Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HETEROCYCLE SUBSTITUTED PURINES AS ANTIPROLIFERATIVE AGENTS
Document Type and Number:
WIPO Patent Application WO/2003/022805
Kind Code:
A2
Abstract:
The compounds of the present invention are 2,6,9-trisubstituted purine derivatives which are inhibitors of cyclin/cdk complexes. The compounds of the current invention also are potent inhibitors of human cellular proliferation. As such, the compounds of the present invention constitute pharmaceutical compositions with a pharmaceutically acceptable carrier. Such compounds are useful in treating a disorder mediated by elevated levels of cell proliferation in a mammal compared to a healthy mammal by administering to such mammal an effective amount of the compound. Examples of the compounds of the present invention are represented by the following chemical structures (I) and (II) with Y, V, A, R1, R2, R3, R4, R7, and n1 defined herein.

Inventors:
TROVA MICHAEL PETER
Application Number:
PCT/US2002/028730
Publication Date:
March 20, 2003
Filing Date:
September 09, 2002
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ALBANY MOLECULAR RES INC (US)
International Classes:
A61P35/00; C07D473/16; C07D473/18; C07D473/24; C07D473/34; (IPC1-7): C07D/
Foreign References:
JPH11222435A1999-08-17
Attorney, Agent or Firm:
Goldman, Michael L. (Clinton Square P.O. Box 3105, Rochester NY, US)
Download PDF:
Claims:
WHAT IS CLAIMED:
1. A compound of the following formula: Formula I wherein: Ri are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain ; C3C7cycloalkyl ; CH2 (C3C7cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH ; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of RI, OR,, SR1, S (O) RI, S (02) RI, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Ri, C (O) NHRI, phenyl, and C (O) NHCHR1CH2OH ; 1naphthyl ; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl; 4pyridyl; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl; thiophene3yl; 2furanyl; 3furanyl; oxazol2yl; oxazol4yl; oxazol5yl; thiazol2yl; thiazol4yl; thiazol5yl; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl; isoxazol3yl; isoxazol4yl; isoxazol5yl; isothiazol3yl; isothiazol4yl; isothiazol5yl; 1, 3, 4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl ; 2pyrrolyl; 3pyrrolyl; 1, 3,5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl; 2quinolinyl ; 3quinolinyl; 4quinolinyl; 1isoquinolinyl ; 3isoquinolinyl ; and 4isoquinolinyl ; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ru, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of: H; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2)nPh ; and (CH2)nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; ClC4straight chain alkyl ; or C3C4branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 58membered saturated or unsaturated ring; nl= 03; n= 03; A= CH2 ; (CH2) 2; (CH2) 3; OCH2CH2; or CHCH3; Y= H; OR1; N(R1)2; N(R1)C(O)R3; N (R1) C (O) R5; N (Rl) C (O) CH (R6) NH2; N (Rs) SO2R3 ; N (R1) C (O) NHR3; or N (R1)C(O) OR6; R5= C3C7cycloalkyl ; R6= ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2)Ph ; or (CH2)nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1, wherein R3 are the same or different and independently selected from the group consisting of : H; C,C4straight chain alkyl ; and C3C4branched chain alkyl ; Y= H; OR, ; N (Ri) 2 ; N (RI) C (O) R3; N (Rl) SO2R3 ; or N (RI) C (O) NHR3.
3. A compound of the following formula: Formula III wherein: Rl are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain; C3C7cycloalkyl ; CH2(C3C7cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2 ; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Rl, ORI, SRl, S (O) Rl, S (02) RI5 NHR,, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Rl, C (O) NHRI, phenyl, and C (O) NHCHRlCH2OH ; 1naphthyl ; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl; 4pyridyl; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl; thiophene3yl; 2furanyl; 3furanyl; oxazol2yl; oxazol4yl; oxazol5yl ; thiazol2yl; thiazol4yl; thiazol5yl ; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl; isoxazol3yl; isoxazol4yl; isoxazol5yl; isothiazol3yl; isothiazol4yl; isothiazol5yl; 1, 3,4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl; 2pyrrolyl; 3pyrrolyl; 1, 3,5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl; 2quinolinyl; 3quinolinyl; 4quinolinyl ; 1isoquinolinyl ; 3isoquinolinyl ; and 4isoquinolinyl ; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; n= 03 ; A= CH2; <BR> <BR> (CH2)2;<BR> (CH)2)3; OCH2CH2 ; or CHCH3; Y= H; OR1; N(R1)2; N(R1)C(O)R3; N (RI) C (O) R5 ; N (Rl) C (O) CH (R6) NH2; N (RI) SO2R3 ; N (Rl) C (O) NHR3; or N (R1) C (O) OR6 ; R3 are the same or different and independently selected from the group consisting of : H; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2)nPh ; and (CH2)nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R5= C3C7cycloalkyl ; R6= C,C4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; or (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan ; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof.
4. A method of treating a mammal with cancer selected from the group consisting of breast cancer, colon cancer, central nervous system cancer, leukemia, melanoma, lung cancer, ovarian cancer, prostate cancer, and renal cancer, comprising: administering a therapeutically effective amount of the compound of claim 1 to the mammal under conditions effective to treat the cancer.
5. The method of claim 4, wherein the mammal is human.
6. A pharmaceutical composition of matter comprising the compound of claim 1 and one or more pharmaceutical excipients.
7. A method of treating a mammal with cancer selected from the group consisting of breast cancer, colon cancer, central nervous system cancer, leukemia, melanoma, lung cancer, ovarian cancer, prostate cancer, and renal cancer, comprising: administering a therapeutically effective amount of the compound of claim 3 to the mammal under conditions effective to treat the cancer.
8. The method of claim 7, wherein the mammal is human.
9. A pharmaceutical composition of matter comprising the compound of claim 3 and one or more pharmaceutical excipients.
10. A process for preparation of a purine derivative compound of the formula: Formula I wherein: R, are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain; C3C7cycloalkyl ; CH2(C3C7cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2 ; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of R1, OR1, SR1, S (O) RI, S (02) R1, NHR1, NO2, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHR), phenyl, and C (O) NHCHRlCH2OH ; 1naphthyl ; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl; 4pyridyl; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl; thiophene3yl ; 2furanyl ; 3furanyl; oxazol2yl; oxazol4yl; oxazol5yl; thiazol2yl; thiazol4yl; thiazol5yl; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl ; isoxazol3yl; isoxazol4yl; isoxazol5yl; isothiazol3yl; isothiazol4yl; isothiazol5yl; 1, 3,4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl; 2pyrrolyl ; 3pyrrolyl; 1, 3, 5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl; 2quinolinyl; 3quinolinyl; 4quinolinyl; 1isoquinolinyl; 3isoquinolinyl; and 4isoquinolinyl; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Rl, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; and (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; ClC4straight chain alkyl ; or C3C4branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 58membered saturated or unsaturated ring; n1= 03; n= 03 ; A= CH2 ; (CH2) 2 ; (CH2) 3; OCH2CH2 ; or CHCH3; Y= H; OR1; N(R1)2; N(R1)C(O)R3; N (Ri) C (O) R5; N (Rl) C (O) CH (R6) NH2; N(R1)SO2R3; N (Rl) C (O) NHR3; or N (Rl) C (O) OR6 ; R5= C3C7cycloalkyl ; R6= ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; or (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof; said process comprising: reacting a first intermediate compound of the formula: Formula IX where Z= Br or I ; with a compound of the formula: (R2B (OH) 2) or (R2Sn (nBu) 3 or R2SnMe3), or mixtures thereof, under conditions effective to form the purine derivative compound.
11. A process according to claim 10, wherein if Y is NHRI, said process further comprises: reacting the purine derivative compound with R3C (O) C1 or R5C (O) CI or R3S02CI or R3NCO or RbOC (O) C1 under conditions effective to form a final product having the same formula as the purine derivative compound except that Y is NRlC (O) R3 or NR1C(O)R5 or NR1SO2R3 or NR1C(O) NHR3 or NRlC (O) OR6.
12. A process according to claim 10 further comprising: reacting a second intermediate compound of the formula: Formula VII with a second compound of the formula: Formula VIII wherein: Vl= NH2; OH; SH; under conditions effective to form the first intermediate compound.
13. A process according to claim 12 further comprising: reacting a third intermediate compound of the formula: Formula VI with a compound of the formula: RlZ under conditions effective to form the second intermediate compound.
14. A process according to claim 10, wherein the purine derivative compound has the formula: Formula III.
15. A process for preparation of a purine derivative compound of the formula: Formula I wherein: R, are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain; C3C7cycloalkyl ; CH2(C3C7cycloalkyl) ; CH2CF3 ; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2 ; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Rl, OR1, SRi, S (O) RI, S (02) Rl, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHR,, phenyl, and C (O) NHCHRlCH2OH ; 1naphthyl; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl ; 4pyridyl ; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl; thiophene3yl; 2furanyl; 3furanyl; oxazol2yl; oxazol4yl; oxazol5yl; thiazol2yl; thiazol4yl; thiazol5yl; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl; isoxazol3yl; isoxazol4yl; isoxazol5yl ; isothiazol3yl; isothiazol4yl; isothiazol5yl; 1, 3,4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl; 2pyrrolyl; 3pyrrolyl; 1, 3,5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl ; 2quinolinyl; 3quinolinyl; 4quinolinyl; 1isoquinolinyl ; 3isoquinolinyl; and 4isoquinolinyl; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; and (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; ClC4straight chain alkyl ; or C3C4branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 58membered saturated or unsaturated ring; nl= 03; n= 03; A= CH2; (CH2) 2 ; (CH2) 3 ; OCH2CH2; or CHCH3; Y= H; OR1; N(R1)2; N(R1)C(O)R3; N (R1) C (O) Rs ; N (Rl) C (O) CH (R6) NH2; N (RI) SO2R3 ; N (Rl) C (O) NHR3; or N (RI) C (O) OR6; R5= C3C7cycloalkyl ; R6= ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; or (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof; said process comprising: reacting a first intermediate compound of the formula: Formula XV under reductive or hydrogenation conditions effective to form the purine derivative compound.
16. A process according to claim 15 further comprising: reacting a second intermediate compound of the formula: Formula XIV with a second compound of the formula: Formula XII under conditions effective to form the first intermediate compound.
17. A process according to claim 16 further comprising: reacting a third intermediate compound of the formula: Formula VII with a compound of the formula: (R2B (OH) 2) or (R2Sn (nBu) 3 or R2SnMe3), or mixtures thereof, under conditions effective to form the second intermediate compound.
18. A process according to claim 15 further comprising: reacting a fourth intermediate compound of the formula: Formula XIII with a compound of the formula: (R2B (OH) 2) or (R2Sn (nBu) 3 or R2SnMe3), or mixtures thereof, under conditions effective to form the first intermediate compound.
19. A process according to claim 18 further comprising: reacting a fifth intermediate compound of the formula: Formula VII with a compound of the formula: Formula XII under conditions effective to form the fourth intermediate compound.
20. A process according to claim 15, wherein the purine derivative compound has the formula: Formula III.
21. A process for preparation of a purine derivative compound of the formula: Formula I wherein: R, are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain; C3C7cycloalkyl ; CH2 (C3C7cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of RI, OR1, SR1, S (O) Rl, S (02) R1, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) R1, C (O) NHR,, phenyl, and C (O) NHCHRlCH20H ; 1naphthyl ; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl; 4pyridyl; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl; thiophene3yl; 2furanyl; 3furanyl ; oxazol2yl; oxazol4yl; oxazol5yl ; thiazol2yl; thiazol4yl; thiazol5yl; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl; isoxazol3yl; isoxazol4yl; isoxazol5yl ; isothiazol3yl; isothiazol4yl; isothiazol5yl; 1, 3,4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl; 2pyrrolyl; 3pyrrolyl; 1, 3,5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl; 2quinolinyl; 3quinolinyl; 4quinolinyl; 1isoquinolinyl ; 3isoquinolinyl; and 4isoquinolinyl; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; and (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; C,C4straight chain alkyl ; or C3C4branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 58membered saturated or unsaturated ring; nl= 03; n= 03 ; A= CH2 ; (CH2) 2 ; (CH2) 3 ; OCH2CH2; or CHCH3; Y= H; OR, ; N (RI) 2 ; N (R1) C (O) R3; N (Rl) C (O) R5 ; N (R1) C (O) CH (R6) NH2; N (RI) SO2R3 ; N (Rl) C (O) NHR3; or N (Rl) C (O) OR6; R^= C3C7cycloalkyl ; R6= CC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; or (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof; said process comprising: reacting a first intermediate compound of the formula: Formula XIV with a compound of the formula : Formula VIII where ViNH2 ; OH; or SH; under conditions effective to form the purine derivative compound.
22. A process according to claim 21, wherein if Y is NHRI, said process further comprises: reacting the purine derivative compound with R3C (O) CI or R5C (O) CI or R3SO2CI or R3NCO or R60C (O) CI under conditions effective to form a final product having the same formula as the purine derivative compound except that Y is NRLC (O) R3 or NR1C (O) Rs or NRIS02R3 or NRIC (O) NHR3 or NRIC (O) OR6.
23. A process according to claim 21 further comprising: reacting a second intermediate compound of the formula: Formula XVIII with a compound of the formula R IZ where Z= Br or I, under conditions effective to form the first intermediate compound.
24. A process according to claim 23 further comprising: reacting a third intermediate compound of the formula: Formula XVII with a compound of the formula 2,6dichloropurine (Formula IV) under conditions effective to form the second intermediate compound.
25. A process according to claim 24 further comprising: reacting a fourth intermediate compound of the formula: Formula V with a compound of the formula: (R2B (OH) 2) or (R2Sn (nBu) 3 or R2SnMe3), or mixtures thereof, under conditions effective to form the third intermediate compound.
26. A process according to claim 21, wherein the purine derivative compound has the formula: Formula III.
27. A process for preparation of a purine derivative compound of the formula: Formula XX wherein: Rl are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain; C3C7cycloalkyl ; CH2 (C3C7cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Rl, OR1, SR1, S (O) R,, S (O2) RI, NHR,, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Ri, C (O) NHRI, phenyl, and C (O) NHCHRICH20H ; 1naphthyl; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl; 4pyridyl; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl ; thiophene3yl ; 2furanyl; 3furanyl; oxazol2yl; oxazol4yl; oxazol5yl ; thiazol2yl; thiazol4yl; thiazol5yl; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl; isoxazol3yl; isoxazol4yl; isoxazol5yl; isothiazol3yl ; isothiazol4yl; isothiazol5yl ; 1, 3, 4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl; 2pyrrolyl; 3pyrrolyl; 1, 3, 5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl; 2quinolinyl; 3quinolinyl; 4quinolinyl; 1isoquinolinyl ; 3isoquinolinyl; and 4isoquinolinyl; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H ; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; and (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H ; C,C4straight chain alkyl ; or C3C4branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 58membered saturated or unsaturated ring; nl= 03; n= 03 ; A= CH2 ; (CH2) 2 ; (CH2) 3 ; OCH2CH2 ; or CHCH3; Y= NRIC (O) R3; NRISO2R3 ; NRIC (O) NHR3; NRIC (O) OR6; or NRI C (O) R5 ; R5= C3C7cycloalkyl ; R6= ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh; or (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof ; said process comprising : reacting a first intermediate compound having the same formula as the purine derivative compound except that Y=NHRl, with R3COC1 or R5COC1 or R3S02C1 or R3NCO or R60C (O) Cl under conditions effective to form the purine derivative compound.
28. A process according to claim 27 further comprising: reacting a second intermediate compound having the same formula as the first intermediate compound except that Y is NH2, with R8CH2Z or R8CHO under conditions effective to form the first intermediate compound where Y=NHR or N (R1) 2, wherein Z is Br or I, wherein R8 is ClC5straight chain alkyl, 205straight chain alkenyl, C3C5branched alkyl chain, C3C5branched alkenyl chain, (C3C7 cycloalkyl), CF3, or CH2CF3.
29. A process for preparation of a purine derivative compound of the formula: Formula XIX wherein: Rl are the same or different and independently selected from the group consisting of : H; ClC6straight chain alkyl ; C2C6straight alkenyl chain; C3C6branched alkyl chain; C3C6branched alkenyl chain; C3C7cycloalkyl ; CH2(C3C7cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2 ; R2= phenyl; substituted phenyl, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Ri, OR,, SRI, S (O) RI, S (02) Rs, NHR,, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHRI, phenyl, and C (O) NHCHRICH2OH ; 1naphthyl ; 2naphthyl; heterocycles selected from the group consisting of : 2pyridyl; 3pyridyl; 4pyridyl; 2pyrimidyl; 4pyrimidyl; 5pyrimidyl; thiophene2yl; thiophene3yl; 2furanyl; 3furanyl; oxazol2yl; oxazol4yl; oxazol5yl ; thiazol2yl; thiazol4yl; thiazol5yl; imidazol2yl; imidazol4yl; pyrazol3yl; pyrazol4yl; isoxazol3yl; isoxazol4yl; isoxazol5yl; isothiazol3yl; isothiazol4yl; isothiazol5yl; 1, 3,4thiadiazol2yl ; benzo [b] furan2yl; benzo [b] thiophene2yl; 2pyrrolyl; 3pyrrolyl; 1, 3,5triazin2yl ; pyrazin2yl; pyridazin3yl; pyridazin4yl; 2quinolinyl; 3quinolinyl; 4quinolinyl; 1isoquinolinyl ; 3isoquinolinyl; and 4isoquinolinyl; or substituted heterocycle, wherein the substituents (12 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; and (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; ClC4straight chain alkyl ; or C3C4branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 58membered saturated or unsaturated ring; nl= 03; n= 03; A= CH2; <BR> <BR> (CH2)2;<BR> (CH2)3; OCH2CH2; or CHCH3; Y= NHC (O) CH (R6) NH2 R5= C3C7cycloalkyl ; R6= ClC4straight chain alkyl ; C3C4branched chain alkyl ; C2C4alkenyl chain; (CH2) nPh ; or (CH2) nsubstituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4thiadiazole; or a pharmaceutically acceptable salt thereof ; said process comprising : reacting a first intermediate compound having the same formula as the purine derivative compound except that Y is NH2, with a compound of the formula: PNHCH (R6) C02H under conditions effective to form the purine derivative compound after a suitable deprotection strategy, wherein P= C (O) OtBu; C (O) OCH2Ph; Fmoc; Benzyl; or Alloc.
Description:
HETEROCYCLE SUBSTITUTED PURINE DERIVATIVES AS POTENT ANTIPROLIFERATIVE AGENTS This application claims the benefit of U. S. Provisional Patent Application Serial No. 60/318,569, filed on September 11, 2001.

