibritumomab Tiuxetan (Zevalin®); idarubicin (Idamycin®); ifosfamide (IFEX®); imatinib mesylate (Gleevec®); interferon alfa 2a (Roferon A®); Interferon alfa-2b (Intron A®); iobenguane I 123 injection (AdreView®); irinotecan (Camptosar®); ixabepilone (Ixempra®); lapatinib tablets (Tykerb®); lenalidomide (Revlimid®); letrozole (Femara®); leucovorin (Wellcovorin®, Leucovorin®); Leuprolide Acetate (Eligard®); levamisole (Ergamisol®); lomustine, CCNU (CeeBU®); meclorethamine, nitrogen mustard (Mustargen®); megestrol acetate (Megace®); melphalan, L-PAM
(Alkeran®); mercaptopurine, 6-MP (Purinethol®); mesna (Mesnex®); mesna (Mesnex tabs®); methotrexate (Methotrexate®); methoxsalen (Uvadex®); mitomycin C
(Mutaniycin®); mitotane (Lysodren®); mitoxantrone (Novantrone®); nandrolone
phenpropionate (Durabolin-50®); nelarabine (Arranon®); nilotinib (Tasigna®);
Nofetumomab (Verluma®); ofatumumab (Arzerra®); Oprelvekin (Neumega®); oxaliplatin (Eloxatin®); paclitaxel (Paxene®); paclitaxel (Taxol®); paclitaxel protein-bound particles (Abraxane®); palifermin (Kepivance®); pamidronate (Aredia®); panitumumab (Vectibix®); pazopanib tablets (Votrienttm®); pegademase (Adagen (Pegademase Bovine)®);
pegaspargase (Oncaspar®); Pegfilgrastim (Neulasta®); pemetrexed disodium (Alimta®); pentostatin (Nipent®); pipobroman (Vercyte®); plerixafor (Mozobil®); plicamycin, mithramycin (Mithracin®); porfimer sodium (Photo frin®); pralatrexate injection (Folotyn®); procarbazine (Matulane®); quinacrine (Atabrine®); Rasburicase (Elitek®); raloxifene hydrochloride (Evista®); Rituximab (Rituxan®); romidepsin (Istodax®); romiplostim
(Nplate®); sargramostim (Leukine®); Sargramostim (Prokine®); sorafenib (Nexavar®);
streptozocin (Zanosar®); sunitinib maleate (Sutent®); talc (Sclerosol®); tamoxifen
(Nolvadex®); temozolomide (Temodar®); temsirolimus (Torisel®); teniposide, VM-26 (Vumon®); testolactone (Teslac®); thioguanine, 6-TG (Thioguanine®); thiotepa (Thioplex®); topotecan (Hycamtin®); toremifene (Fareston®); Tositumomab (Bexxar®); Tositumomab/I- 131 tositumomab (Bexxar®); Trastuzumab (Herceptin®); tretinoin, ATRA (Vesanoid®); Uracil Mustard (Uracil Mustard Capsules®); valrubicin (Valstar®); vinblastine (Velban®); vincristine (Oncovin®); vinorelbine (Navelbine®); vorinostat (Zolinza®); and zoledronate (Zometa®).
Non-limiting examples of other suitable anti-cancer agents for combination with the instant compounds are selected from the group consisting of a Cytostatic agent, Cisplatin, Deforolimus (described in PCT publication No. 2003/064383), Doxorubicin, liposomal doxorubicin (e.g., Caelyx®, Myocet®, Doxil®), Taxotere, Taxol, Etoposide, Irinotecan, Camptostar, Topotecan, Paclitaxel, Docetaxel, Epothilones, Tamoxifen, 5- Fluorouracil, Methoxtrexate, Temozolomide, cyclophosphamide, SCH 66336, Rl 15777®, L778,123®, BMS 214662®, Iressa®, Tarceva®, Antibodies to EGFR, antibodies to IGFR (including, for example, those published in US 2005/0136063 published June 23, 2005), ESK inhibitors, KSP inhibitors (such as, for example, those published in WO 2006/098962 and WO 2006/098961 ; ispinesib, SB-743921 from Cytokinetics), Centrosome associated protein E ("CENP-E") inhibitors (e.g., GSK-923295), Gleevec®, Intron, Ara-C, Adriamycin, Cytoxan, Gemcitabine, Uracil mustard, Chlormethine, Ifosfamide, Melphalan, Chlorambucil,
Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine,
Lomustine, Streptozocin, Dacarbazine, Floxuridine, Cytarabine, 6 Mercaptopurine, 6
Thioguanine, Fludarabine phosphate, Oxaliplatin, Leucovirin, ELOXATINTM, Vinblastine, Vincristine, Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Mithramycin, Deoxycoformycin, Mitomycin C, L Asparaginase, Teniposide 17a- Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone,
Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone,
Methyltestosterone, Prednisolone, Triamcinolone, Chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, Goserelin, Cisplatin, Carboplatin, Hydroxyurea, Amsacrine, Procarbazine, Mitotane, Mitoxantrone, Levamisole, Navelbene, Anastrazole, Letrazole, Capecitabine, Reloxafine, Droloxafme, Hexamethylmelamine, Avastin, herceptin, Bexxar, bortezomib ("Velcade"), Zevalin, Trisenox, Xeloda, Vinorelbine, Porfimer, Erbitux, Liposomal, Thiotepa, Altretamine, Melphalan, Trastuzumab, Lerozole, Fulvestrant, Exemestane, Fulvestrant, Ifosfomide, Rituximab, C225®, Satriplatin, mylotarg, Avastin, Rituxan, Panitubimab, Sutent, Sorafinib, Sprycel (dastinib), Nilotinib, Tykerb (Lapatinib) and Campath.
In one embodiment, the invention provides a method of treating cancer, the method comprising administering an amount of an Imidazopyrazine Compound or a pharmaceutically acceptable salt thereof, and an amount of one additional anticancer agent selected from the group consisting of Adriamycin, Altretamine, Amidox, Aminoglutethimide, Amsacrine, Anastrazole, Antibodies to EGFR, 3-AP, Aphidicolon, Ara-C, Arsenic trioxide, L Asparaginase, Bevacizumab, Bleomycin, BMS 214662, Bortezomib, Busulfan, Campath, Camptostar, Capecitabine, Carboplatin, Carmustine, Centrosome associated protein E
("CENP-E") inhibitors, Cetuximab, Cladribine, Chlorambucil, Chlormethine,
Chlorotrianisene, Cisplatin, Clofarabine, cyclophosphamide, Cytarabine, a Cytostatic agent, Cytoxan, Dacarbazine, Dactinomycin, Daunorubicin, Dasatinib, Deforolimus,
Deoxycoformycin, Didox, Diethylstilbestrol, Docetaxel, Doxorubicin, Dromostanolone, Droloxafme, Epirubicin, Epothilones, ERK inhibitors, Erlotinib, Etoposide, 17a-
Ethinylestradiol, Estramustine, Exemestane, Floxuridine, Fludarabine, Fludarabine phosphate, 5-Fluorouracil, Fluoxymesterone, Flutamide, Fulvestrant, Gefitinib, Gemcitabine,
Gemtuzumab ozogamcicin, Goserelin, GSK-923295, Hexamethylmelamine,
Hydroxyprogesterone, Hydroxyurea, Ibritumomab Tiuxetan, Idarubicin, Ifosfamide, Imatinib mesylate, Intron, Irinotecan, ispinesib, KSP inhibitors, L778,123, Lapatinib, Leucovirin,
Leuprolide, Lerozole, Letrazole, Levamisole, Liposomal Doxorubicin, Liposomal, Lomustine, Lonafarnib, Medroxyprogesteroneacetate, Megestrolacetate, Melphalan, 6 Mercaptopurine, Methoxtrexate, Methylprednisolone, Methyltestosterone, Mithramycin, Mitomycin C, Mitotane, Mitoxantrone, Navelbene, Nilotinib, Oxaliplatin, Paclitaxel, Panitubimab,
Pentostatin, Pipobroman, Porfimer, Prednisolone, Prednisone propionate, Procarbazine, Reloxafine, Rituximab, Satriplatin, SB-743921, Smll, Sorafinib, Streptozocin, Sunitinib, Tamoxifen, Taxotere, Taxol, Temozolomide, Teniposide, Testolactone, Testosterone, Tezacitabine, 6 Thioguanine, Thiotepa, Tipifarnib, Topotecan, Toremifene, Tositumomab, Trastuzumab, Triamcinolone, Triapine, Triethylenemelamine, Triethylenethiophosphoramine, Trimidox, Uracil mustard, Vinblastine, Vincristine, Vindesine, and Vinorelbine.
In one embodiment, the invention provides a method of treating cancer, the method comprising administering an amount of an Imidazopyrazine Compound or a pharmaceutically acceptable salt thereof, and an amount of one or more of a MAP Kinase pathway inhibitor such as bRaf, MEK, or ERK inhibitors to a patient in need thereof.
In another embodiment, the invention provides a method of treating cancer, the method comprising administering an amount of an Imidazopyrazine Compound or a pharmaceutically acceptable salt thereof, and an amount of one or more of ERK inhibitors (for example, compounds described in WO2008/156739, WO2007/070398, WO 2008/156739 and US publication 2007/0232610) to a patient in need thereof.
In one embodiment, the invention provides a method of treating cancer, the method comprising administering an amount of an Imidazopyrazine Compound or a pharmaceutically acceptable salt thereof, and an amount of one or more of an anti-IGF-lR antibody. Specific anti-IGF-lR antibodies include, but are not limited to, dalotuzumab, figitumumab, cixutumumab, SHC 717454, Roche R1507, EM 164 or Amgen AMG479.
The instant invention also includes a pharmaceutical composition useful for treating or preventing cancer that comprises a therapeutically effective amount of a compound of Formula I and a second compound selected from: an estrogen receptor modulator, an androgen receptor modulator, a retinoid receptor modulator, a cytotoxic/cyto static agent, an antiproliferative agent, a prenyl-protein transferase inhibitor, an HMG-CoA reductase inhibitor, an HIV protease inhibitor, a reverse transcriptase inhibitor, an angiogenesis inhibitor, a PPAR-γ agonist, a PPAR-δ agonist, an inhibitor of cell proliferation and survival signaling, a bisphosphonate, an aromatase inhibitor, an siRNA therapeutic, γ-secretase inhibitors, agents that interfere with receptor tyrosine kinases (RTKs) and an agent that interferes with a cell cycle checkpoint.
The use of all of these approaches in combination with the instant compounds described herein are within the scope of the present invention.
Compositions and Administration
This invention is also directed to pharmaceutical compositions which comprise at least one Imidazopyrazine Compound, or a pharmaceutically acceptable salt of said compound and at least one pharmaceutically acceptable carrier. When administered to a patient, the Imidazopyrazine Compounds can be administered as a component of a composition that comprises a pharmaceutically acceptable carrier or vehicle. The present invention provides pharmaceutical compositions comprising an effective amount of at least one Imidazopyrazine Compound and a pharmaceutically acceptable carrier. In the pharmaceutical compositions and methods of the present invention, the active ingredients will typically be administered in admixture with suitable carrier materials suitably selected with respect to the intended form of administration, i.e., oral tablets, capsules (either solid-filled, semi-solid filled or liquid filled), powders for constitution, oral gels, elixirs, dispersible granules, syrups, suspensions, and the like, and consistent with conventional pharmaceutical practices. Examples of pharmaceutically acceptable carriers and methods of manufacture for various compositions may be found in A. Gennaro (ed.),
Remingto 's Pharmaceutical Sciences, 18th Edition, (1990), Mack Publishing Co., Easton, Pennsylvania. For example, for oral administration in the form of tablets or capsules, the active drug component may be combined with any oral non-toxic pharmaceutically acceptable inert carrier, such as lactose, starch, sucrose, cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, talc, mannitol, ethyl alcohol (liquid forms) and the like. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets and suppositories. Powders and tablets may be comprised of from about 0.5 to about 95 percent inventive composition. Tablets, powders, cachets and capsules can be used as solid dosage forms suitable for oral administration.
Moreover, when desired or needed, suitable binders, lubricants, disintegrating agents and coloring agents may also be incorporated in the mixture. Suitable binders include starch, gelatin, natural sugars, corn sweeteners, natural and synthetic gums such as acacia, sodium alginate, carboxymethylcellulose, polyethylene glycol and waxes. Among the lubricants there may be mentioned for use in these dosage forms, boric acid, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrants include starch, methylcellulose, guar gum, and the like. Sweetening and flavoring agents and preservatives may also be included where appropriate.
Liquid form preparations include solutions, suspensions and emulsions and may include water or water-propylene glycol solutions for parenteral injection.
Liquid form preparations may also include solutions for intranasal administration. Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas.
Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for either oral or parenteral administration.
Such liquid forms include solutions, suspensions and emulsions.
For preparing suppositories, a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted, and the active ingredient is dispersed
homogeneously therein as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool and thereby solidify.
The Imidazopyrazine Compounds of the present invention may also be delivered transdermally. The transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
Additionally, the compositions of the present invention may be formulated in sustained release form to provide the rate controlled release of any one or more of the components or active ingredients to optimize therapeutic effects, i. e. , anti-cancer activity and the like. Suitable dosage forms for sustained release include layered tablets containing layers of varying disintegration rates or controlled release polymeric matrices impregnated with the active components and shaped in tablet form or capsules containing such impregnated or encapsulated porous polymeric matrices.
In one embodiment, the Imidazopyrazine Compound is administered orally.
In another embodiment, the Imidazopyrazine Compound is administered intravenously.
In another embodiment, the Imidazopyrazine Compound is administered topically.
In still another embodiment, the Imidazopyrazine Compounds is administered sublingually.
In one embodiment, a pharmaceutical preparation comprising at least one Imidazopyrazine Compound is in unit dosage form. In such form, the preparation is subdivided into unit doses containing effective amounts of the active components.
Compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present compositions can contain, in one embodiment, from about 0.1% to about 99% of the Imidazopyrazine Compound(s) by weight or volume. In various embodiments, the present compositions can contain, in one embodiment, from about 1% to about 70% or from about 5% to about 60% of the Imidazopyrazine Compound(s) by weight or volume.
The quantity of Imidazopyrazine Compound in a unit dose of preparation may be varied or adjusted from about 0.1 mg to about 5000 mg. In various embodiments, the quantity is from about 10 mg to about 5000 mg, about 10 mg to about 1000 mg, 1 mg to about 500 mg, 1 mg to about 100 mg, and 1 mg to about 50 mg.