FIELD OF THE INVENTION The present invention relates to compounds that are shown to be potent cyclin/cyclin dependent kinase (cdk) inhibitors. Compounds with these properties are shown to be potent inhibitors of cell growth and proliferation. Such compounds can be used to treat the following conditions: rheumatoid arthritis, lupus, type 1 diabetes, multiple sclerosis, cancer, restenosis, gout and other proliferative diseases mediated by elevated levels of cell proliferation compared to a healthy mammal. Compounds of the present invention which are biaryl substituted purine derivatives are shown to be potent antiproliferative agents against a number of human transformed cell lines, and also inhibitors of human cyclin/cdk kinase complexes.

BACKGROUND OF THE INVENTION Cellular Proliferation and Cancer.

The disruption of external or internal regulation of cellular growth can lead to uncontrolled proliferation and in cancer, tumor formation. This loss of control can occur at many levels and, indeed, does occur at multiple levels in most tumors.

Further, although tumor cells can no longer control their own proliferation, they still must use the same basic cellular machinery employed by normal cells to drive their growth and replication.

Cyclin Dependent Kinases and Cell Cycle Regulation.

Progression of the normal cell cycle from the Gl to S phase, and from the G2 phase to M phase is dependent on cdks (Sherr, C. J. , Science 274: 1672-1677 (1996) ). Like other kinases, cdks regulate molecular events in the cell by facilitating the transfer of the terminal phosphate of adenosine triphosphate (ATP) to a substrate protein. Isolated cdks require association with a second subunit, called cyclins (Desai

et al. , Mol. Cell. Biol., 15: 345-350 (1995) ). Cyclins cause conformational changes at the cdk active site, allowing ATP access and interaction with the substrate protein.

The balance between its rates of synthesis and degradation controls the level of each cyclin at any point in the cycle (Elledge, S. J. , et al. , Biochim. Biophys. Acta, 1377: M61-M70 (1998) ). The influences of cyclin/cdk activity on the cell cycle and cellular transformation are summarized in Table 1.

Abnormal Cyclin/cdk Activity in Cancer.

In a normal cell, interlocking pathways respond to the cell's external environment and internal checkpoints monitor conditions within the cell to control the activity of cyclin/cdk complexes. A reasonable hypothesis is that the disruption of normal control of cyclin/cdk activity may result in uncontrolled proliferation. This hypothesis appears to hold in a number of tumor types in which cyclins are expressed at elevated levels (Table 1). Mutations in the genes encoding negative regulators (proteins) of cyclin/cdk activity are also found in tumors (Larsen, C. -J., Prog. Cell Cycle Res., 3: 109-124 (1997)) ; (Kamb, A. , Trends in Genetics, 11: 136-140 (1995) ).

Members of the Cip family of cdk inhibitors form a ternary complex with the cyclin/cdk and require binding to cyclinA, cyclinE, or cyclinD (Hall, M. , et al., Oncogene, 11: 1581-1588 (1995) ). In contrast, Ink family members form a binary complex with cdk4 or cdk6 and prevent binding to cyclinD (Parry, D.; et al. , EMBO J., 14: 503-511 (1995) ).

Table 1. Associations Among Cyclins and Cancers Cyclin Cell Cycle Role Associated cdk Cancer A S, G2 to M cdkl, cdk2 hepatocellular carcinoma (Wang, J. ; et al., Oncogene, 8:1653-1656 (1992)) B1/B2 G2 to M cdk1 none yet defined parathyroid adenoma (Motokura, T. , et al., Nature, 350: 512-515 (1991)) centrocytic B cell lymphom (Withers, D. A. , et al. , Mol. Cell. Biol. , 11: 4846- 4853 (1991)) esophageal carcinoma (Jiang, W. , et al., D1 G1 cdk4, cdk6 Cancer Res., 52: 2980-2983 (1992)) breast cancer (Dickson, C. , et al., Cancer Lett. , 90: 43-50 (1995) ) squamous cell carcinoma (Bartkova, J., et al., Cancer Res. , 55: 949-956 (1995)) hepatocellular carcinoma (Nishida, N., et al., Cancer Res. , 54: 3107-3110 (1994)) D2 Gl cdk4, cdk6 colorectal carcinoma (Leach, F. S. , et al., Cancer Res., 53: 1986-1989 (1993)) breast cancer (Keytomarsi, K. , et al., Cancer Res., 54: 380-385 (1994) ) E rit to S cdk2 gastric carcinoma (Akama, Y.; et al., Jap. J. Cancer Res. , 86: 617-621 (1995)) colorectal carcinoma (Kitihara, K. ; et al., Int. J. Cancer 62: 25-28 (1995))

Inhibitors of Cyclin/cdk Complexes as Potential Anticancer Agents.

Tumors with elevated cyclin/cdk activity, whether from the over expression of cyclins or the loss of an endogenous cdk inhibitor, are prime targets for potential therapies based on small molecule cyclin/cdk inhibitors. In fact, several small molecule inhibitors of cyclin/cdks are reported (Meijer, L. , et al. ,"Progress in Cell Cycle Research, "Plenum Press: New York, 351-363 (1995) ) and appear to bind at the ATP site of the kinase. Some information is known about small molecule inhibitors of other kinases, such as PKC (serine kinase) (Murray, K. J. et al.,"Ann.

Rep. Med. Chem. ,"J. Bristol, Ed. , Academic Press, Inc.: New York, Chapter 26 (1994) ) and tyrosine kinases (Fantl, W. J. , et al. , Ann. Rev. Biochem., 62: 453 (1993); Burke, T. R., Drugs of the Future, 17: 119-1131 (1992); Dobrusin, E. M. et al.,"Ann.

Rep. Med. Chem, "J. Bristol, Ed. , Academic Press, Inc.: New York, Chapter 18 (1992); Spence, P. , Curr. Opin. Ther. Patents, 3: 3 (1993)). A number of known inhibitors were obtained from commercial sources or were synthesized by literature procedures.

Purine Compounds as Cyclin/cdk Inhibitors.

There are several reports of 2, 6-diamino substituted purine derivatives as cyclin/cdk inhibitors and as inhibitors of cellular proliferation. Among those are reports by U. S. Patent No. 5,583, 137 to Coe, et al. , olomoucine (Vesely, J. , et al. , Eur.

J. Biochem. , 224: 771-786 (1994) ), roscovitine (Meijer, L. , Eur. J. Biochem., 243: 527- 536 (1997) ), WO 97/16452 to Zimmerman, Imbach, P. , et al., Bioorg. Med. Chem.

Lett., 9: 91-96 (1999), Norman, T. C. , et al. , J. Amer. Chem. Soc. , 118: 7430-7431 (1996), Gray, N. S. , et al. , Tetrahedron Lett., 38: 1161-1164 (1997), Gray, N. S. , et al., Science, 281: 533-538 (1998), WO 98/05335 to Lum, et al. , Schow, S. R. , et al., Bioorg. Med. Chem. Lett, 7: 2697-2702 (1997), US Patent No. , 5,886, 702 to Mackman, et al. , Nugiel, D. A. , et al. , J. Ore. Chem., 62: 201-203 (1997), and Fiorini, M. T. et al., Tetrahedron Lett, 39: 1827-1830 (1998). Many of these reported compounds are shown to inhibit cyclin/cdk complexes and have modest cellular proliferation inhibition properties.

The compounds of the present invention are shown to have far superior biological activities as cyclin/cdk complex inhibitors as well as inhibitors of cellular proliferation compared to those previously reported. In fact, the art (e. g. , Fiorini, M. T. et al., Tetrahedron Lett, 39: 1827-1830 (1998) ) teaches away from compounds of this invention, claiming lack of cellular proliferation inhibition.

SUMMARY OF THE INVENTION The compounds of the present invention are 2,6, 9-trisubstituted purine derivatives which are inhibitors of cyclin/cdk complexes. The compounds of the current invention also are potent inhibitors of human cellular proliferation. As such, the compounds of the present invention constitute pharmaceutical compositions with a pharmaceutically acceptable carrier. Such compounds are useful in inhibiting cellular

proliferation in a mammal by administering to such mammal an effective amount of the compound.

In one embodiment, the compounds of the present invention are represented by the chemical structure found in Formula I Formula I wherein: Rl are the same or different and independently selected from the group consisting of : H; C,-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH ; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Ri, ORl, SR1, S (O) RI, S (O2) RI, NHR,, NO2, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHR1, phenyl, and C (O) NHCHR1CH2OH ; 1-naphthyl ; 2-naphthyl;

heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl ; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl ; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl; 1, 3, 4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, R,, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H;

C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; C1-C4-straight chain alkyl ; or C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; nl= 0-3; n= 0-3 ; A= CH2; (CH2) 2; (CH2) 3 ; OCH2CH2 ; or CHCH3 ; Y= H; <BR> <BR> <BR> <BR> <BR> <BR> <BR> OR1;<BR> N(R1)2; N(R1)C(O)R3; N (R,) C (O) R5 ; N (R1) C (O) CH (R6) NH2 ; N (R1) SO2R3 ; N (R1) C (O) NHR3; or N (R1) C (O) OR6; R5= C3-C7-cycloalkyl ; R6= C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2)n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole ;

pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof.

Another aspect of the present invention is directed to a compound of the following formula: Formula III wherein: Ri are the same or different and independently selected from the group consisting of : H; Cl-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2- (C3-C7-cycloalkyl) ; CH2CF3 ; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Rl,

ORI) SRI, S (O) RI, S (02) R1,NHR1,NO2, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) R,, C (O) NHRI, phenyl, and C (O) NHCHR, CH20H ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl ; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl ; 3-furanyl ; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl ; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl ; 3-quinolinyl ; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or

substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; n= 0-3; A= CH2 ; <BR> <BR> (CH2) 2 ;<BR> <BR> <BR> <BR> (CH2) 3 ; OCH2CH2 ; or CHCH3 ; Y= H; OR, ; N (Rl) 2; N (Rl) C (O) R3; N (RI) C (O) R5 ; N (RI) C (O) CH (R6) NH2; N (RI) SO2R3 ; N (R1) C (O) NHR3; or N (RI) C (O) OR6 ; R3 are the same or different and independently selected from the group consisting of : H ; C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R5= C3-C7-cycloalkyl ; R6= Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole;

isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof.

The present invention is also directed to a process for preparation of a purine derivative compound of the formula: Formula I wherein: Rl are the same or different and independently selected from the group consisting of : H; Cl-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Rl, OR,, SR1, S (O) RI, S (02) RI, NHRI, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHR,, phenyl, and C (O) NHCHR1CH2OH ; 1-naphthyl ;

2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3, 5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Rl, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of :

H; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; C,-C4-straight chain alkyl ; or C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; ni= 0-3; n= 0-3 ; A= CH2 ; (CH2) 2; (CH2) 3 ; OCH2CH2 ; or CHCH3; Y= H; OR1; N(R1)2; N(R1)C(O)R3; N (R1) C (O) R5 ; N (Rl) C (O) CH (R6) NH2; N (RI) SO2R3 ; N (RI) C (O) NHR3; or N (R1) C (O) OR6 ; R5= C3-C7-cycloalkyl ; R6= C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole;

thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof, said process comprising: reacting a first intermediate compound of the formula:

Formula IX where Z = Br or I ; with a compound of the formula: (R2-B (OH) 2) or (R2-Sn (n-Bu) 3 or R2-SnMe3), or mixtures thereof, under conditions effective to form the purine derivative compound.

Another aspect of the present invention is directed to a process for preparation of a purine derivative compound of the formula:

Formula I wherein: Ri are the same or different and independently selected from the group consisting of : H; Cl-C6-straight chain alkyl ;

C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2- (C3-C7-cycloalkyl) ; CH2CF3 ; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Ri, OR,, SR1, S (O) Rl, S (02) R1, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Rl, C (O) NHR1, phenyl, and C (O) NHCHRlCH2OH ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl;

1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl ; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H ; C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; C,-C4-straight chain alkyl ; or C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; n, = 0-3; n= 0-3; A= CH2; <BR> <BR> (CH2) 2 ;<BR> <BR> <BR> <BR> (CH2) 3 ; OCH2CH2; or CHCH3; Y= H ; OR, ; N (Ri) 2 ;

N (R,) C (O) R3; N (RI) C (O) R5 ; N (RI) C (O) CH (R6) NH2 ; N (R,) SO2R3 ; N (R1) C (O) NHR3 ; or N (RI) C (O) OR6; R5= C3-C7-cycloalkyl ; R6= C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof, said process comprising: reacting a compound of the formula: Formula XV under reductive or hydrogenation conditions effective to form the purine derivative compound.

Another aspect of the present invention is directed to a process for preparation of a purine derivative compound of the formula:

Formula I wherein: R1 are the same or different and independently selected from the group consisting of : H; Cl-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Rl, OR,, SRI, S (O) RI, S (02) RI, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHR,, phenyl, and C (O) NHCHRlCH20H ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl;

2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl ; thiazol-4-yl ; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl ; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl; 1,3, 4-thiadiazol-2-yl; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; Cl-C4-straight chain alkyl ; or

C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; nl= 0-3; n= 0-3; A= CH2; (CH2)2; (CH2)3; OCH2CH2; or CHCH3 ; Y= H; OR, ; N (RI) 2; N (RI) C (O) R3; N (RI) C (O) R5; N (Ri) C (O) CH (R6) NH2; N (R,) SO2R3 ; N (RI) C (O) NHR3; or N (RI) C (O) OR6; R5= C3-C7-cycloalkyl ; R6= Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof, said process comprising: reacting a first intermediate compound of the formula:

Formula XIV with a compound of the formula:

Formula VIII where Vl= NH2; OH; or SH; under conditions effective to form the purine derivative compound.

Another aspect of the present invention is directed to a process for preparation of a purine derivative compound of the formula: R2 R, \ nu R3 N N N Y 4V f Nu Ruz Formula XX

wherein: Rl are the same or different and independently selected from the group consisting of : H; Cl-C6-straight chain alkyl ;

C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2- (C3-C7-cycloalkyl) ; CH2CF3 ; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Rl, ORI, SRI, S (O) Rl, S (02) RI, NHR,, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Ri, C (O) NHRI, phenyl, and C (O) NHCHR1CH2OH ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl ; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl ; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl;

1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H ; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; C,-C4-straight chain alkyl ; or C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; n, = 0-3; n= 0-3 ; A= CH2; (CH2)2; (CH2)3; OCH2CH2 ; or CHCH3; Y= NRlC (O) R3; NR, C (O) Rs ; NR, SO2R3 ;

NR, C (O) NHR3; or NR, C (O) OR6 ; R5= C3-C7-cycloalkyl ; R6= C-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof ; said process comprising: reacting a first intermediate compound having the same formula as the purine derivative compound except that Y=NHR1, with R3COC1 or RsCOCI or R3SO2C1 or R3NCO or R6OC (O) C1 under conditions effective to form the purine derivative compound.

Yet another aspect of the present invention is directed to a process for preparation of a purine derivative compound of the formula : R2 I R7 As A NH R3 N N N YX V1a N Ri Formula XIX wherein: R, are the same or different and independently selected from the group consisting of : H ; C-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2- (C3-C7-cycloalkyl) ; CH2CF3 ; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Ri, OR,, SRI, S (O) RI, S (02) RI, NHRI, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Rl, C (O) NHRI, phenyl, and C (O) NHCHRlCH20H ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl ; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl ; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl ; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl;

isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl ; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; C,-C4-straight chain alkyl ; or C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; n, = 0-3; n= 0-3 ; A= CH2 ; (CH2) 2 ; (CH2) 3; OCH2CH2 ; or

CHCH3; Y= NHC (O) CH (R6) NH2 R5= C3-C7-cycloalkyl ; R6= Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof; said process comprising: reacting a first intermediate compound having the same formula as the purine derivative compound except that Y is NH2, with a compound of the formula: PNHCH (R6) C02H under conditions effective to form the purine derivative compound after a suitable deprotection strategy, wherein P= C (O) OtBu; C (O) OCH2Ph; Fmoc; Benzyl; or Alloc.

The compounds of the present invention, as described in Formula I, show significantly improved growth inhibition of human transformed cell lines and/or cyclin/cdk inhibition relative to compounds of the prior art. These compounds have been demonstrated to be potent growth inhibitors in dozens of human transformed cell lines. Olomoucine, a structurally related purine derivative, is a poor human

transformed cell growth inhibition agent with GIso values in the 20,000-100, 000 nM range over 60-transformed cell lines. By contrast, the compounds of the present invention demonstrate GIso values over 60-transformed cell lines in the <10-25,000 nM range, preferably in the <10-100 nM range over 60-transformed cell lines, and, most preferably, <10 nM across 60-human transformed cell lines. This finding is unexpected from the prior art, which specifically teaches that compounds of the present invention would not be potent human transformed cell line growth inhibitors.

The R2 group in Formula I imparts unexpected and significant improvement in growth inhibition in human transformed cell lines, while substitution of various groups at R3 and R4 found in Formula I impart important features that contribute to cyclin/cdk inhibition and growth inhibition of human transformed cell lines. Specifically, the combination of the R2 group and the substitutions within R3 and R4 result in compounds with superior biological activity. Compounds which are cyclin/cdk inhibitors and/or human transformed cell line growth inhibitors have utility in treating human proliferative cellular disorders.

DETAILED DESCRIPTION OF THE INVENTION The compounds of the present invention are represented by the chemical structure found in Formula I.

Formula I wherein: RI are the same or different and independently selected from the group consisting of : H;

C,-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3 ; CH2CH2CF3; and CH (CF3) 2; V= NH ; O ; S; or CH2 ; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Rl, ORI, SRI, S (O) Rl, S (02) R1, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Rl, C (O) NHRI, phenyl, and C (O) NHCHR, CH20H ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl ; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl ; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl ; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl;

isothiazol-5-yl; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl ; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; R3 are the same or different and independently selected from the group consisting of : H; C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh; and (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; C,-C4-straight chain alkyl ; or C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form with intervening atoms a 5-8-membered saturated or unsaturated ring; nui-0-3 ; n= 0-3 ; A= CH2; <BR> <BR> (CH2) 2 ;<BR> <BR> <BR> <BR> <BR> <BR> (CH2) 3 ; OCH2CH2; or CHCH3; Y= H; OR, ;

N (R1) 2; N (R,) C (O) R3; N (R1) C (O) Rs ; N (R1) C (O) CH (R6) NH2; N (R,) SO2R3 ; N (R,) C (O) NHR3; or N (RI) C (O) OR6 ; R5= C3-C7-cycloalkyl ; R6= C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2)nPh ; or (CH2)n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from the group consisting of : thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole; or a pharmaceutically acceptable salt thereof.

More preferably, the compounds of the current invention are represented by the chemical structure found in Formula III. Formula III wherein:

R, are the same or different and independently selected from the group consisting of : H; C-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3 ; and CH (CF3) 2; V= NH; O ; S; or CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of RI, ORI, SRI, S (O) Rl, S (02) Rl, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) RI, C (O) NHRI, phenyl, and C (O) NHCHRoCH2OH ; 1-naphthyl ; 2-naphthyl; heterocycles selected from the group consisting of : 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl ; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl;

isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl ; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl ; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl ; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; and 4-isoquinolinyl; or substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from the group consisting of Br, Cl, F, Ri, and C (O) CH3; n= 0-3; A= CH2; (CH2) 2 ; (CH2) 3 ; OCH2CH2 ; or CHCH3; Y= H; ORI ; N (Ri) 2 ; N (RI) C (O) R3; N (Ri) C (O) R5 ; N (Rl) C (O) CH (R6) NH2 ; N (R,) SO2R3 ; N (R1) C (O) NHR3; or N (R1) C (O) OR6 ; R6= C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; or (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R7 is a heterocycle selected from the group consisting of :

thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; and 1,3, 4-thiadiazole ; or a pharmaceutically acceptable salt thereof.

In another embodiment, the present invention is directed to a method of treating a mammal with a disorder mediated by elevated levels of cellular proliferation comprising administering a therapeutically effective amount of the compound of the present invention to the mammal under conditions effective to treat the disorder mediated by elevated levels of cell proliferation.

The compounds of the present invention can be administered orally, parenterally, for example, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes. They may be administered alone or with suitable pharmaceutical carriers, and can be in solid or liquid form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.

Based on the results obtained in the standard pharmacological test procedures described below, the compounds of the present invention are useful as antineoplastic agents. More particularly, the compounds of the present invention are useful for inhibiting the growth of neoplastic cells, causing cell death of neoplastic cells, and eradicating neoplastic cells. The compounds of the present invention are, therefore, useful for treating solid tumors, including sarcomas and carcinomas, such as astrocytomas, prostate cancer, breast cancer, small cell lung cancer, and ovarian cancer, leukemias, lymphomas, adult T-cell leukemia/lymphoma, and other neoplastic disease states.

In addition to the utilities described above, many of the compounds of the present invention are useful in the preparation of other compounds.

The active compounds of the present invention may be orally administered, for example, with an inert diluent, or with an assimilable edible carrier, or they may be enclosed in hard or soft shell capsules, or they may be compressed

into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, these active compounds may be incorporated with excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compound in these compositions may, of course, be varied and may conveniently be between about 2% to about 60% of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained. Preferred compositions according to the present invention are prepared so that an oral dosage unit contains between about 1 and 250 mg of active compound.

The tablets, capsules, and the like may also contain a binder such as gum tragacanth, acacia, corn starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose, or saccharin. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.

Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar, or both. A syrup may contain, in addition to active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye, and flavoring such as cherry or orange flavor.

These active compounds may also be administered parenterally.

Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils.

Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions.

Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.

The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e. g. , glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.

The compounds of the present invention may also be administered directly to the airways in the form of an aerosol. For use as aerosols, the compounds of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants. The materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.

General Synthetic Schemes The compounds of the present invention can be prepared by conventional methods of organic synthesis practiced by those skilled in the art. The general reaction sequences outlined below are general methods useful for preparing the compounds of the present invention and are not meant to be limiting in scope or utility.

Reaction of 2, 6-dichloropurine (Formula IV) with various amines of Formula V, many of which are commercially available or prepared by literature methods or modifications of literature methods, in the presence of a polar solvent, such as ethanol, provides purines of Formula VI (General Flowsheet I, infra).

Reaction of purines of Formula VI with alkyl halides (R,-Z) in the presence of a base such as potassium carbonate provides Nl-alkylated purines of Formula VII.

Chloride displacement of N1-alkylated purines of Formula VII with amines, thiols or alcohols of structure Formula VIII, either in neat solution or in an inert solvent such as ethanol or butanol, with or without a base such as sodium hydride as appropriate, at

an appropriate temperature provides purines of Formula IX (V=NH, O, S).

Transition metal-mediated cross-coupling reaction of purines of Formula IX with boronic acid (R2-B (OH) 2) or tin reagents (R2-Sn (n-Bu) 3 or R2-SnMe3) provides purines of Formula X (V=NH, O, S). If in Formula X (Y=NH2), then subsequent reaction of Formula X (Y=NH2) with acid chloride (R3COC1), or sulfonyl chloride (R3S02C1), or isocyanate (R3NCO), or chloroformate (C1C (O) OR6) reagents provides purines of Formula XI wherein Y=NHC (O) R3, NHS02R3, or NHC (O) NHR3, or NHC (O) OR6, respectively. On the other hand, if in Formula X, Y already is ORl or NHC (O) R3 or NHS02R3 or NHC (O) NHR3 or NHC (O) OR6, as a result of what Y started out as in Formula VIII, then this last step is unnecessary.

Reaction of purines of Formula VII, with alkenyl tin reagents of Formula XII, which are prepared by conventional methods described in the literature, in the presence of a transition metal catalyst, such as Pd (0), provides purines of Formula XIII (General Flowsheet II, infra). Subsequent reaction of purines of Formula XIII with boronic acid (R2-B (OH) 2) or tin reagents (R2-Sn (n- Bu) 3 or R2-SnMe3) in the presence of a transition metal catalyst, such as Pd (0), provides purines of Formula XV. Alternatively, by switching the order of reactions dependent on the precise reactivity of the purine of Formula VII, reaction of purines of Formula VII with boronic acid (R2-B (OH) 2) or tin reagents (R2-Sn (n-Bu) 3 or R2- SnMe3) in the presence of a transition metal catalyst, such as Pd (0), provides purines of Formula XIV. Subsequent reaction of purines of Formula XIV, with alkenyl tin reagents of Formula XII, which are prepared by conventional methods described in the literature, in the presence of a transition metal catalyst, such as Pd (0), provides purines of Formula XV. Finally reduction of the olefin within Formula XV provides purines of Formula X (V=CH2).