For convenience, the total daily dosage may be divided and administered in portions during the day if desired. In one embodiment, the daily dosage is administered in one portion. In another embodiment, the total daily dosage is administered in two divided doses over a 24 hour period. In another embodiment, the total daily dosage is administered in three divided doses over a 24 hour period. In still another embodiment, the total daily dosage is administered in four divided doses over a 24 hour period.
For administration to human patients, the amount and frequency of
administration of the Imidazopyrazine Compounds will be regulated according to the judgment of the attending clinician considering such factors as age, condition and size of the patient as well as severity of the symptoms being treated. Generally, a total daily dosage of the Imidazopyrazine Compounds range from about 0.1 to about 5000 mg per day, although variations will necessarily occur depending on the target of therapy, the patient and the route of administration. In one embodiment, the dosage is from about 1 to about 200 mg/day, administered in a single dose or in 2-4 divided doses. In another embodiment, the dosage is from about 10 to about 5000 mg/day, administered in a single dose or in 2-4 divided doses. In another embodiment, the dosage is from about 100 to about 5000 mg/day, administered in a single dose or in 2-4 divided doses. In still another embodiment, the dosage is from about 500 to about 5000 mg/day, administered in a single dose or in 2-4 divided doses.
The compositions of the invention can further comprise one or more additional therapeutic agents, selected from those listed above herein. Accordingly, in one embodiment, the present invention provides compositions comprising: (i) at least one Imidazopyrazine Compound or a pharmaceutically acceptable salt thereof; (ii) one or more additional therapeutic agents that are not a Imidazopyrazine Compound; and (iii) a pharmaceutically acceptable carrier, wherein the amounts in the composition are together effective to treat disease or disorder associated with dysregulated mTOR activity, such as a cancer. In Vitro and In Vivo METHODS:
The present invention also provides methods of using the imidazopyrazine compounds of the present invention for inducing terminal differentiation, cell growth arrest and/or apoptosis of neoplastic cells thereby inhibiting the proliferation of such cells. The methods can be practiced in vivo or in vitro.
In one embodiment, the present invention provides in vitro methods for selectively inducing terminal differentiation, cell growth arrest and/or apoptosis of neoplastic cells, thereby inhibiting proliferation of such cells, by contacting the cells with an effective amount of any one or more of the imidazopyrazine compounds described herein.
In a particular embodiment, the present invention relates to an in vitro method of selectively inducing terminal differentiation of neoplastic cells and thereby inhibiting proliferation of such cells. The method comprises contacting the cells under suitable conditions with an effective amount of one or more of the imidazopyrazine compounds described herein.
In another embodiment, the invention relates to an in vitro method of selectively inducing cell growth arrest of neoplastic cells and thereby inhibiting proliferation of such cells. The method comprises contacting the cells under suitable conditions with an effective amount of one or more of the imidazopyrazine compounds described herein.
In another embodiment, the invention relates to an in vitro method of selectively inducing apoptosis of neoplastic cells and thereby inhibiting proliferation of such cells. The method comprises contacting the cells under suitable conditions with an effective amount of one or more of the imidazopyrazine compounds described herein.
In another embodiment, the invention relates to an in vitro method of inducing terminal differentiation of tumor cells in a tumor comprising contacting the cells with an effective amount of any one or more of the imidazopyrazine compounds described herein.
Although the methods of the present invention can be practiced in vitro, it is contemplated that the preferred embodiment for the methods of selectively inducing terminal differentiation, cell growth arrest and/or apoptosis of neoplastic cells, and of inhibiting mTor will comprise contacting the cells in vivo, i.e., by administering the compounds to a subject harboring neoplastic cells or tumor cells in need of treatment.
Thus, the present invention provides in vivo methods for selectively inducing terminal differentiation, cell growth arrest and/or apoptosis of neoplastic cells in a subject, thereby inhibiting proliferation of such cells in the subject, by administering to the subject an effective amount of any one or more of the imidazopyrazine compounds described herein. In a particular embodiment, the present invention relates to a method of selectively inducing terminal differentiation of neoplastic cells and thereby inhibiting proliferation of such cells in a subject. The method comprises administering to the subject an effective amount of one or more of the imidazopyrazine compounds described herein.
In another embodiment, the invention relates to a method of selectively inducing cell growth arrest of neoplastic cells and thereby inhibiting proliferation of such cells in a subject. The method comprises administering to the subject an effective amount of one or more of the imidazopyrazine compounds described herein.
In another embodiment, the invention relates to a method of selectively inducing apoptosis of neoplastic cells and thereby inhibiting proliferation of such cells in a subject. The method comprises administering to the subject an effective amount of one or more of the imidazopyrazine compounds described herein.
In another embodiment, the invention relates to a method of treating a patient having a tumor characterized by proliferation of neoplastic cells. The method comprises administering to the patient one or more of the imidazopyrazine compounds described herein. The amount of compound is effective to selectively induce terminal differentiation, induce cell growth arrest and/or induce apoptosis of such neoplastic cells and thereby inhibit their proliferation.
Kits
Another aspect of this invention is a kit comprising a therapeutically effective amount of at least one Imidazopyrazine Compound, or a pharmaceutically acceptable salt of said compound, and a pharmaceutically acceptable carrier, vehicle or diluent.
Yet another aspect of this invention is a kit comprising an amount of at least one Imidazopyrazine Compound, or a pharmaceutically acceptable salt of said compound and an amount of at least one additional anti-cancer agent listed above, wherein the amounts of the two or more active ingredients result in a desired therapeutic effect. In one embodiment, the at least one Imidazopyrazine Compound and the at least one additional anti-cancer agent are provided in the same container. In one embodiment, the at least one Imidazopyrazine
Compound and the at least one additional anti-cancer agent are provided in separate containers.
The invention is illustrated in the examples in the Experimental Details Section that follows. This section is set forth to aid in an understanding of the invention but is not intended to, and should not be construed to limit in any way the invention as set forth in the claims which follow thereafter.
EXPERIMENTAL DETAILS SECTION
Solvents, reagents, and intermediates that are commercially available were used as received. Reagents and intermediates that are not commercially available were prepared in the manner as described below. 1H NMR spectra were obtained on a Varian spectrometer (400 MHz and 500 MHz) are reported as ppm down field from Me4Si with number of protons, multiplicities, and coupling constants, in Hertz indicated parenthetically. Where LC/MS data are presented, analyses was performed using an Agilent 1100 Series LC w/ MicroMass Quattro MS Varian Pursuit XRs CI 8, 5micron, 150mm x 4.6mm ID gradient flow (0.1% TFA or 0.2% FA): 0 min - 5% ACN, 7.5 min - 100% ACN, 8.5 min -100 ACN, 8.51 min - 5% ACN, 10 min - stop 3 ml/min. The retention time and observed parent ion are given. Where the description indicates the reaction mixture was purified by HPLC, the description refers to using a preparative Agilent 1 100 Series LC/MSD SL system: Column Reverse Phase- Varian Pursuit XRs 10DC-18 250 X 21.2mm; elution with gradient Acetonitrile/water with 0.1%TFA or 0.2% formic acid. The desired product was detected and collected by a mass-triggered automatic sample collector. Flash column chromatography was performed using pre-packed normal phase silica from Biotage, Inc.
The following solvents, reagents and reaction conditions may be referred to by their abbreviations:
Aq: aqueous
g or gm: grams
psi: pounds per square inch
pH: concentration of hydronium ions in a solution
°C: degrees Celsius
h: hours
THF: Tetrahydrofuran
Et20: diethyl ether
SEM: 2-(trimethylsilyl)ethoxymethyl
LC-MS: Liquid chromatography mass spectrometry
DCM: dichloromethane
N: Normal
ml: milliliter
NBS: N-Bromosuccinimide
NCS: N-Chlorosuccinimide NIS: N-iodosuccinimide
r. : room temperature
MeOH: methanol
DIEA: diisopropylethylamine
EtOAc: ethyl acetate
EtOH: ethanol
DMF: dimethylformamide
wt%: weight percent
m/z: mass per charge
LiOH: lithium hydroxide
DMSO: dimethylsulfoxide
HPLC: high performance liquid chromatography
IPA: isopropanol
Ret: retention
Rt: retention time
RP: reverse phase
ACN: acetonitrile
CH3CN: acetonitrile
MeCN: acetonitrile
Mel: iodomethane
r.t.: room temperature
pTSA: para-toluene sulfonic acid
CDI: N,N'-carbonyldiimidazole
mg: milligram
PMA: phosphomolybdic acid
LiHMDS: Lithium bis(trimethylsilyl)amide
HMDS: hexamethyldisilazane
Pd/C: palladium on carbon
H2: hydrogen gas
PdCl2(dppf): [l ,l '-bis(diphenylphosphino)ferrocene] dichloropalladium(II) μιηοΐ: micromole
TFA: trifluoroacetic acid
NMP: N-methyl-2-pyrrolidone
min: minute DME: dimethylethane
AcOH: acetic acid
BBN: 9-borabicyclo[3.3.1]nonane
BOC: tertiary-butyloxycarbonyl
M: Molar
mmol: millimolar
DIE A: diisopropylethylamine
Bu3SnCN: tributyltin cyanide
Pd[P(t-Bu)3]2: bis(tributyl)Phosphine) palladium
Pd(PPh3)4: tetrakis(triphenylphosphine) palladium
EDCI: l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
UV: ultraviolet
LDA: lithium diisopropylamide
Tf: trifluoromethanesulfonyl
General Scheme
Figure imgf000053_0001
VII VIII IX Substituted 6-bromoimidazo[l ,2-a]pyrazine I is reacted with boronic ester II in an appropriate solvent or solvent mixture such as Dioxane-H20 in the presence of appropriate catalyst and base to provide the corresponding intermediate III (General Scheme).
Intermediate III was hydrogenated in the presence of 10% Pd/C in an appropriate solvent or solvent mixture such as EtOH to provide a mixture of the corresponding intermediate IV and IV. The mixture of intermediate IV and IV could be transferred to V when treating with appropriate base such as KOtBu. Intermediate V is reacted with N-Iodosuccinimide (NIS) in an appropriate solvent or solvent mixture such as DMF to provide the corresponding iodide intermediate VI. Iodide intermediate VI is heated with boronic acid or boronic ester in an appropriate solvent or solvent mixture such as Dioxane-H20 in the presence of appropriate catalyst and base to provide the corresponding intermediate VII. Intermediate VII is treated with an appropriate acid, such as TFA, to afford the corresponding intermediate VIII.
Intermediate VIII is then transferred to IX.
Example 1
Preparation of (( 1 R,3s,5 S)-3 -(8-amino-3-(6-phenylpyridin-3 -yl)imidazo[ 1 ,2-a]pyrazin-i
azabicyclo[3.2. l]octan-8-yl)(4H-l ,2,4-triazol-3-yl)methanone
Step 1 : Preparation of 6-bromo-8-[bis[(l,l -dimethylethoxy)carbonyl]amino]imidazo[l,2- a]pyrazine
Figure imgf000054_0001
6-bromo-8-[bis[(l , 1 -dimethylethoxy)carbonyl] amino] imidazo[l ,2-a]pyrazine was prepared from 6-bromoimidazo[l ,2-a]pyrazin-8-amine according to reference procedure for similar compound (Lind, Kenneth E. etc. WO2008005457).
Step 2: Preparation of (lR,5S)-tert-butyl 3-(8-(bis(tert-butoxycarbonyl)amino)imidazo[l ,2- a]pyrazin-6-yl)-8-azabicyclo[3.2.1 ]oct-3-ene-8-carboxylate
Figure imgf000055_0001
To a mixture of 6-bromo-8-[bis[(l,l-dimethylethoxy)carbonyl]amino]imidazo[l,2- ajpyrazine (1057.3 mg, 2.57 mmol) in dioxane (14 niL) and H20 (1.4 mL) was added tert- butyl 3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-8-azabicyclo[3.2.1]oct-2-ene-8- carboxylate (1720.2 mg, 5.13 mmol), K3P04 (1631.9 mg, 7.70 mmol), and
PdCl2(dppf)-CH2Cl2 (209.4 mg, 0.25 mmol). The reaction was heated at 60°C for 30 min under microwave condition, at which time LC/MS analysis confirmed full consumption of starting material. On cooling, filtration through celite and concentration gave a crude product. Gradient column chromatography on silica provided (lR,5S)-tert-butyl 3-(8-(bis(tert- butoxycarbonyl)amino)imidazo[l,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]oct-3-ene-8- carboxylate: LCMS tR = 1.43 Min (5 min run, UV254nm). Mass calculated for, M+ 541.2, observed m/z 542.2 (M+H).
Step 3: Preparation of (lR,3s,5S)-tert-butyl 3-(8-(bis(tert-butoxycarbonyl)amino)imidazo[l,2- a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate and (lR,3r,5S)-tert-butyl 3-(8- (bis(tert-butoxycarbonyl)amino)imidazo[l ,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8- carboxylate
Figure imgf000055_0002
A mixture of (lR,5S)-tert-butyl 3-(8-(bis(tert-butoxycarbonyl)amino)imidazo[l ,2- a]pyrazin-6-yl)-8-azabicyclo[3.2.1]oct-3-ene-8-carboxylate (1024 mg, 1.89 mmol), 10% Pd/C (100 mg) in EtOH (15 ml) was stirred at room temperature under hydrogen (balloon pressure) for 2 days. Filtration and concentration provided a mixture of two isomers: (lR,3s,5S)-tert- butyl 3-(8-(bis(tert-butoxycarbonyl)amino)imidazo[l,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octane-8-carboxylate and (lR,3r,5S)-tert-butyl 3-(8-(bis(tert- butoxycarbonyl)amino)imidazo[l ,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate, which was used for step 4. Step 4: Preparation of (lR,3s,5S)-tert-butyl 3-(8-(tert-butoxycarbonylamino)-3- iodoimidazo l,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000056_0001
At 0°C, KOtBu (1.0M in THF, 3.14 mL, 3.14 mmol) was added dropwise to a mixture of (lR,3s,5S)-tert-butyl 3-(8-(bis(tert-butoxycarbonyl)amino)imidazo[l,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octane-8-carboxylate and (lR,3r,5S)-tert-butyl 3-(8-(bis(tert- butoxycarbonyl)amino)imidazo[l ,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate (852 mg, 1.57 mmol) in THF (20 mL) in 5 min. The mixture was stirred at 0°C for another 10 min, diluted with H20 and then extracted with ethyl acetate (x2). The combined organic layers were washed with brine and dried with Na2S04. Evaporation afforded crude (lR,3s,5S)-tert- butyl 3-(8-(tert-butoxycarbonylamino)imidazo[l,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane- 8-carboxylate: LCMS tR = 1.09 Min (5 min run, UV25 nm). Mass calculated for, M+ 443.2, observed m/z 444.2 (M+H).