Definitions of the groups include: Z= Br; I ; V, = NH2; OH; SH ; R, are the same or different and independently selected from:

H; Cl-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3 ; CH (CF3) 2; V= NH; O ; S ; CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from Rl, ORl, SRI, S (O) RL S (02) Rl, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) Rl, C (O) NHR1, phenyl, C (O) NHCHRlCH20H ; 1-naphthyl ; 2-naphthyl; heterocycles including: 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl ; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl;

isothiazol-4-yl; isothiazol-5-yl; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl ; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl; 3-isoquinolinyl; 4-isoquinolinyl; substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from Br, Cl, F, Ri, C (O) CH3; R3 are the same or different and independently selected from: H; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form a 5-8-membered saturated or unsaturated ring; nl= 0-3 ; n= 0-3; A= CH2 ; (CH2) 2 ; (CH2) 3; OCH2CH2; CHCH3; Y= H; OR,;

N(R1)2; <BR> <BR> N(R1)C(O)R3;<BR> N(R1)C(O)R5; N (R,) C (O) CH (R6) NH2 ; N(R1)SO2R3; N (R1) C (O) NHR3; N (R1) C (O) OR6 ; R5= C3-C7-cycloalkyl ; R6= Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; (CH2)n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R7 is a heterocycle selected from: thiophene; furan; pyrrole; thiazole; pyrazole; imidazole; isoxazole; isothiazole; 1,3, 4-thiadiazole.

General Flowsheet I cri z I t0N) Solvent + NU ci N N K7 Formula IV NH2 N N Formula V CprN N H Formula VI | Rl-Z ; base Z Z R ? R R 7 R4 R3 A, NH Y-1 -Vi NH nui R4 R3 N Formula VIII N N 4 3 I Solvent, heat I Y-'1" V N N C1N R, R FormutaVH Formula IX RZ-B (OH) Z Formula VII or R2-Sn (n-Bu) 3 or R2-Sn (Me) 3 ; Pd (O) catalyst IR2 R3C (O) CI R2 or R7 R3SO2CI K7 Or A, NH R3NCO NH or R60C (O) CI Ra R3 I N Ra Rs N J 0 > Solvent base vV N N Ni ni Ri ri Rt R Formula X Formula XI General Flowsheet II Z Z z I z K7 I i n 7 A Ra R3 | 7 r Y\% Sn (nBu H N n. zon Formula XII R4 R3 N C Cl N N Pd (O) catalyst ; Rl Solvent, heat Formula VII Formula XIII RZ-B (OH} R2-B (O H) 2 R2-B (OH) 2 or or or R2-Sn (n-Bu) 3 R2-Sn (n-Bu) 3 or or R2-Sn (Me) 3 ; R2-Sn (Me) 3 ; Pd (O) catalyst Pd (O) catalyst Iz R2 Rl 2 lR2 Rl 7 gR4 gR3 gR7 NH yw Sn (nBu) 3 A s NH NH Y NH ,,, \ N Formula XII RUZ 1 Pd (0) catalyst ; Cl N N Solvent, heat y N N nez Ri R Formula XIV Formula XV Hydrogenate Solvent R2 I lR2 I NH R, R3 N . R, Formula X

Additional, general non-limiting syntheses of compounds of the present invention of Formula X and Formula XI are shown below in General Flowsheet III.

General Flowsheet III z Ru AU Mi2 Formula V R2-B (OH) 2 or R2-Sn (n-Bu) 3 or R2-Sn (Me) 3 ; Rl R2 Pd (O) catalyst I I K7 K7 NH NH Ruz N R-Z ; basé Formula Il I I Solvent I Cl,-N N CI N H NHZ H cr N" cr N' NH Rl Formula XIV Formula XVIII Formula XVII Formula VIII Solvent, heat R2 R3C (O) CI R2 ou R R3SO2C'R7 or JAI, NH R3NCO A,, NH or R60C (O) Cl N Solvent, base Y) YMV--N"N Y ("I i V N N Y 'l V N N R Formula X Formula XI

Reaction of various amines of Formula V, many of which are commercially available or prepared by literature methods or modifications of literature methods, with boronic acid (R2-B (OH) 2) or tin reagents (R2-Sn (n-Bu) 3) or (R2-SnMe3) in the presence of a transition metal catalyst, such as Pd (0), provides biaryl amines of Formula XVII.

Reaction of 2, 6-dichloropurine (Formula IV) with various amines of Formula XVII, in the presence of a polar solvent, such as ethanol, provides purines of Formula XVIII. Reaction of purines of Formula XVIII with alkyl halides (Rl-Z) in the presence of a base such as potassium carbonate provides Nl-alkylated purines of Formula XIV. Chloride displacement of N 1-alkylated purines of Formula XIV with

amines, thiols or alcohols of Formula VIII, either in neat solution or in an inert solvent such as ethanol or butanol, with or without a base such as sodium hydride as appropriate, at an appropriate temperature provides purines of Formula X (V=NH, O, S). If in Formula X (Y=NH2), then subsequent reaction of Formula X (Y=NH2) with acid chloride (R3COC1), or sulfonyl chloride (R3SO2C1), or isocyanate (R3NCO), or chloroformate (ClC (O) OR6) reagents provides purines of Formula XI wherein Y=NHC (O) R3, NHS02R3, or NHC (O) NHR3, or NHC (O) OR6, respectively.

On the other hand, if in Formula X, Y already is ORl or NHC (O) R3 or NHS02R3 or NHC (O) NHR3 or NHC (O) OR6, as a result of what Y started out as in Formula VIII, then this last step is unnecessary.

Definitions of the groups include: Z= Br; I ; V, = NH2; OH; SH; Ri are the same or different and independently selected from: H; C1-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3; CH (CF3) 2; V= NH; O ; S; CH2; R2= phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from R"ORl, SRl, S (O) RL S (02) Rl, NHR1, N02, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) R1, C (O) NHRI, phenyl, C (O) NHCHRICH20H ; 1-naphthyl ;

2-naphthyl; heterocycles including: 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl; 1, 3,4-thiadiazol-2-yl ; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl; 4-quinolinyl; 1-isoquinolinyl; 3-isoquinolinyl ; 4-isoquinolinyl; substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from Br, Cl, F, Ri, C (O) CH3; R3 are the same or different and independently selected from: H;

Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R4= H; C1-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form a 5-8-membered saturated or unsaturated ring; nl= 0-3; n= 0-3; A= CH2 ; (CH2) 2 ; (CH2) 3; OCH2CH2; CHCH3; Y= H; OR, ; N(R1)2; N (R1) C (O) R3; N (R1) C (O) R5 ; N (Rl) C (O) CH (R6) NH2 ; N (RI) SO2R3 ; N (R1) C (O) NHR3; N (R1) C (O) OR6; R5= C3-C7-cycloalkyl ; R6= Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; (CH2)n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R7 is a heterocycle selected from: thiophene; furan; pyrrole; thiazole;

pyrazole; imidazole; isoxazole; isothiazole; 1,3, 4-thiadiazole.

Additional, general non-limiting syntheses of compounds of the present invention of Formula XVI, Formula XVII and Formula XVIII are shown below in General Flowsheet IV.

General Flowsheet IV R2 R2 r R7 RBCH2-Z, solvent, base A, NU N or N Ra R3 N \ _ R4 R3 N \ R$CHO, reducing agent y N ? solvent N y y Rl Rl Formula X Formula XVI R3C (O) CI R6 or R5C (O) CI or PHN"Y R3SO2CI ou R3NCO 1. Coupling Agent, solvent or 2. Deprotection sequence R60C (O) CI Solvent, base R2 R2 7 R7 A A NH NH R, l R3 N R, R, Y _ : CN Y/YVN R.'R. Formula XIX Formula XX

If in Formula X (Y=NH2), then subsequent reaction of Formula X (Y=NH2) with alkyl halide (RgCH2Z), an appropriate base, and a solvent; or reaction of Formula X (Y=NH2) with aldehyde (R8CHO) in the presence of a solvent and a suitable reducing agent provides purines of Formula XVI wherein Y=NHR"or N (RI) 2. On the other hand, if in Formula X, Y already is NHRI, or N (RI) 2, as a result of what Y started out as in Formula X, then this last step is unnecessary. If in

Formula XVI (Y=NHRI), then subsequent reaction of Formula XVI (Y=NHRs) with acid chloride (R3COC1 or RsCOCl), or sulfonyl chloride (R3S02C1), or isocyanate (R3NCO), or chloroformate (ClC) (O) OR6) reagents provides purines of Formula XX wherein Y=NRlC (O) R3, or NRlC (O) R5, or NR1SO2R3, or NRlC (O) NHR3, or NRlC (O) OR6, respectively. On the other hand, if in Formula XVI, Y already is NRI C (O) R3, or NR, C (O) Rs, or NR1SO2R3, or NR1C(O) NHR3, or NRlC (O) OR6, as a result of what Y started out as in Formula XVI, then this last step is unnecessary.

If in Formula X (Y=NH2), then subsequent reaction of Formula X (Y=NH2) with acid (PNHCH (R6) C02H), in a suitable solvent in the presence of an appropriate coupling agent provides a purine derivative; which upon suitable deprotection provides purines of Formula XIX wherein Y=NHC (O) CH (R6) NH2. On the other hand, if in Formula X, Y already is NHC (O) CH (R6) NH2, as a result of what Y started out as in Formula X, then this last step is unnecessary.

Definitions of the groups include: Z= Br; I ; P= C (O) OtBu; C (O) OCH2Ph ; Fmoc; Benzyl; Alloc; Rl are the same or different and independently selected from: H; C,-C6-straight chain alkyl ; C2-C6-straight alkenyl chain; C3-C6-branched alkyl chain; C3-C6-branched alkenyl chain; C3-C7-cycloalkyl ; CH2-(C3-C7-cycloalkyl) ; CH2CF3; CH2CH2CF3; CH (CF3) 2 ; V= NH; O ; S ; CH2; R2 can be in any position on the ring and selected from: phenyl; substituted phenyl, wherein the substituents (1-2 in number) are in any position and are independently selected from Ri, ORl, Suri, S (O) RI, S (02) RI, NHR,, NO2, OC (O) CH3, NHC (O) CH3, F, Cl, Br, CF3, C (O) R1, C (O) NHRI, phenyl, C (O) NHCHRlCH2OH ; 1-naphthyl ; 2-naphthyl; heterocycles including: 2-pyridyl; 3-pyridyl; 4-pyridyl; 2-pyrimidyl; 4-pyrimidyl; 5-pyrimidyl; thiophene-2-yl ; thiophene-3-yl; 2-furanyl; 3-furanyl; oxazol-2-yl; oxazol-4-yl; oxazol-5-yl; thiazol-2-yl; thiazol-4-yl; thiazol-5-yl; imidazol-2-yl; imidazol-4-yl; pyrazol-3-yl; pyrazol-4-yl; isoxazol-3-yl; isoxazol-4-yl; isoxazol-5-yl; isothiazol-3-yl; isothiazol-4-yl; isothiazol-5-yl; 1,3, 4-thiadiazol-2-yl; benzo [b] furan-2-yl; benzo [b] thiophene-2-yl ; 2-pyrrolyl; 3-pyrrolyl; 1, 3,5-triazin-2-yl ; pyrazin-2-yl; pyridazin-3-yl; pyridazin-4-yl; 2-quinolinyl; 3-quinolinyl;

4-quinolinyl; 1-isoquinolinyl ; 3-isoquinolinyl; 4-isoquinolinyl ; substituted heterocycle, wherein the substituents (1-2 in number) are in any position and are independently selected from Br, Cl, F, RI, C (O) CH3; R3 are the same or different and independently selected from: H; C1-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain; (CH2) nPh ; (CH2) n-substituted phenyl, wherein the phenyl substituents are as defined above in R2; R4= H; Cl-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; R3 and R4 can be linked together by a carbon chain to form a 5-8-membered saturated or unsaturated ring; nl= 0-3; n= 0-3 ; A= CH2; (CH2) 2 ; (CH2) 3 ; OCH2CH2 ; CHCH3; Y= H; OR1; N(R1)2; N(R1)C(O)R3; N (R1) C (O) Rs ; N (R1) C (O) CH (R6) NH2 ; N (R1) SO2R3 ; N (R1) C (O) NHR3; N (R,) C (O) OR6; R5= C3-C7-cycloalkyl ; R6= C,-C4-straight chain alkyl ; C3-C4-branched chain alkyl ; C2-C4-alkenyl chain;

(CH2) nPh ; (CH2)n-substituted phenyl, wherein the phenyl substituents are as defined above in R2 ; R7 is a heterocycle selected from : thiophene; furan ; pyrrole; thiazole; pyrazole ; imidazole; isoxazole; isothiazole; 1,3, 4-thiadiazole.

R8= C1-C5-straight chain alkyl ; C2-C5-straight alkenyl chain; C3-C5-branched alkyl chain; C3-C5-branched alkenyl chain; (C3-C7-cycloalkyl) ; CF3 ; CH2CF3. The synthesis of compound 5 is shown below in Scheme I.

Scheme I Br ci Br NH N \ N EtOH N N I H CIN ri M NH2 2 1 K2C03 DMSO Br Br \ \ NH HOJNH2 NH > N H oil HO NJN N Cl N H 4 3 H ¢, NCH3 /O B (OH) 2 Pd (PPh3) 4 Na2C03 toluene, water H N y CH3 0 NH H HONJ N H 5 The synthesis of compound 11 is shown below in Scheme II.

Scheme II I I (COCI) 2 NH40H DMF DME CH2C12 CO2H CH2CI2 0 NH2 6 7 BH3-THF THF t I /I 1 NIi Hiinigs base N H20 I 8 9 I / K2C03, DMSO I I ! NH HN N ci NXN 10 11 /L J v NN 10 11 NH2 The syntheses of compounds 12,13 and 14 are shown below in Scheme III.

Scheme III The synthesis of compound 17 is shown below in Scheme IV.

Scheme IV nBuLi I \ N Br nBU3SnCl N Sn (n-BU) 3 15 18 16 4, Pd (PPh3) 4 toluene 1 NH NU I NH N N AN N N H 17 The synthesis of compound 17 is shown below in Scheme V.

Scheme V The synthesis of compound 25 is shown below in Scheme VI.

Scheme VI N / N Pd (PPh3) a 4 + I nu Sn (nBu) 3 toluene H o) HO H 25 An alternative synthesis of compound 25 is shown below in Scheme VII.

Scheme VII The synthesis of compound 32 is shown below in Scheme VIII.

Scheme VIII Br Sn (nBu) 3 ¢t nBuLi ¢) N (nBu) 3SnCI 31 N 1 Pd (PPh3) 4 A 4 + 31 toluene NH N N HO AN N N 32' The syntheses of compounds 33 and 34 are shown below in Scheme IX.

Scheme IX I / Pd (PPh3) a 4 + toluene N B (OH) 2 HOJ \> 33' Os04 pyr toluene HA HO oh I NU N-H HO N N 34 The syntheses of compounds 36,38, and 40 are shown below in Scheme X.