The crude (lR,3s,5S)-tert-butyl 3-(8-(tert-butoxycarbonylamino)imidazo[l,2- a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate was heated with NIS (529.5 mg, 2.35 mmol) in DMF (10 mL) at 70°C for 1.5 h. The mixture was diluted with H20 and then extracted with ethyl acetate (x2). The combined organic layers were washed with brine and dried with Na2S04. Evaporation and purification by column chromatography afforded
(lR,3s,5S)-tert-butyl 3-(8-(tert-butoxycarbonylamino)-3-iodoimidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octane-8-carboxylate: LCMS tR = 1.45 Min (5 min run, UV254nm)- Mass calculated for, M+ 569.1, observed m/z 570.1 (M+H). Step 5: Preparation of (lR,3s,5S)-tert-butyl 3-(8-(tert-butoxycarbonylamino)-3-(6- phenylpyridin-3-yl)imidazo[l,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate
Figure imgf000057_0001
2-phenyl-5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)pyridine (136.6 mg, 0.49 mmoL), K3P04 (154.5 mg, 0.73 mmol), and PdCl2(dppf)-CH2Cl2 (19.8 mg, 0.024 mmol) was added to a mixture of (lR,3s,5S)-tert-butyl 3-(8-(tert-butoxycarbonylamino)-3- iodoimidazo[l,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octane-8-carboxylate (138.3 mg, 0.24 mmol) in dioxane (5 mL) and H20 (0.5 mL). The resulting solution was stirred at 70° C under microwave condition for 30 min. The mixture was filtered through celite and then
concentrated. Purification by column chromatography afforded (lR,3s,5S)-tert-butyl 3-(8- (tert-butoxycarbonylamino)-3-(6-phenylpyridin-3-yl)imidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octane-8-carboxylate: LCMS tR = 1.48 Min (5 min run, UV254nm)- Mass calculated for, M+ 596.3, observed m/z 597.3 (M+H).
Step 6: Preparation of 6-((lR,3s,5S)-8-azabicyclo[3.2.1]octan-3-yl)-3-(6-phenylpyridin-3- yl)imidazo[l ,2-a]pyrazin-8-amine
Figure imgf000057_0002
At 0°C, CH2C12 (2 mL) and then TFA (2 mL) was added to (lR,3s,5S)-tert-butyl 3-(8- (tert-butoxycarbonylamino)-3-(6-phenylpyridin-3-yl)imidazo[l,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octane-8-carboxylate (144 mg, 0.24 mmol). After stirring at room
temperature for 30 min, the mixture was concentrated to afford 6-((lR,3s,5S)-8- azabicyclo [3.2.1] octan-3 -yl)-3 -(6-phenylpyridin-3 -yl)imidazo [ 1 ,2-a]pyrazin-8-amine which was used for next step without further purification: LCMS tR = 0.75 Min (5 min run, UV254nm)- Mass calculated for, M+ 396.2, observed m/z 397.1 (M+H). Step 7: Preparation of ((lR,3s,5S)-3-(8-amino-3-(6-phenylpyridin-3-yl)imidazo[l,2-a]pyrazin- 6-yl)-8-azabicyclo[3.2.1]octan-8-yl)(4H-l ,2,4-triazol-3-yl)methanone
Figure imgf000058_0001
A mixture of crude 6-((lR,3s,5S)-8-azabicyclo[3.2.1]octan-3-yl)-3-(6-phenylpyridin- 3-yl)imidazo[l,2-a]pyrazin-8-amine from step 7, lH-l ,2,4-triazole-3-carboxylic acid (33.0 mg, 0.29 mmol), EDCI (92.9 mg, 0.49 mmol), 1-hydroxybenzotriazole (65.6 mg, 0.49 mmol) and DIEA (253 ul, 1.46 mmol) in DMF (5 mL) was stirred at room temperature for lh.
Purification with prep-LC and conversion to HCl salt provided ((lR,3s,5S)-3-(8-amino-3-(6- phenylpyridin-3-yl)imidazo[l,2-a]pyrazin-6-yl)-8-azabicyclo[3.2.1]octan-8-yl)(4H-l ,2,4- triazol-3-yl)methanone as HCl salt: LCMS tR = 2.25 Min (10 min run, UV254nm). Mass calculated for, M+ 491.2, observed m/z 492.3 (M+H).
Example 2
By applying the chemistry described in Example 1, steps 5-7, and using suitable starting materials, compounds 2-7 were synthesized.
Figure imgf000058_0002
Figure imgf000059_0001
azabicyclo[3.2.1 ] octan
Figure imgf000060_0001
Example 3
mTOR Kinase Assay
Methods: An HTRF mTOR enzyme assay was developed to assess the compounds' inhibitory activity. The mTOR assay buffer contained 10 mM Hepes (pH 7.4), 50 mM NaCl,
100 g/ml BSA, 50 mM μ-glycerophosphate, 10 mM MnCl2 and 0.5 mM DTT. An active truncated mTOR enzyme was prepared similarly to that reported by Toral-Barza et al., Biochemical and Biophysical Research Communications 332, pp 304-310 (2005). 20 ng of human mTOR enzyme (< 5 % pure was preincubated with the compound for 10 minutes followed by the addition of 5 μΜ ATP and 0.2 μΜ GST-S6K (Zhang et al., Protein
Expression and Purification 46, pp 414^420 (2006)). The reaction was incubated for one hour at 30°C. Anti phospho p70-S6K(Thr389) (-1.7 ng/well, anti-phospho-p70S6K-cryptate (Pho- p70S6-Kin-K cat# 64CUSKAY, from Cisbio)) and anti GST-XL665 (1 : 1 Ratio with the substrate GST-S6K, anti GST-XL665, cat# 61GSTXLB) Cisbio) were added after the reaction was stopped. The plates were read (PHERAstar, BMG) at least 2 hours after adding the anti phospho p70-S6K and the anti GST-XL665.
IC50 DETERMINATIONS: Dose-response curves were plotted from the inhibition data generated, each in duplicate, from 8 point serial dilutions of inhibitory compounds.
Concentration of compound was plotted against the HTRF em665/em590 ratio signal. To generate IC50 values, the dose-response curves were fitted to a standard sigmoidal curve and IC50 values were derived by nonlinear regression analysis. Example 4
mTOR Target Engagement Assay
The target engagement of mTOR kinase inhibitors was evaluated using an
immuno fluorescent cell-based assay. In this assay, inhibition of mTORCl activity was measured by the reduction in the level of phosphorylated 4E-BPlThr37/46 (p4E-BPl
Thr37/46), and inhibition of mTORC2 activity was measured by the reduction of
phosphorylated AKTSer473 (pAKT S473).
PC3 cells (prostate tumor cell-line that contains a mutation in the tumor suppressor PTEN, that promotes the phosphorylation and activation of AKT and 4E-BP1) were used in the immunofluorescence assay. PC3 cells were seeded on 384 well plates (black clear bottom, Matrix #4332) overnight. PC3 cells were then treated with 40 μΐ of the serially diluted test compounds (in 5% fetal bovine serum, F12 medium containing 0.25% DMSO) for ninety minutes at 37°C. The test compound solution was removed, and the plates were washed gently two times with 25 μΐ phosphate buffered saline (PBS). The cells were fixed by adding 25 μΐ of Prefer reagent (from Anatech LTD, Cat#414, a glyoxal fixative for fixing proteins within a cell) for sixty minutes followed by three washes with PBS. 5% Goat serum in
PBS/0.3% Triton was used to block non-specific binding (60 minutes).
The primary antibodies targeting pAKT S473 and p4E-BPl Thr37/46 were diluted into PBS/0.3% Triton and incubated with the cells overnight at 4°C. The antibodies targeting pAKTS473 (Cat# 4085, Cell signaling) and p4E-BPl Thr37/46 (Cat#2855, Cell signaling) were used at a 1 : 100 dilution. Plates were washed three times with PBS/0.1 % Tween 20 before adding the secondary antibody at a 1 :200 dilution, (goat anti-rabbit containing a fluorescent label, Alexa Fluor 488, Cat# Al 1008, Invitrogen) in PBS/0.3% Triton for 60 minutes. Finally, the plates were washed three times with PBS/0.1% Tween 20 and the fluorescent intensity was read using an Analyst HT from Molecular Devices. The fluorescent intensity values from the serially diluted compound treatment group were analyzed using the Xlfit 4 program (Microsoft) (Formula 205: Y=Bottom+(Top-Bottom)/(l+(IC50/X)AHillslope) to generate the IC5o value. Where Top is the maximum signal without Compound (+ DMSO only) and Bottom represents maximum inhibition. Y is the fluorescence at some compound concentration. The control used to determine the fluorescent intensities for 100% pAKT S473 or 100% phosphorylated p4E-BPl Thr37/46 were measured from untreated wells that contained only DMSO, instead of test compound.
The above tables list representative compounds of the invention with activity data whereby the IC50 values are rated "A", "B," "C," or "D." The IC50 values are rated "A" for IC50 values in the range of 1 nM to 100 nM, "B" for IC50 values in the range from 100 nM to 1000 nM, "C" for IC50 values in the range from 1000 nM to 10000 nM, "D" for IC50 values more than 10000 nM.
While this invention has been particularly shown and described with references to embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the meaning of the invention described. Rather, the scope of the invention is defined by the claims that follow.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula I
Figure imgf000063_0001
I
U is N or CH;
W is absent, or W is selected from the group consisting of C(O), S(O), S(0)2, C 1 -C4 alkylene, C3-C8 cycloalkyl, phenyl, 5- to 6-membered heteroaryl, and 3- to 8-membered heterocyclyl;
V is absent, or V is selected from the group consisting of C(O), O, S, N(H), N(C]-C3 alkyl), N(C3-C8 cycloalkyl), S(O), S(0)2, and C,-C4 alkylene;
or W and V together form a C3-C cycloalkyl, phenyl, 5- to 6-membered heteroaryl, or 3 to 8-membered heterocyclyl ring;
R1 is selected from the group consisting of
(i) Ci-C6 alkyl or C3-Ci0 cycloalkyl, wherein said alkyl or cycloalkyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of hydroxy, C]-C6 alkoxy, halo, trifluoromethyl, carboxy, 5- to 6-membered heteroaryl, -S02H, Ci-C6 alkyl-C(0)-NH-, Ci-C6 alkyl- S02-NH-, and Ci-C6 alkyl-SO-NH-;
(ii) 3- to 8-membered heterocyclyl wherein said heterocyclyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of halo, Ci-C6 alkyl, C]-C6 alkoxy, cyano, hydroxy, amino, Ci-C6 alkylamino, and Ci-C6 dialkylamino;
(iii) C6-C10 aryl or 5- to 10-membered heteroaryl, wherein said aryl or heteroaryl of R1 is unsubstituted or is substituted with one to three moieties independently selected from the group consisting of halo, trifluoromethyl, trifluoromethoxy, C]-C6 alkyl, C]-C6 alkoxy, cyano, hydroxy, amino, Ci-C6 alkylamino, and Ci-C6 dialkylamino; and
(iv) -N(H)OH or -N(H)-d-C3 alkoxy; L and Z are bonded to any two carbons of the ring and are independently selected from the group consisting of CH2, C(H)(R10),C(R10)(Rn), N(R10), C(O), O, S, S(O) and S(0)2;
T is not present such that L is bonded directly to Z, or T is selected from the group consisting of CH2, C(H)(R10),C(R10)(Rn), N(R10), C(O), O, S, S(O) and S(0)2 and CrC4 alkylene, wherein said alkylene of T is unsubstituted or substituted with 1 to two substituents selected from the group consisting of Ci-C3 alkyl, halo, hydroxyl, C]-C3 alkoxy, amino, Ci-C3 alkylamino and Ci-C3 dialkylamino;
Ra and Rb are independently selected from H, halogen and C]-C6 alkyl;
R3 is selected from the group consisting of H, halogen, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halo-d-C6alkyl, -CF3, -C(0)R9, C6-C10aryl, C3-C8cycloalkyl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclyl, 5- to 10-membered heterocyclenyl, C6-CioarylCi- C6alkyl, C3-C8cycloalkylCi-C6alkyl, 5- to 10-membered heteroarylCi-C6alkyl, 5- to 10- membered heterocyclylCi-C6alkyl and 5- to 10-membered heterocyclenylCi-C6alkyl, wherein each of said aryl, cycloalkyl, heteroaryl, heterocyclyl, heterocyclenyl, arylalkyl,
cycloalkylalkyl, heteroarylalkyl, heterocyclylalkyl and heterocyclenylalkyl is unsubstituted or substituted with one to three moieties which can be the same or different, each moiety being selected from the group consisting of halogen, Q-Qalkyl, C3-C8cycloalkyl, -CF3, -CN, - C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -(CRaRb)nOR9, -(CRaRb)nC(0)R9, -(CRaRb)nNR8R9, -
(CRaRb)nNR8,
-NR8R9, -(CRaRb)nC(0)0-Ci-C6alkyl, -0-haloC,-C6alkyl, -(CRaRb)nC(0)NR8R9, -(CRaRb)n C(0)NR8S(0)2R9, -(CRaRb)nNR8C(0)R9, -(CRaRb)nNR8C(0)OR9,
-(CRaRb)nNR8C(0)NR8R9, -(CRaRb)nS(02)NR8R9, -(CRaRb)nS(02)NR8C(0)R9, -(CRaRb)n NR8S(02)R9, -(CRaRb)nSR9, -(CRaRb)nS(02)R9, C6-C10aryl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclenyl, 5- to 10-membered heterocyclyl, C6-Ci0arylalkyl, 5- to 10- membered heteroarylalkyl, 5- to 10-membered heterocyclenylalkyl and 5- to 10-membered heterocyclylalkyl wherein each of said alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, heterocyclenyl, arylalkyl, cycloalkylalkyl, heteroarylalkyl, heterocyclylalkyl and heterocyclenylalkyl is unsubstituted or substituted with one to five moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Cj- C6alkyl, C3-C8cycloalkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O-haloCi- C6alkyl, -OR9, -C(0)R9, -NR8R9, -C(0)0-C1-C6alkyl, -C(0)NR8R9, -NR8C(0)R9, - S(02)NR8R9, -NR8S(02)R9, -SR9, and -S(02)R9;