Scheme X NH2 4 NH2 Pd (PPh3) a 4 + I *HCI Na2C03 N toluene N B (OH) 2 H2O HOv N N ~ N N N 35 H 36 CF3 CF3 Pd (PPh3) 4 ¢ ; 4 DME NH /DME H20 B (OH) 2 Na2C03 HO NN HO tll, 37 N'N N H 38 Pd (PPh3) 4 NH 4 + B (OH) 2 t3 uene N B (OH) 2 toluene 39 H20 N N N H 40 The synthesis of compound 43 is shown below in Scheme XI.

Scheme XI OCH3 OCH3 i. n-BuLi . B (OCH3) 3 B (OH) 2 I iii. AcOH, EtOH 41 42 OCH3 OCH3 Pd (PPh3) 4 td 4 + 42 Na03 toluene HO . H20 NH N HO J N 43 The synthesis of compound 46 is shown below in Scheme XII.

Scheme XII O i. s-BuLi TMEDA e B (OH ii. B (OCH3) 3 45 44 iii. HCI 45 Pd (PPh3) 4 W N 4 + 45 I I Nazi03 0 toluene H20 NH N I N HO N H 2 46 The syntheses of compound 48 and 50 are shown below in Scheme XIII.

Scheme XIII 3, NO2 N02 Pd (PPh3) 4 4 + nui Na2C03 B (OH) 2 DME H20 HO 48 48 zu 0 nu Pd (nPh3) 4 4 + B (OH) 2 N N toluene HO 49 H20 H 50 50 The synthesis of compound 53 is shown below in Scheme XIV.

Scheme XIV The synthesis of compound 54 is shown below in Scheme XV.

Scheme XV The synthesis of compound 56 is shown below in Scheme XVI.

Scheme XVI Br NH O N ; EtOH NaN ry, HAN N N 55 N Sn (nBu) 3 16 Pd (PPh3) 4, toluene N NH NH N HN N 56 56 The synthesis of compound 58 is shown below in Scheme XVII.

Scheme XVII The synthesis of compound 60 is shown below in Scheme XVIII.

Scheme XVIII The syntheses of compounds 61, and 62 are shown below in Scheme XIX.

Scheme XIX The syntheses of compounds 64, and 65 are shown below in Scheme XX.

Scheme XX The syntheses of compounds 66, and 67 are shown below in Scheme XXI.

Scheme XXI Br NH WO J W N 61 + NEt3 HN N O CCH3 toluene Br z 0 HN'N 66 Ber 66 Br I DMAP 0 pyr NH O pyr NH --, 11 CHCH2 HN-NN ho ici HN Y4 0 67 The synthesis of compound 73 is shown below in Scheme XXII.

Scheme XXII W W W NH40H BH3-THF NaBH3CN THF CH30H CHO CHO NH2 NH2 68 69 70 Ii r Hiinigs base 69 + 1 H20 K2C03 DMSO NE NU N N N umso NH NH N-H C1 N H Cl N 71 72 HOo ; 1 NH2 -NH, AN N N 73 73 " 73 HO N 73 The syntheses of compounds 74,75, and 76 are shown below in Scheme XXIII. Scheme XXIII 2 nu i EtOH NH NH2 l HN N N NH2 NHZ 74 "-"74 / NHZ ) EtOH NH 72 + / N/ NH2 HN N N 75 NHZ AcCI, DMAP 4 NEt3, CH2C12 nu ru NH N HN N I N 76 HN f CH3 I I T 0 The synthesis of compound 77 is shown below in Scheme XXIV.

Scheme XXIV

The synthesis of compound 78 is shown below in Scheme XXV.

Scheme XXV i ¢3N zu NH Pd (PPh3) 4 ! 61 + 16 N N toluene HN N N 78 NH2 An alternative synthesis of compound 78, and the synthesis of compound 79 are shown below in Scheme XXVI.

Scheme XXVI

The synthesis of compound 80 is shown below in Scheme XXVII.

Scheme XXVII if NCO 43 benzene 74 +6 NH / in HN N N kl)-0 ¢ W b 80 0 80 The syntheses of compounds 86, and 87 are shown below in Scheme XXVIII.

Scheme XXVIII 0 0 ! ! zu 0 PhB (OH) 2 NH2 N Pd (PPh3) 4 81 Na2CO3 82 toluene, EtOH Ho0 NaBH4, HOAc 1, 4-dioxane N NH2 II 2 I S 1 m NH2 N 83 83 , I N> H 84 84 - K2C03 DMSO nu2 2 N N N au20 A 20 DRAP 2 NH b Y AC20 V ! y rfjr') ! 7 N H NH DMAP NH \> n-BuOH HN N N HN N N Cul'-N 85 86 NU2 HNCH3 0 87 The synthesis of compound 88 is shown below in Scheme XXIX.

Scheme XXIX The syntheses of compounds 93, and 94 are shown below in Scheme XXX.

Scheme XXX 0 0 NaBH4 (5 eq) NH2 HOAc (5 eq) N Sn (Me) g \ N 1, 4-Dioxane CI N I/ PdC12 (PPh3) 2 Ne DME 150-1600C NH2 90 1 EtOH \ \ \ N N ZON z T 1 1 N tN 4 N S N NH2 NH K2CO3 NH NH « EtOH N N DMSO N N or n-BuOH N N N N H 92 91 93 NH2 Ac20 DMAP pyr « CH2C12 N N I NH HN N b 3 HNCH3 94 o 0 The syntheses of compounds 95, and 96 are shown below in Scheme XXXI Scheme XXXI The synthesis of compound 97 is shown below in Scheme XXXII.

Scheme XXXII The syntheses of compounds 98, and 99 are shown below in Scheme XXXIII.

Scheme XXXIII The synthesis of compound 100 is shown below in Scheme XXXIV.

Scheme XXXIV The syntheses of compounds 101, and 102 are shown below in Scheme XXXV.

Scheme XXXV The syntheses of compounds 103, and 104 are shown below in Scheme XXXVI.

Scheme XXXVI The syntheses of compounds 106,107, and 108 are shown below in Scheme XXXVII.

Scheme XXXVII

The syntheses of compounds 109, and 110 are shown below in Scheme XXXVIII.

Scheme XXXVIII The syntheses of compounds 111, and 112 are shown below in Scheme XXXIX.

Scheme XXXIX The synthesis of compound 113 is shown below in Scheme XL.

Scheme XL The syntheses of compounds 114,115, 116, and 117 are shown below in Scheme XLI. Scheme XLI COsH C02H < < Pd (dbda) W 61 + PPh3 dé B (OH) 2 DME B (OH) 2 DME VJ NH NIFS \> HN N Ac2O 114 pyr, DMAP H2p 2 1, 4-dioxane MeOH TMSCI C02CH3 C02H COZCH3 i MeOH TMSCI N N N N N HN N HN N HN N 6 6-6 117 116 115 HÑCH3 NH CH3 NH2 T T O O

The synthesis of compound 118 is shown below in Scheme XLII.

Scheme XLII The syntheses of compounds 123 and 124 are shown below in Scheme XLIII.

Scheme XLIII The syntheses of compounds 131 and 132 are shown below in Scheme XLIV.

Scheme XLIV I I I 1 Water /iPr2EtN L NH K., CO.) NH NH2 K CO NH z 125 N DK4S8 N zon 126 127 NHZ MeOH S NHC I \ I/I I I \ I nu2 s NH gocp NH N Water, THF HN N N BH2 N y NaHC03 I j y NH lV NH Boc20 NH PPh3, Pd (0) N N HN N N PPh Pd (0) HN N N « HN N o) Water, DME Na2CO3 130 129 128 NHBoc NHBoc ÑH2 HCI, EtOAc VJ NH N-H AC20 NH Ac20 NH DMAP, Pyr NA N CHZCIz HN N N b A N . HIV'CH3 132 NHZ 131 . hazy

The syntheses of compounds 134 and 135 are shown below in Scheme XLV.

Scheme XLV w I w w I w w I I I I NH NH NH N N N\ HCI, N W HN N /_ v v N } PPh3, Pd (0) HN N N HN N N Water, dé Q 0 0 MR 3-134 Na2C03 129 NHBoc Ac20 DMAP, Pyr CH2CI2, NH N N J y HN N 135 HN CH3 T 0 The synthesis of compound 137 is shown below in Scheme XLVI.

Scheme XLVI

NH2 Ç "I ZON if Iodoethane NH NH 71--DMSO--"k NH 21 2CO., NU NH 2MK H ETOH C1 N N liN I N 136 137 NHZ The syntheses of compounds 139 and 140 are shown below in Scheme XLVII.

Scheme XLVII \ \ I/NH2 I/I/ \ Ac20 DMAP, Pyr /CHZCIZ " ISJYJ Iodomethane NH 2M KzC03 NH EtOH N N Nez han N b C1 N \ 139 140 NH2 HNyCH3 z 0 The synthesis of compound 142 is shown below in Scheme XLVIII.

Scheme XLVIII

C NH2 Ç /nu2 \ \ / 1 1-Iodopropane D O X b X I N \ J N CIN N N Cl* I 141 142 a b a 141 142 NH2 The synthesis of compound 144 is shown below in Scheme XLIX.

Scheme XLIX 0 f I NH2 I \ zu < NH t Iodocyclopentane DMSO NH 2M K2C03 ETOH N N nu C1N N N 143 144 NH2 The synthesis of compound 146 is shown below in Scheme L.

Scheme L \ \ Q i li Alkyl bromide DMSO NH 2M K2C03 EtOH b t b C1N 145 146 ÑH2 The synthesis of compound 148 is shown below in Scheme LI.

Scheme LI

C NH2 Ç I I 1 2-Iodobutane 2M K2C03 EtOH N N HAN N Cl* 147 148 NH2 The syntheses of compounds 149-152 are shown below in Scheme LII.

Scheme LII Arc20 DMAP, Pyr B (OH) 2 I , CHZCIZ/ Pd (0), PPh3 61 /DME 2M Na2C03 HN N N HN N N 6 6 HN Jv N N HN N N u 149 NHAc 150 NE2 Propionaldehyde Na (OAc) 3BH DCE NH"NH NH NH N N N N i i i HN N HN J\ N N 151 N152 The syntheses of compounds 153-156 are shown below in Scheme LIII.

Scheme LIII OCH3 I OCH3 H3CO H3CO Arc20 H3CO B (OH) 2 ß DMAP, Pyr fq Pd (0), PPh3 /CHZCl2/ DME 2M Na2COg NU OCH3 . - OCH3 HN aN HN aN NH2 1S3 NHAC 1S4 2 Propionaldehyde Na (OAc) 3BH DCE OCH3 OCH3 H3CO H3CO NH NH : N Z HN N RN N N ~NH 1SS ~N 1S6 The syntheses of compounds 157-159 are shown below in Scheme LIV.

Scheme LIV s \ Au20 DRAP, PUR Ac20 DMAP, Pyr Pd (0), PPh3 CH2C'2 DME 61 S 2M Na2COI NH NH B (OH) 2 N N N B (OH) Z N J \ N/i v HN N N HN N ÑH2 1S7 NHAC 1S8 Propionaldehyde Na (OAc) 3BH DCE s S NH zu N N HN N ! CN ~ NH lS9 The syntheses of compounds 160-163 are shown below in Scheme LV.

Scheme LV s Vs S S Ac20 DMAP, Pyr 3 CH2CI2 Pd (0), PPh3 I/CHZCIz/ DME zon az 3 NH B (OH) Z nez HN N N HN N N u 160 NHAC 161 nu2 Propionaldehyde Na (OAc) 3BH DCE tS tS \ S \ S nu nu NH NH HN XN HN XN NH 162 N 163 162 N 163 The syntheses of compounds 164-166 are shown below in Scheme LVI.

Scheme LVI zu Au20 DMAP Pyr I 9 Pd (0), PPh3 ta CH2C12 \i 61 (+Ò 2M Na2CO3 NH NH DME 2M NazC03 NU HN N u NHZ N Hc 165 Propionaldehyde Na (OAc) 3BH DCE ? \ NH N N v HN N 166 N The syntheses of compounds 167-168 are shown below in Scheme LVII.

Scheme LVII i i Acetaldehyde Na (OAc) 3BH DCE 75 NH NH N N N N NH NH HN AN HN1AN 66 167 N 168 Ac20 DMAP, Pyr ,, CH2CI2 / NH N N HN N N han N , NAc 267 The syntheses of compounds 169-171 are shown below in Scheme LVIII.

Scheme LVIII Butryaldehyde Na (OAc) 3BH 75 > < 4 DCE 75 NH NH N N N N NH"NH nu 169 HNN'N HNN""N 66 [169 . Ac20 170 DMAP, Pyr CH2CI2 / NH N N y HN N NAc 171 The syntheses of compounds 172-173 are shown below in Scheme LIX.

Scheme LIX Cyclopropane- carboxaldehyde Na (OAc) 3BH 75 DCE NH NH HN 1WN HN 1> N N N N N HN N N HN N IRH 172 N ANH 172 Z\NA 173 The syntheses of compounds 174-176 are shown below in Scheme LX.

Scheme LX I I i i Propionaldehyde Na (OAc) 3BH DCE 75 NH NH HN1aN HN1aN b 3 t X NE 174 174 AC20 175 DMAP, Pyr t, CH2CI2 / NH AU y HN ß XzÑAC 176 The syntheses of compounds 177-178 are shown below in Scheme LXI.

Scheme LXI

The syntheses of compounds 179-180 are shown below in Scheme LXII.

Scheme LXII The syntheses of compounds 181-182 are shown below in Scheme LXIII.

Scheme LXIII The syntheses of compounds 187-188 are shown below in Scheme LXIV.

Scheme LXIV F (P Ph3) a \ F F 81 + ( EtOH, Toluene I/BH3 THF y 2M Na2CO3 B (OH) z I I1 I N // O NH2 NH2 0"NH2 "2 183 184 1 iPr2EtN EtOH ¢)/F 3 F ¢ 1 F F F F /// NHC I N NH NH2 NH K CO NH T 0 v JL Y ETOH N N umbu N jew HN CN N 187 186 185 NHZ Ac20 DMAP, Pyr CHUCK F N NH N N N HN aN ffN N N o The syntheses of compounds 193 and 194 are shown below in Scheme LXV.