R6 is independently selected from the group consisting of H, halogen, Ci-C6alkyl, C3- Cgcycloalkyl, -CF3, -CN, -(CRaRb)nC(0)OH, -OCF3, -OR9, -C(0)R9, -NR8R9, -C(0)0-Ci- C6alkyl, -CRaRb, -ORa, -S(0)Ra, -C(0)ORa, -S(02)NRaRb, -NRaC(0)Rb, -NRaS(02)Rb,- C(0)NR8R9, -SR9, and -S(02)R9;
R8 and R9 are independently selected from the group consisting of H, OH, Ci-C6alkyl, C3- C8cycloalkyl, C6-Ci0aryl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclenyl, 5- to 10-membered heterocyclyl, C3-C8cycioalkylCi-C6alkyl, C6-CioarylCi-C6aikyl, 5- to 10- membered heteroarylCi-C6alkyl, 5- to 10-membered heterocyclylCi-Cealkyl, 5- to 10- membered heterocyclenylCi-C alkyl, and said alkyl, cycloalkyl, aryl, heteroaryl,
heterocyclenyl, heterocyclyl, cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclenylalkyl or heterocyclylalkyl is optionally substituted with halogen, Ci-C6alkyl, C3-C8cycloalkyl, -CF3, -CN, -(CRaRb)nC(0)OH, -OCF3, -ORa, -C(O), amino, -C(0)0-Ci-C6alkyl, -C(0)NRaRb,-SRa, and -S(02)Ra; or R8 and R9 together with the nitrogen atom to which they are attached form a
3- to 6-membered heterocyclyl ring;
R10 and R11 are independently selected from the group consisting of H, C]-C3alkyl, halo, hydroxyl, Ci-C3alkoxy, amino, Ci-C3alkylamino and Ci-C3dialkylamino; n is independently 0, 1, 2, 3 or 4;
m is independently 0, 1 , 2, 3 or 4;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, wherein
L and Z are both CH2i and T is not present;
Ra and Rb are independently selected from H and C]-C6 alkyl; R3 is selected from the group consisting of C6-C!0aryl, 5- to 10-membered heteroaryl, wherein each of said aryl or heteroaryl is unsubstituted or substituted with one to three moieties which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, C6-Ci0aryl, 5- to 10-membered heteroaryl, -CF3, -CN,
-C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloC C6alkyl, -OR8, -C(0)R8, -NR8R9,
-C(0)0-Ci-C6alkyl, -C(0)NR8R9, -NR8C(0)R9, -S(02)NR8R9, -NR8S(02)R9, -SR8, and -S(02)R8, wherein each of said heteroaryl or aryl is unsubstituted or substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, C C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O- haloC,-C6alkyl, -ORa, -C(0)Ra, -NRaRb, -C(0)0-Ci-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, - S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra;
R6 is independently selected from the group consisting of H, -ORa, -NRaR , -SRa, -S(0)Ra, - S(02)Ra, -C(0)C, -C6alkyl, -C(0)NRaRb, -C(0)ORa, -S(02)NRaRb, -NRaC(0)Rb, -
NRaS(02)Rb, C!-C6alkyl, C C6alkoxy, halogen, hydroxyl, amino and -CN;
R and R are independently selected from the group consisting of H, OH, Ci-C6alkyl, C3- C8cycloalkyl, C6-Cioaryl, 5- to 10-membered heteroaryl, 5- to 10-membered heterocyclenyl, 5- to 10-membered heterocyclyl, C3-C8cycloalkylCi-C6alkyl, C6-Ci0arylCi-C6alkyl, 5- to 10- membered heteroarylCi-C6alkyl, 5- to 10-membered heterocyclylC]-C6alkyl, 5- to 10- membered heterocyclenylCi-C alkyl, and said alkyl, cycloalkyl, aryl, heteroaryl,
heterocyclenyl, heterocyclyl, cycloalkylalkyl, arylalkyl, heteroarylalkyl, heterocyclenylalkyl or heterocyclylalkyl is optionally substituted with halogen, Ci-C6alkyl, C3-C8cycloalkyl, -CF3, -CN, -(CRaRb)nC(0)OH, -OCF3, -ORa, -C(O), amino, -C^O-Q-Cealkyl, -C(0)NRaRb,-SRa, and -S(02)Ra; n is independently 0, 1 or 2;
m is 1 ; or a pharmaceutically acceptable salt thereof.
3. The compound of any one of claims 1 to 2, wherein W is C(O).
4. The compound of any one of claims 1 to 3, wherein V is absent.
5. The compound of any one of claims 1 to 4, wherein R1 is selected from the group consisting of
(i) C]-C6 alkyl or C3-Ci0 cycloalkyl, wherein said alkyl or cycloalkyl of R1 is unsubstituted or substituted with one to three moieties independently selected from the group consisting of hydroxy, Ci-C6 alkoxy, fluoro, trifluoromethyl, carboxy, tetrazolyl, -S02H, Cj- C6 alkyl-C(0)-NH-, CrC6 alkyl-S02-NH-, and Ci-C6 alkyl-SO-NH-; and
(ii) 5- to 6-membered heterocyclyl containing 1 to 3 heteroatoms selected from the group consisting of N, O, S, and S(0)2 wherein said heterocyclyl of R1 is unsubstituted or substituted with one to two moieties independently selected from the group consisting of halo, C]-C6 alkyl, Cj-C alkoxy, cyano, hydroxy, amino, Ci-C6 alkylamino, and C]-C6
dialkylamino.
6. The compound of any one of claims 1 to 5, wherein R6 is amino.
7. The compound of any one of claims 1 to 6, wherein
R3 is a 5- to 6-membered heteroaryl or phenyl unsubstituted or substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, phenyl, 5- to 6-membered heteroaryl, -CF3, -CN,
-C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloC,-C6alkyl, -ORa, -C(0)Ra, -NRaRb,
-C(0)0-C,-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra, wherein the alkyl, phenyl or heteroaryl is optionally substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, C]-C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O- haloC C6alkyl, -OR8, -C(0)Ra, -NRaRb, -C(0)0-C C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, - S(02)NRaRb, -NRaS(02)R , -SRa, and -S(02)Ra;
Wherein all other substituents are as defined in any one of claims 1 to 6.
8. The compound of claim 7, wherein R3 is pyrazolyl, isoquinolinyl, pyrimidinyl, phenyl or pyridyl, unsubstituted or substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Ci- C alkyl, phenyl, 5- to 6-membered heteroaryl, -CF3, -CN,
-C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O-halod-Cealkyl, -ORa, -C(0)Ra, -NRaRb, -C(0)0-C C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra, wherein the alkyl, phenyl or heteroaryl is optionally substituted with one to three moieties, which can be the same or different, each moiety being selected from the group consisting of halogen, Ci-C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O- haloC1-C6alkyl, -ORa, -C(0)Ra, -NRaRb, -C(0)0-C,-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -
S(02)NRaRb, -NRaS(02)Rb, -SRa, and -S(02)Ra.
9. The compound of claim 8, wherein R is
Figure imgf000068_0001
1 12 Ar is phenyl or a 5- to 6-membered heteroaryl optionally substituted with one to three of R , which can be the same or different, each R12 being selected from the group consisting of halogen, Ci-C6alkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -0-haloC,-C6alkyl, - ORa, -C(0)Ra, -NRaRb, -C(0)0-Ci-C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, - NRaS(02)Rb, -SRa, and -S(02)Ra.