Scheme LXV OCH PPh OCH3 OCH3 34 81 + I EtOH Toluene I/BH THF 2M Na2C03 U 2MNa2C03 T---"--- ! ! 'N fN B (OH) 2 N 0A NH2 NH2 189 190 1 | iPr lEtN EtOH OC3 N-N-N NHC N N NH H2 NH K CO NH EtOH \ N N."I N C1 N C1 N < 193 192 ÑH2 193 192 191 NHZ Ac20 DMAP, Pyr CH2CI2 OCH3 ? NH N NH N N HN aN HÑ CH3 0 0 The syntheses of compounds 199-200 are shown below in Scheme LXVI.

Scheme LXVI Pd (PPh3) 4 81 + S EtOH, Toluene S BH THF 2M Na2C03 3 B (OH) 2 N N O NHZ NHZ 195 196 1 | iPr lEtN EtOH S s S NHC N N NH NHZ NH K CO NH ETCH ETOH N N z ZON HN-N ci N N c N 199 198 197 NHZ Ac20 DMAP, Pyr CH2CI2 S T NH N N HN-N"N HN CH3 o T 0 The syntheses of compounds 205-206 are shown below in Scheme LXVII.

Scheme LXVII Pd (PPh3) 4 81 + ETOH, Toluene 0 BH3THF 0 2M Na2C03 B (OH) Z w N I N / OW NH2 NH2 O-NH NH2 201 202 1 iPr2EtN EtOH 0 0 0 \ O-\-\ N N N 2 N N NH NHz ! NH2 ! Y ETOH N N N Z N BtOH,, Sd- NH Cl N C1 N H 205 204 203 NHZ Ac20 DMAP, Pyr CH2CI2 Co 0 NH NH HN N N 6 206 I I f o 0 The syntheses of compounds 207-210 are shown below in Scheme LXVIII.

Scheme LXVIII au20 II w /Ac20 I/ HZNNHZ NH NH 72 EtCH ZON Pur AcHNNN H " 207 208 Propionaldehyde Na (OAc) 3BH DCE NH NH N N N N N N H H 209 210

The syntheses of compounds 211-212 are shown below in Scheme LXIX.

Scheme LXIX The syntheses of compounds 213-215 are shown below in Scheme LXX.

Scheme LXX Ac II I ao d Ac20 t NH NH H 213 214 213 214 H H 213 214 Propionaldehyde Na (OAc) 3BH DCE nu NH z H II /N N aN H 215 The syntheses of compounds 216-217 are shown below in Scheme LXXI.

Scheme LXXI

I I HZNNH Ac20 I/ t ! J AcO LtJ Hunig's base DMAP, Pyr 1-Propanol CHZCIz nu \> N 216 217 AcHN-, U 216 217 The syntheses of compounds 218-219 are shown below in Scheme LXXII.

Scheme LXXII

The synthesis of compounds 221 is shown below in Scheme LXXIII.

Scheme LXXIII nu2 I I 1. CbzCl, NaOH Pd/C, MeOH Water, CH2CI2 NH4HC02 NIFS NE 2. 72, Propanol N N H N CbzNCs r N p HNWH N 220 221 The synthesis of compound 222 is shown below in Scheme LXXIV.

Scheme LXXIV Nu2 ka I/ NHZ I/ 72, Propanol NH HZN I' N N-1 H 222 The synthesis of compound 223 is shown below in Scheme LXXV.

Scheme LXXV I NH2 I Pd (0), PPh3 ANH2 l-propanol 2M Na2C03 NH N I----N'r o"o JJL JL un In HNIN-XN HUNCH3 223 T 0

The synthesis of compound 224 is shown below in Scheme LXXVI.

Scheme LXXVI \ Pd (0), PPh3 /l-propanol 61 2M Na2C03 N B (OH) z HN N N z NH2 224 The synthesis of compound 229 is shown below in Scheme LXXVII.

Scheme LXXVII

The syntheses of compounds 230-233 are shown below in Scheme LXXVIII.

Scheme LXXVIII F F F N N N N Boc-L-alanine, NH HATU, Hunig's base NH NH DMF HCI, EtOH N N N N HN N N HNIN N _ 230 231 NHZ HN NHBoc NH2 Boc-glycine, ° ° HATU, Hunig's base DMF F zu zon nu HCI, EtOH N han N 233 han NHBoc NHZ O O 0 0 The syntheses of compounds 239-241 are shown below in Scheme LXXIX.

Scheme LXXIX Br Br Br pur i /EtOAc NH NH ZON ci N N ci N CI N H 234 235 236 NH2 ! C EtOH NHZ Br nu nu NU N NH NH 1. Boc2O, H20, THF N N'N ITA, CH2C'2 N NaHC03 N N if HN N N RN N N 2. Phenylboronic acid Pd (PPh3) a DMA 2M Na2CO3 dz 239 NHBoc 238 Ac2O, DMAP \ Propionaldehyde CH2CI2, Pyr Na (OAc) 3BH DCE NH NH ,,, % N i,, i N HN N , HN N N HN N N HN N N 240 241 NHAc IVH The syntheses of compounds 242-243 are shown below in Scheme LXXX.

Scheme LXXX Brus I i 1. Boc20, HzO, THF NH NaHCO N NH _ NH 2. 3-Thiopheneboronic acid N N Pd (PPh3) 4 DME HN'ill /2 3 HN N N 237 NHZ ÑH2 237 kJ NHBoc 242 s 4 1 HCIv MeOH NH NH N N Han N NHC 243 The syntheses of compounds 248-250 are shown below in Scheme LXXXI.

Scheme LXXXI ber, NHBoc 1. TFA, CH, Cll THF, NaOH EtOHtPr2EtN O NH OH NHBoc NX N ! ! l-T-unxTr. u f !) 2. 1, ; Pr2EtN"Y" ! h THF, NaOH Lf p.-,',- ! un ttUH Q 244 245 > ci N 246 I KI) CO 246 O NH O NH b o Ac20, DMAP N _ ONA CH'CIz, NEt3 II > NH Z N N EtOH C NHAc Cl NH2 248 247 \ Propionaldehyde Na (OAc) 3BH DCE NY N N HN N N /NH 250 The syntheses of compounds 256 and 257 are shown below in Scheme LXXXII.

Scheme LXXXII NHZ Br Br Br Br ipr1tN Etch nu2 /-- NH NH ETOH NH : cN> HN N, N N HNN 251 C1N H C1N 252 253 254 NHZ n | Boc2O, THF Br NaHC03, H2O 1. Phenylboronic acid NH Pd (PPh3) 4 DME NH 2M Na2CO3 N \ N \ 2. HCI, MEOH HN N HN N 6 NHZ NHBoc 1. 3-Thiopheneboronic acid Pd (PPh3), DME 2M Na2C03 2. HCI, MeOH NH NU non N NH2 257 The syntheses of compounds 258-260 are shown below in Scheme LXXXIII. Scheme LXXXIII i i Propionyl Chloride NH DMAP, Pyr CH2CI2 N N 75 I I 75') t 1 > HAN N 6 258 NH O T o Methyl Chloroformate NH DMAP, Pyr CH, CI, N N 75 > HN N 259 , oy NH O 0 o J ! Methanesulfonyl Chloride NH DMAP, Pyr CH, C12 N N 75') t HN N N 260 s NH X, 0 The syntheses of compounds 261-263 are shown below in Scheme LXXXIV. Scheme LXXXIV i i Cyclopropanecarbonyl Chloride NH DMAP, Pyr CH2C12 N N 75 ru HN N N 261 NU 0 O Succinic anhydride NH Xylene N N HN N Js N HNN'N zon 0 o HJ W CF3SO2CI, Hunig's base NH CH2C12 N 75 I I HN N N 263 3 ost O NO 0 0 The syntheses of compounds 264-265 are shown below in Scheme LXXXV.

Scheme LXXXV Ci cl s s Pd (PPh3) a Toluene EtOH 61 + AS NaOH NH NEt3 NEt3 NH I N w JN N \/ B (OH)) Han N N NH2 264 NHAc 265 The synthesis of compound 266 is shown below in Scheme LXXXVI.

Scheme LXXXVI

The syntheses of compounds 271 and 272 are shown below in Scheme LXXXVII and begin with the known amine 267 (Finch, H. , et al. , J. Chem. Soc. Perkin Trans. I, 9: 1193, (1994), which is hereby incorporated by reference in its entirety).

Scheme LXXXVII

The syntheses of compounds 279 and 280 are shown below in Scheme LXXXVIII and begin with the known acid 273 (Lawesson, et al., Ark Kemi., 11: 317,322, (1957); Campaigne, et al. , J. Amer. Chem. Soc. , 76: 2445, (1954); Fournari, P. et al. , Bull.

Soc. Chim. Fr., 4115, (1967), which are hereby incorporated by reference in their entirety).

Scheme LXXXVIII The syntheses of compounds 287 and 288 are shown below in Scheme LXXXIX and begin with the known acid 281 (Gilman, et al. , J. Amer. Chem. Soc. , 55: 2903,2908 (1933) ; Johansson, G. , et al. , J. Med. Chem., 40 (23): 3804, (1997), which are hereby incorporated by reference in their entirety).

Scheme LXXXIX

The syntheses of compounds 295 and 296 are shown below in Scheme XC and begin with the known acid 289 (Gilman, et al. , J. Amer. Chem. Soc., 55: 2903, (1933); Bury, P. , et al. , Tetrahedron, 50 (29): 8793, (1994); Tanis, S. , et al., Tetrahedron Lett., 25 (40): 4451, (1984); Sornay, et al. , Bull. Soc. Chim. Fr., 990,995, 999 (1971), which are hereby incorporated by reference in their entirety).

Scheme XC The synthesis of compound 303 is shown below in Scheme XCI and begins with the known aldehyde 297 (Farnier, et al. , Bull. Soc. Chim. Fr. , 351,356 (1973), which is hereby incorporated by reference in its entirety).

Scheme XCI 1. Jones Ox. 2. BOP, HOBT OHC Hunig's base H2NOC H., N 4 CH2CI2 4 NaCNBH3 4 N 3. NH40H N N H H H 297 298 299 1 EtOH HN HN NH YNH NH NH NH NH KCOg NH nu2 3 N N N N\) J i HN N N or Cl/N N Cl N n-Bull 302 301 302 NH B (OH) 2 PdCl2 (PPh3) 2 DMF 150-160°C HN- Ph NH N,, J,, N HN N NH, 303

The synthesis of compound 310 is shown below in Scheme XCII and begins with the known acid 304 (Huettel, et al. , Justus Liebigs Ann. Chem. , 625: 55,60, (1959), which is hereby incorporated by reference in its entirety).

Scheme XCII 1. BOP, HOBT, HO2C Br Hunig's base, H2NOC Br H2N Br CH2CII NaCNBH3 N H H H H 304 305 306 1 EtOH NHy HN - ber Br N Br 1 NH NH NH NH Nui N, N _N > ETOH N : N\ DMSO N or LN Cl \N n-BuOH H 308 307 309 ÑH2 n B (OH) 2 PdCl2 (PPh3) 2 DMF 150-160°C zu Ph Nymph N fPh NH HN N N b S ÑH2 310

The syntheses of compounds 315 and 316 are shown below in Scheme XCIII and begin with the known amine 311 (Jpn. Hokai Tokkyo Koho, 2000143648,26 May 2000; Jpn. Hokai Tokkyo Koho, 05286936,02 Nov 1993; PCT Int. Appl., 2000021943,20 April 2000; Ger. Offen., 19653586,25 Jun 1998, which are hereby incorporated by reference in their entirety).

Scheme XCIII

The syntheses of compounds 321 and 322 are shown below in Scheme XCIV and begin with the known amine 317 (U. S. Patent 4, 358, 435 ; Chem. Pharm. Bull., 38 : 951, (1990), which are hereby incorporated by reference in their entirety).

Scheme XCIV

The syntheses of compounds 327 and 328 are shown below in Scheme XCV and begin with the known amide 323 (Can. J. Chem. , 55: 243 (1977), which is hereby incorporated by reference in its entirety).

Scheme XCV

EXAMPLES Proton NMR spectra were obtained on a Bruker AC 300 spectrometer at 300 MHz or a Bruker 500 MHz spectrometer and were referenced to tetramethylsilane as an internal standard. The IR spectrometer used was a single beam Perkin-Elmer Spectrum 1000 FT-IR. All IR spectra obtained were prepared in a pressed disc of KBr. All IR spectra obtained were acquired with a total of 4 accumulations at a resolution of 4.00 cm~'. Melting points were obtained on a Mel- Temp II apparatus and are uncorrected. Mass spectra were obtained on either a Shimadzu QP-5000 or a PE Sciex API 150 Mass Spectrometer.

Example 1-Preparation of Compound 2 To the starting material 1 (1.0 g, 5.29 mmol) was added 4- bromobenzylamine (2.53 g, 11.4 mmol), and EtOH (11 mL). The mixture was stirred and heated at 50 °C in a round-bottomed flask and then H20 (1 mL) and EtOH (10 mL) were added to dissolve the solids. The mixture was refluxed for 1 h. Hunig's base (3.68 mL, 21.2 mmol) was added and refluxed overnight, during which time a precipitate formed. The solution was filtered to provide a light yellow solid. The solid was dried in vacuo (1.08 g, 60%):'H NMR (300 MHz, DMSO-d6) 8 8.75 (bs, 1 H), 8. 15 (s, 1 H), 7.52 (d, 2 H), 7.30 (d, 2 H), 4.63 (bs, 2 H) ; CI MS m/z = 340 [C, 2H9BrClNs+H] +.

Example 2-Preparation of Compound 3 To the starting material 2 (1.08 g, 3.19 mmol) was added DMSO (11 mL), K2C03 (2.20 g, 15.95 mmol), and 2-iodopropane (1 mL, 9.57 mmol). The solution was stirred overnight then poured into H20 (75 mL) and stirred. Additional H2O (25-50 mL) was added to the mixture to form a yellow solid. The stirring was continued at 0 °C. The solid was filtered in vacuo. The crude product was purified by silica gel chromatography to provide 3 (0.66 g, 50%) as a white solid: mp 136-140 °C ; IH NMR (300 MHz, CDC13) 8 7.78 (s, 1 H), 7.49 (d, 2 H), 7.28 (d, 2 H), 6.12 (bs, 1 H), 4.90-4. 70 (m, 3 H), 1.61 (d, 6 H).

Example 3-Preparation of Compound 4 To starting material 3 (1.44 g, 3. 78 mmol) was added 2-amino-1- butanol (5.06 g, 56.7 mmol) and ethanol (5 mL) and the mixture was heated in a sealed tube in an oil bath at 150-160 °C for 48 h. The cooled solution was transferred to a round-bottomed flask and the ethanol was removed in vacuo. The crude product was purified by flash column chromatography on silica gel to give 4 (0.90 g, 55%): 'H NMR (300 MHz, CDCl3) 8 7.44-7. 41 (m, 3 H), 7.23 (d, 2 H), 6.22 (s, 1 H), 5.06 (s, 1 H), 4.90 (d, 1 H), 4.78-4. 68 (m, 2 H), 4.65-4. 55 (m, 1 H), 3.91-3. 80 (m, 2 H), 3.66- 3.60 (m, 1 H), 1.66-1. 47 (m, 8 H), 1.04-0. 99 (t, 3 H).