10. The compound of claim 9 wherein Ar1 is phenyl, pyridyl, pyrimidinyl,
imidazolyl, pyrazinyl, pyrazolyl, or thiazolyl, optionally substituted with one to three of R12, which can be the same or different, each R12 being selected from the group consisting of halogen, d-Qalkyl, -CF3, -CN, -C(0)OH, -(CRaRb)nC(0)OH, -OCF3, -O-haloQ-Cealkyl, - ORa, -C(0)Ra, -NRaRb, -C(0)0-C C6alkyl, -C(0)NRaRb, -NRaC(0)Rb, -S(02)NRaRb, -
NRaS(02)Rb, -SRa, and -S(02)Ra.
1 1. The compound of claim 9 or 10, wherein R12 is selected from the group consisting of halogen, Ci-C6alkyl, -CF3, and -OCF3.
12. A compound selected from the group consisting of:
((lR,3s,5S)-3-(8-amino-3-(6-phenylpyridin-3-yl)imidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1 ]octan-8-yl)(4H- 1 ,2,4-triazol-3-yl)methanone;
((lR,3r,5S)-3-(8-amino-3-(6-phenylpyridin-3-yl)imidazo[l,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octan-8-yl)(4H-l,2,4-triazol-3-yl)methanone; l-((lR,3s,5S)-3-(7-amino-6-fluoro-3-(6-phenylpyridin-3-yl)pyrazolo[l ,5-a]pyrimidin-5-yl)-8- azabicyclo[3.2.1 ]octan-8-yl)-2-hydroxyethanone;
(( 1 R,3 s,5 S)-3 -(8-amino-3 -(6-( 1 -methyl- 1 H-pyrazol-3 -yl)pyridin-3 -yl)imidazo [ 1 ,2-a]pyrazin- 6-yl)-8-azabicyclo[3.2.1]octan-8-yl)(4H-l,2,4-triazol-3-yl)methanone;
((lR,3s,5S)-3-(8-amino-3-(l-phenyl-lH-pyrazol-4-yl)imidazo[l ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2. l]octan-8-yl)(4H-l ,2,4-triazol-3-yl)methanone;
1 -(( 1 R,3 s,5 S)-3 -(3-(6-( 1 H-imidazol-2-yl)pyridin-3 -yl)-8-aminoimidazo[ 1 ,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1]octan-8-yl)-2-hydroxyethanone; and
((lR,3s,5S)-3-(3-(6-(lH-imidazol-2-yl)pyridin-3-yl)-8-aminoimidazo[l,2-a]pyrazin-6-yl)-8- azabicyclo[3.2.1 ]octan-8-yl)(4H- 1 ,2,4-triazol-3-yl)methanone;
Or a stereoisomer thereof;
Or a pharmaceutically acceptable salt thereof;
Or a pharmaceutically acceptable salt of the stereoisomer thereof.
13. A pharmaceutical composition comprising a therapeutically effective
amount of the compound of any one of claims 1-12 and a pharmaceutically acceptable carrier and optionally other therapeutic ingredients.
14. A compound according to any one of claims 1-13 for the treatment of cancer.
PCT/US2012/047522 2011-07-26 2012-07-20 NOVEL IMIDAZO[1,2-A]PYRAZINE DERIVATIVES AS mTOR INHIBITORS WO2013016160A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161511613P 2011-07-26 2011-07-26
US61/511,613 2011-07-26

Publications (1)

Publication Number Publication Date
WO2013016160A1 true WO2013016160A1 (en) 2013-01-31

Family

ID=47601461

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/047522 WO2013016160A1 (en) 2011-07-26 2012-07-20 NOVEL IMIDAZO[1,2-A]PYRAZINE DERIVATIVES AS mTOR INHIBITORS

Country Status (1)

Country Link
WO (1) WO2013016160A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000044755A1 (en) * 1999-01-29 2000-08-03 Abbott Laboratories Diazabicyclic derivatives as nicotinic acetylcholine receptor ligands
US20040127491A1 (en) * 2002-11-11 2004-07-01 Dan Peters Novel diazabicyclic biaryl derivatives
US20070105864A1 (en) * 2005-11-10 2007-05-10 Schering Corporation Methods for inhibiting protein kinases
WO2010069684A1 (en) * 2008-12-17 2010-06-24 Biomarin Iga, Ltd. Compounds for treatment of duchenne muscular dystrophy
WO2011090935A1 (en) * 2010-01-19 2011-07-28 Merck Sharp & Dohme Corp. PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS mTOR INHIBITORS

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000044755A1 (en) * 1999-01-29 2000-08-03 Abbott Laboratories Diazabicyclic derivatives as nicotinic acetylcholine receptor ligands
US20040127491A1 (en) * 2002-11-11 2004-07-01 Dan Peters Novel diazabicyclic biaryl derivatives
US20070105864A1 (en) * 2005-11-10 2007-05-10 Schering Corporation Methods for inhibiting protein kinases
WO2010069684A1 (en) * 2008-12-17 2010-06-24 Biomarin Iga, Ltd. Compounds for treatment of duchenne muscular dystrophy
WO2011090935A1 (en) * 2010-01-19 2011-07-28 Merck Sharp & Dohme Corp. PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS mTOR INHIBITORS

Similar Documents

Publication Publication Date Title
EP2608669B1 (en) NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012027239A1 (en) NOVEL PYRAZOLO[1,5-a]PYRROLO[3,2-e]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
EP2755482B1 (en) Combination of mk-1775 and mk-8776 for treating cancer
EP2934535B1 (en) Substituted pyrrolopyrimidines as hdm2 inhibitors
EP2935263B1 (en) Substituted imidazopyridines as hdm2 inhibitors
EP2770987B1 (en) Novel compounds that are erk inhibitors
EP2584903B1 (en) Novel heterocyclic compounds as erk inhibitors
US20210309687A1 (en) Prmt5 inhibitors
EP2699567A1 (en) Insulin-like growth factor-1 receptor inhibitors
US20140315916A1 (en) MACROCYCLES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
US20130261125A1 (en) Indazole derivatives useful as erk inhibitors
EP2953470B1 (en) 2,6,7 substituted purines as hdm2 inhibitors
EP2632462A1 (en) Novel heteroaryl-carboxamide derivatives as pdk1 inhibitors
EP2736338B1 (en) FUSED TRICYCLIC COMPOUNDS AS mTOR INHIBITORS
EP2608668B1 (en) Fused tricyclic inhibitors of mammalian target of rapamycin
WO2013148227A1 (en) Insulin-like growth factor-1 receptor inhibitors
EP2616451A1 (en) Novel thiazol-carboximide derivatives as pdk1 inhibitors
EP2632263A1 (en) Novel thiazole-carboxamide derivatives as pdk1 inhibitors
WO2013016160A1 (en) NOVEL IMIDAZO[1,2-A]PYRAZINE DERIVATIVES AS mTOR INHIBITORS
US20230219962A1 (en) PYRAZOLO[4,3-d]PYRIMIDINE DERIVATIVES AND METHODS OF USE THEREOF FOR THE TREATMENT OF CELLULAR PROLIFERATIVE DISORDERS

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12818280

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12818280

Country of ref document: EP

Kind code of ref document: A1