Example 4-Preparation of Compound 5 To starting material 4 (0.13 g, 0.29 mmol) was added 3- acetamidophenylboronic acid (0.21 g, 1.19 mmol) and Pd (PPh3) 4 (0.08 g, 0.07 mmol), Na2CO3 (2M, 0.60 mL), and toluene (5 mL). The solution was degassed with argon for 10 min then heated at 130 °C for 6 h. The cooled solution was diluted with water and then extracted with CH2C12 (3 x 50 mL). The combined organic phases were washed with brine, dried over anhydrous Na2S04, filtered, and concentrated to yield a viscous orange oil. The oil was purified by flash column chromatography on silica gel and then the product crystallized upon standing to give 5 (0.06 g, 41%) as a pale yellow solid :'H NMR (300 MHz, CDC13) 8 8.01-7. 21 (m, 9 H), 6.48 (s, 1 H), 4.97 (d, 1 H), 4.82-4. 70 (m, 2 H), 4.65-4. 53 (m, 1 H), 3.98-3. 25 (m, 2 H), 3.20-3. 05 (m, 1 H), 2.20 (s, 3 H), 1.69-1. 45 (m, 8 H), 1.07-0. 98 (t, 3 H).

Example 5-Preparation of Compound 7 To 4-iodobenzoic acid (52.2 g, 0.21 mol) was added CH2C12 (500 mL) and DMF (2 drops) at room temperature. Oxalyl chloride (32 g, 0.25 mol) was added dropwise in 0.5 h and stirred for 2 d. The volatiles were removed in vacuo to a volume of 150 mL to give the acid chloride and CH2C12. To a mixture of ice (500 mL) and NH40H (29%; 100 mL) was added the CH2C12 solution during 15 min. The resulting solids were collected, washed with CH2Cl2, and dried in vacuo. The solids were slurried in H20 for 1 h. The solids were collected by filtration, washed in water and acetone, and dried in vacuo to give 7 (48 g; 92%): mp 213-216 °C.

Example-Preparation of Compound 8 To a suspension of 7 (11 g, 45 mmol) in THF (50 mL) was added BH3- THF (1M, 22.5 mL, 22.5 mmol). The resulting solution was heated under reflux overnight. The reaction was cooled in an ice bath and MeOH-HCl (60 mL) was slowly added dropwise. The resulting precipitate was filtered and dried to give 8 (10.8 g, 88%) as a white solid: mp 256-262 °C dec. ; 1H NMR (300 MHz, DMSO-d6) 8 8.55 (bs, 3 H), 7.79 (d, 2 H), 7.32 (d, 2 H), 3.98 (s, 2 H).

Example 7-Preparation of Compound 9 To compound 1 (7.63 g, 40.4 mmol) was added compound 8 (10.8 g, 40.4 mmol), water (123 mL), and Hunig's base (14 mL, 81 mmol). The mixture was heated to reflux for 5 h and stirred overnight at room temperature to give a pale yellow solution. An additional quantity of water (150 mL) was added, refluxed for 3 h, then cooled overnight. A pale yellow solid was formed which was filtered, washed with water, rinsed with EtOH (2 x), and dried in vacuo to give yield 9 (13.3 g, 80%): lH NMR (300 MHz, DMSO-d6) 8 8.68 (bs, 1 H), 8.28 (s, 1 H), 7.68 (d, 2 H), 7.50 (d, 2 H), 5.08 (bs, 1 H), 4.50 (d, 2 H).

Example 8-Preparation of Compound 10 To compound 9 (12.2 g, 31.7 mmol) was added K2CO3 (35 g, 0.25 mol), 2-iodopropane (13 g, 0.13 mol) and DMSO (210 mL). The reaction mixture was stirred under N2 at room temperature overnight, then poured into H2O (1.5 L) and stirred for 2 d. The precipitate was collected as an off-white solid and washed with Et2O. The aqueous layer was extracted with EtOAc (2 x) and the combined organic phases were washed with brine, dried over Na2S04, filtered, and evaporated to give an off-white foam (6.4 g). This off-white foam was combined with the precipitate and washed with Et20 to give 10 (11.0 g) :'H NMR (300 MHz, DMSO-d6) 8 8.91 (m, 1 H), 8.38 (s, 1 H), 7.74 (d, 2 H), 7.21 (d, 2 H), 5.11 (bs, 1 H), 4.68 (m, 1 H), 4.60 (d, 2 H), 1.48 (d, 6 H).

Example 9-Preparation of Compound 11 Compound 10 (1.52 g, 3.55 mmol), trans-1, 4-diaminocyclohexane (6.35 g, 55.60 mmol), and EtOH (18 mL) were placed in a sealed tube. The reaction mixture was heated at 120-190 °C for 24 h. The reaction was then allowed to cool to room temperature. The reaction mixture was filtered and the filtrate evaporated. The residue was purified by column chromatography, and dried in vacuo for 16 h to yield 11 (1.60 g, 89%) as a yellow sticky oil :'H NMR (300 MHz, CDCl3) 6 7.62 (d, 2 H), 7.44 (s, 1 H), 7.08 (d, 2 H), 6.14 (br, 1 H), 4.75-4. 63 (m, 2 H), 4.63-4. 54 (m, 2 H),

3.75-3. 63 (m, 1 H), 2.72-2. 57 (m, 2 H), 2.18-2. 00 (m, 2 H), 2.00-1. 75 (m, 4 H), 1.54 (d, 6 H), 1.39-1. 00 (m, 3 H) ; API MS m/z = 506 [C21H28IN7+H] +.

Example 10-Preparation of Compound 12 To compound 11 (0. 133 g, 0.26 mmol) was added DME (2.5 mL) and 3-thiopheneboronic acid (0.12 g, 0.97 mmol) in a round-bottomed flask and equipped with a condenser purged with argon. To this was added DME (3 mL) followed by tris (dibenzylidoneacetone) dipalladium (0.01 g, 0.01 mmol) and PPh3 (0.04 g, 0.15 mmol). Na2C03 (2M, 0.6 mL) and DME (1 mL) was added to the reaction mixture and the reaction mixture was allowed to reflux for 18.5 h, then stirred at room temperature under argon for 46 h. The reaction mixture was diluted with H20 and extracted with CH2Cl2. The combined organic phases were washed with brine, dried over anhydrous Na2S04, filtered, and concentrated in vacuo. The residue was purified by column chromatography to yield 12 (0.050 g, 41%) as a tan solid :'H NMR (300 MHz, CDC13) 8 7.56-7. 50 (m, 4 H), 7.44-7. 35 (m, 3 H), 6.02 (br, 1 H), 4.78 (d, 2 H), 4.69-4. 54 (m, 2 H), 3.75 (br, 1 H), 2.69 (br, 1 H), 2.15 (br, 2 H), 1.88 (br, 3 H), 1.54 (d, 7 H), 1.33-0. 97 (m, 4 H); API MS m/z= 462 [C25H3lN7S+H] +.

Example 11-Preparation of Compound 13 DME (3 mL), tris (dibenzylidoneacetone) dipalladium (0.01 g, 0.01 mmol), and PPh3 (0.04 g, 0.15 mmol) were placed in a round-bottomed flask fitted with a condenser and maintained under argon. Compound 11 (0.13 g, 0.26 mmol), and 4-methylbenzeneboronic acid (0.13 g, 0.98 mmol) dissolved in Na2C03 (2M, 0.6 mL) and DME (1 mL) were added to the reaction mixture. The reaction mixture was refluxed for 19.5 h and stirred at room temperature for 4 h. The reaction mixture was diluted with water and extracted with CH2C12. The combined organic phases were washed with brine, dried over anhydrous Na2SO4, and evaporated. The crude product was purified by column chromatography and dried in vacuo for 22 h to yield the desired product 13 (54 mg, 44%) as an off-white solid :'H NMR (300 MHz, CDC13) 8 7.56-7. 41 (m, 7 H), 7.23 (s, 1 H), 5.92 (br, 1 H), 4.83 (d, 2 H), 4.74-4. 58 (m, 2 H),

3.77 (br, 1 H), 2.70 (br, 1 H), 2.40 (s, 3 H), 2.16 (d, 3 H), 1.88 (d, 3 H), 1.55 (d, 7 H), 1.33-0. 97 (m, 4 H); API MS m/z = 470 [C2sH3sN7+H] +.

Example 12-Preparation of Compound 14 DME (3 mL), tris (dibenzylideneacetone) dipalladium (0.01 g, 0.01 mmol), and PPh3 (0.04 g, 0. 15 mmol) were placed in a round-bottomed flask with a condenser under argon. Compound 11 (0.13 g, 0.25 mmol) and 3-chloro-4- fluoroboronic acid (0.15 g, 0. 88 mmol) were dissolved in Na2C03 (2M, 0.6 mL) and DME (1 mL) were added to the reaction mixture, refluxed for 19 h then stirred at room temperature for 2 h. The reaction mixture was diluted with water and extracted with CH2C12. The combined organic phases were washed with brine, dried over anhydrous Na2S04, and evaporated. The crude product was purified by repeated column chromatography to yield 14 (0.019 g, 15%) :'H NMR (300 MHz, CDC13) 5 7.59-7. 53 (m, 1 H), 7.47-7. 35 (m, 4 H), 7.26-7. 14 (m, 3 H), 5.81 (br, 1 H), 4.81 (d, 2 H), 4.72-4. 54 (m, 2 H), 3.72 (br, 1 H), 2.69 (br, 1 H), 2.21-2. 03 (m, 3 H), 1.94-1. 78 (m, 3 H), 1.54 (d, 6 H), 1.33-1. 12 (m, 4 H); API MS m/z = 508 [C27H3lClFN7+H] +.

Example 13-Preparation of Compound 16 A solution of 15 (2.5 g, 15.8 mmol) and ether was cooled to-78 °C. In a separate flask, n-BuLi (15.8 mmol) was also cooled to-78 °C. The solution of 15 was added to the n-BuLi solution via cannula to give a dark red solution. The reaction mixture was stirred for 5 min prior to the rapid addition of (n-Bu) 3SnCl (6.2 g, 19 mmol). The resulting bright yellow solution was stirred at-78 °C for 2 h, allowed to warm to room temperature, and stirred for another 10 min. The solution was then diluted with H20 (80 mL) and extracted with ethyl acetate (3 x 50 mL). The organic extracts were combined, washed with brine, dried over Na2S04, filtered, and concentrated in vacuo to yield the crude product as a yellow oil. Purification by column chromatography gave the product 16 (4.89 g, 84%) as a pale yellow liquid: 'H NMR (300 MHz, CDCI3) 8 8.72 (d, 1 H), 7.48-7. 46 (m, 1 H), 7.40-7. 38 (m, 1 H), 7.11-7. 09 (m, 1 H), 1.61-1. 50 (m, 6 H), 1.38-1. 26 (m, 6 H), 1.14-1. 09 (m, 6 H), 0.97- 0.77 (t, 9 H).

Example 14-Preparation of Compound 17 To compound 16 (0.18 g, 0.48 mmol) was added compound 4 (0.14 g, 0.33 mmol), Pd (PPh3) 4 (0.05 g, 0.49 mmol), and toluene (10 mL) in a sealed tube under an argon atmosphere. The solution was degassed with argon and heated at 135 °C in an oil bath for 3 h. The solution was cooled to room temperature, diluted with saturated NaHC03, and extracted with CH2Cl2 (3 x 30 mL). The organic layer was washed with brine, dried over Na2S04, and concentrated in vacuo to give a light brown oil. The residue was purified by flash column chromatography using MeOH/CH2Cl2 (10%) to afford 17 as a white solid. The sample was dissolved into hexane/CH2Cl2/MeOH and then precipitated with diethyl ether, filtered, and rinsed several times with ether to provide in 17 (30.3 mg): mp 95-100 °C ;'H NMR (300 MHz, CDC13) 8 8.68 (d, 1 H), 7.96 (d, 2 H), 7.77-7. 69 (m, 2 H), 7.49-7. 45 (m, 3 H), 7.24-7. 20 (m, 1 H), 5.99 (s, 1 H), 5.11 (s, 1 H), 4.88-4. 83 (m, 3 H), 4.65-4. 56 (m, 1 H), 3.91-3. 80 (m, 2 H), 3.65-3. 60 (m, 1 H), 1.66-1. 52 (m, 8 H), 1.05-0. 99 (t, 3 H); IR (KBr) 3411,2968, 1601,1489 cm~'; CI MS m/z = 432 [C24H29N7+H] +.

Example 15-Preparation of Compound 19 To a solution of n-BuLi (2. 5M hexane solution, 10.9 mL, 27.4 mmol) in ethyl ether 28 mL at-78 °C was added 2-bromopyridine (4.33 g, 27.4 mmol) in ethyl ether (15 mL). After stirring for 30 min, a solution of trimethylstannylchloride (6.0 g, 30 mmol) in THF (10 mL) was added. Stirring was continued at-78 °C for 2 h and the mixture was then warmed up to room temperature and filtered. The precipitate was washed with ether and the combined the ether filtrates were concentrated to give the crude product :'H NMR (500 Hz, CDCl3) 8 8.69-8. 68 (d, 1 H), 7.47-7. 07 (m, 3 H), 0.30 (s, 9 H).

Example 16-Preparation of Compound 21 A mixture of 4-bromobenzonitrile (1.68 g, 9.2 mmol), crude 2- trimethylstannylpyridine (3.33 g, 13.8 mmol), and PdCl2 (PPh3) 2 (321 mg, 0.46 mmol) in DMF (25 mL) was heated at 150-155 °C in pressure tube for 24 h. The DMF was

distilled off under reduced pressure and the residue was filtered through a short column of basic alumina and washed with ethyl acetate and then concentrated. Flash chromatography of the residue on silica gel gave the product (41%) as a white solid: mp 99-100 °C ;'H NMR (500 Hz, CDC13) 8 8.74 (dd, Jl = 1 Hz, J2 = 1. 7 Hz, 1 H), 8.12 (d, J = 8. 6 Hz, 2 H), 7.83-7. 76 (m, 4 H), 7.32 (m, 1 H).

Example 17-Preparation of Compound 22 To LiAlH4 (8 mmol) in THF (25 mL) was added 21 (0.96 g, 5.3 mmol) in THF (15 mL) slowly while the flask was cooled with ice. The mixture was stirred at room temperature for 10-30 min then stirred at reflux for 4 h under nitrogen. The mixture was cooled in an ice bath and aqueous sodium hydroxide solution (0.5 mL, 10%) was added. The mixture was stirred until the residue became white and the solid was filtered and washed with methylene chloride (4 x 5 mL). The methylene chloride solution was dried with anhydrous sodium sulfate, concentrated, and the crude product was chromatographed on silica gel to give the product as a yellow liquid. A small amount of ethanol was added and the pure amine 22 was obtained as a white solid (74%) after filtration: mp 114-117 °C ;'H NMR (500 Hz, CDCl3) 8 8.66 (d, J = 4. 4 Hz, 1 H), 7.94 (d, J = 8. 1 Hz, 2 H), 7.70 (m, 2 H), 7.39 (d, J=8. 0Hz), 7.19 (m, 1 H), 3.90 (s, 2 H), 1.98 (s, 2 H).

Example 18-Preparation of Compound 23 A mixture of 2, 6-dichloropurine (1,0. 19 g, 1 mmol), amine 22 (0. 39 g, 2.15 mmol) in ethanol (13 mL), and water (3.4 mL) was heated at 100-110 °C under nitrogen for 24 h and then it was cooled to room temperature. The mixture was concentrated and water (5 mL) was added. A solid was filtered and washed with water (2 x 5 mL) and dried under vacuum to give the product (93%) as yellow solid: mp 260 °C (dec) ;'H NMR (500 Hz, DMSO-d6) 8 12.4 (bs, 1 H), 8.76 (m, J = 1 Hz, 1 H), 8.28 (s, 1 H), 8.16 (d, J = 8.1 Hz, 2 H), 8.03 (d, J = 7.8 Hz, 1 H), 7.97 (m, 1 H), 7.58 (d = 8.6 Hz, 2 H), 7.45 (m, 1 H), 4.82 (s, 2 H).

Example 19-Preparation of Compound 24 To the solution of compound 23 (0.33 g, 1 mmol) in DMSO (5.2 mL), added potassium carbonate (0.7 g, 5 mmol) and 2-iodopropane (0.5 g, 3 mmol). The mixture was stirred at ambient temperature under nitrogen for 24 h and poured into ice water (30 mL). After filtration, the solid was washed with water (4 x 5 mL), dried under vacuum to give the crude product as a yellow solid. Flash column chromatography of the crude product on silica gel and recrystallization provided the pure product (76%) as white crystals: mp 178-179 °C ;'H NMR (500 Hz, CDC13) 8 8.68 (m, 1 H), 7.96 (d, J = 8 Hz, 2 H), 7.76-7. 70 (m, 2 H), 7.73 (s, 1 H), 7.47 (d, J = 8 Hz, 2 H), 7.22 (m, 1 H), 4.89 (s, 1 H), 4.79 (m, 1 H), 1.54 (d, J = 6.8 Hz, 6 H); CI MS m/z= 379 [C20Hl9ClN6+H] +. Anal. Calcd. for C20HlgClN6 : C, 63.41 ; H, 5. 05 ; N, 22.18. Found: C, 63.07 ; H, 5.01 ; N, 22.01.

Example 20-Preparation of Compound 17 To compound 24 (0.7 g, 1.8 mmol) was added (R)-(-)-2 amino-1- butanol (3.5 g, 3.9 mmol) stirred in a sealed tube for 2 h at 190 °C. The reaction mixture was allowed to cool and then was partitioned between EtOAc and brine. The EtOAc was separated, washed with saturated brine (4 x), dried with Na2S04, and concentrated. The product was air dried to give an oil, then dissolved in EtOAc. The EtOAc solution was cooled again, and the precipitate collected, washed with cold EtOAc (2 x), air dried, and heated in vacuo for 2 h to give 17 (0.54 g, 67%): mp 98- 100 °C ;'H NMR (300 MHz, CDC13) 8 8.00-7. 85 (m, 2 H), 7.75-7. 55 (m, 2 H), 7.50- 7.35 (m, 3 H), 7.30-7. 15 (m, 1 H), 6.40-6. 20 (bs, 1 H), 5.00-4. 82 (m, 1 H), 4.80-4. 68 (bs, 3 H), 4.60 (heptuplet, 1 H), 3.98-3. 70 (m, 2 H), 3.70-3. 54 (dd, 1 H), 2.10 (bs, 1 H), 1.75-1. 53 (m, 2 H), 1.51 (d, 6 H), 1.00 (t, 3 H); IR (KBr) 3406,2969, 1601,1490, 1389, 1254,779 cm~'; API MS m/z= 432 [C24H2gN7O+H] +.

Example 21-Preparation of Compound 25 To compound 4 (0.14 g, 0.33 mmol) was added 3- (tributylstannyl) pyridine (0.15 g, 0.33 mmol), Pd (PPh3) 4 (0.06 g, 0.41 mmol), and toluene (10 mL). The solution was degassed with argon for 8 min in a sealed tube,

and heated in an oil bath for 3 h at 130 °C. The cooled reaction mixture was diluted with saturated NaHC03 and extracted with CH2Cl2 (3 x 50 mL). The combined organic extracts were washed with brine and dried over Na2S04. The reaction mixture was purified by column chromatography on silica gel to give the desired coupling product. The product was dissolved in acetonitrile and washed with hexane (3 x 10 mL) to remove a portion of the tin contaminants. The reaction mixture was again purified by column chromatography on reversed phase silica gel to give compound 25 (0.04 g) :'H NMR (300 MHz, CDC13) 5 8.83 (s, 1 H), 8.58 (d, 1 H), 7.88-7. 83 (m, 1 H), 7.56-7. 46 (m, 5 H), 7.38-7. 33 (m, 1 H), 5.99 (s, 1 H), 5.11 (s, 1 H), 4.90-4. 83 (m, 2 H), 4.63-4. 56 (m, 1 H), 3.92-3. 81 (m, 2 H), 3.67-3. 60 (m, 1 H), 1.69-1. 49 (m, 8 H), 1.05-1. 00 (t, 3 H); CI MS milz = 432 [C24H29N7O+H] +.

Example 22-Preparation of Compound 27 A mixture of diethyl (3-pyridyl) borane (26,540 mg, 3.67 mmol), 4- bromobenzonitrile (803 mg, 4.41 mmol) and Pd (PPh3) 4 (144 mg, 0.13 mmol) in toluene (9 mL), ethanol (1.3 mL) and 2M aqueous sodium carbonate solution (4.1 mL, 8.2 mmol) was heated at 90-100 °C under nitrogen for 27 h. The mixture was cooled to room temperature and water (10 mL) was added. The organic layer was separated and the aqueous layer was extracted with ethyl acetate (2 x 20 mL). The combined organic layers were washed with brine (2 x 15 mL) and dried over anhydrous sodium sulfate. Flash chromatography of the crude product on silica gave the product as a white solid (80%): mp 95-96 °C.

Example 23-An Alternative Preparation of 27 is Described Below A flask charged with 4-bromobenzonitrile (360 mg, 2.0 mmol), bis (pinacolato) diboron (560 mg, 2.2 mmol), potassium acetate (590 mg, 6.0 mmol) and PdCl2 (dppf) (49 mg, 0.06 mmol) was flushed with nitrogen and DMF (12 mL) was added. The mixture was heated at 80-85 °C for 4 h and then cooled to room temperature at which time PdCl2 (dppf) (49 mg, 0.06 mmol), 3-bromopyridine (385 8L, 3.40 mmol), and 2M aqueous sodium carbonate solution (5 mL, 10 mmol) was added. The mixture was stirred at 80-85 °C for 24 h and extracted with ethyl ether (3

x 30 mL) and then washed with brine (3 x 15 mL) and dried with anhydrous sodium sulfate. Flash chromatography of the crude product on silica gel gave the product as white crystals (56%): mp 96-97 °C ;'H NMR (500 Hz, CDC13) 8 8.55 (dd, Ji = I Hz, J2 = 1.4 Hz, 1 H), 8.66 (m, 1 H), 7.90-7. 87 (m, 1 H), 7.77 (d, J = 7.8 Hz, 2 H), 7.69 (d, J = 8. 8 Hz, 2 H), 7.42 (m, 1 H).

Example 24-Preparation of Compound 28 To LiAlH4 (8 mmol) in THF (25 mL) was added 27 (0.96 g, 5.3 mmol) in THF (25 mL) slowly while the flask was cooled with ice. The mixture was stirred at room temperature for 10-30 min then stirred at reflux for 4 h under nitrogen. The mixture was cooled in an ice bath and aqueous sodium hydroxide solution (0.5 mL, 10%) was added. The mixture was stirred until the residue became white and the solid was filtered and washed with methylene chloride (4 x 5 mL). The methylene chloride solution was dried with anhydrous sodium sulfate, concentrated, and the crude product was chromatographed on silica gel to give the product as a yellow liquid. A small amount of ethanol was added and the pure amine 28 was obtained as a white solid (46%) after filtration: mp 94-96 °C ;'H NMR (500 Hz, CDC13) 8 8.74 (d, J = 2. 4 Hz, 1 H), 8.48 (dd, JI=1. 5Hz, J2=4. 7Hz, lH), 7.77 (m, 1 H), 7.45 (d, J = 8. 10 Hz, 2 H), 7.33 (d, J = 8. 0 Hz, 2 H), 7.25 (m, 1 H), 3.83 (s, 2 H), 2.25 (s, 2 H).

Example 25-Preparation of Compound 29 A mixture of 2, 6-dichloropurine (1,0. 19 g, 1 mmol), amine 28 (0.4 g, 2.15 mmol) in ethanol (13 mL), water (3 mL) was heated at 100-110 °C under nitrogen for 24 h and then it was cooled to room temperature. The mixture was concentrated and water (5 mL) was added. A solid was filtered and washed with water (2 x 5 mL) and dried under vacuum to give the product (92%) as a yellow solid: mp 219 °C (dec) ;'H NMR (500 Hz, DMSO-d6) 8 13.2 (bs, 1 H), 8.99 (s, 1 H), 8.66 (d, J = 3. 5 Hz, 1 H), 8.28 (s, 1 H), 8.16 (d, J = 7.3 Hz, 1 H), 7.80 (d, J = 7.6 Hz, 2 H), 7.60-7. 57 (m, 3 H).

Example 26-Preparation of Compound 30 To a solution of 29 (0.3 g, 1 mmol) in DMSO (5 mL), was added potassium carbonate (0.7 g, 5 mmol) and 2-iodopropane (0.5 g, 3 mmol). The mixture was stirred at ambient temperature under nitrogen for 24 h and poured into ice water (30 mL). After filtration, the solid was washed with water (4 x 5 mL), dried under vacuum to give the crude product as a yellow solid. Flash column chromatography of the crude product on silica gel and recrystallization provided the pure product (76%) as white crystals: mp 178-179 °C ; lH NMR (500 Hz, CDC13) 8 8.82 (d, J = 1.3 Hz, 1 H), 8.59-8. 58 (m, 1 H), 7.86-7. 84 (m, 1 H), 7.72 (s, 1 H), 7.56- 7.48 (m, 4 H), 7.37-7. 34 (m, 1 H), 4.88 (s, 2 H), 4.82 (m, 1 H), 1.56 (d, J = 0.7 Hz, 3 H), 1.55 (d, J = 0.8 Hz, 3 H); CI MS milz = 379 [C2oH19ClN6+H] +. Anal. Calcd. for C2oHl9ClN6 : C, 63.41 ; H, 5.05 ; N, 22.18. Found: C, 63.24 ; H, 4.97 ; N, 21.93.

Example 27-Preparation of Compound 32 To a mixture of 4 (0.05 g, 0.11 mmol) was added 4- (tributylstannyl) pyridine (0.06 g, 0.16 mmol), Pd (PPh3) 4 (0.02 g, 0.02 mmol), and toluene (2.5 mL). The reaction mixture was degassed and heated in a sealed tube at 125 °C for 3 h. The reaction mixture was cooled to room temperature then saturated NaHC03 (30 mL) was added followed by extraction with CH2Cl2 (3 x 30). The organic layer was washed with brine (50 mL), dried with MgS04, and concentrated.

The reaction mixture was purified by column chromatography on silica gel to give 32: 'H NMR (300 MHz, CDC13) 8 8.65 (s, 2 H), 7.60-7. 57 (m, 2 H), 7.49-7. 45 (m, 5 H), 6.20 (s, 1 H), 4.93 (d, 1 H), 4.84 (s, 2 H), 4.65-4. 57 (m, 1 H), 3.92-3. 80 (m, 2 H), 3.68-3. 51 (m, 1 H), 1.68-1. 58 (m, 2 H), 1.52 (d, 6 H), 1.05-0. 99 (t, 3 H).

Example 28-Preparation of Compound 33 To compound 4 (0.18 g, 0.43 mmol) was added 4-vinylphenylboronic acid (0.19 g, 1.28 mmol), Pd (PPh3) 4 (0. 09 g, 0.08 mmol), Na2C03 (2M, 0.85 mL), was added toluene (5 mL). The mixture was degassed with argon for 10 min. The resulting solution was heated in a sealed tube at 135 °C for 4.5 h. The cooled solution was diluted with water and extracted with CH2Cl2 (3 x 50 mL). The combined

organic extracts were washed with brine and dried over Na2S04. The solution was purified by flash column chromatography (2 x) on silica gel to give the desired product 33 as a yellow solid (0.09 g): mp 130-131 °C ;'H NMR (300 MHz, CDC13) S 7.57-7. 42 (m, 9 H), 6.80-6. 70 (dd, 1 H), 5.98 (s, 1 H), 5.79 (d, 1 H), 5.27 (d, 1 H), 4.88 (d, 1 H), 4.84-4. 72 (m, 2 H), 4.63-4. 56 (m, 1 H), 3.92-3. 81 (m, 2 H), 3.66-3. 60 (m, 1 H), 1.68-1. 52 (m, 8 H), 1. 05-1. 00 (t, 3 H); IR (CH2C12) 3293,2968, 1601,1489, 1390 cm-1 ; CI MS milz = 457 [C27H32N6O+H] +.

Example 29-Preparation of Compound 34 To compound 33 (0.008 g, 0.016 mmol) was added Os04 (0.007 g, 0.026 mmol), pyridine (0.08 mL), and toluene (0.75 mL). The reaction mixture was stirred at room temperature in the dark for 1 h, concentrated in vacuo, and then slurred in methanol/water (9: 1). Sodium metabisulfite (0.07 g) was added and the reaction was stirred for 1 h. The mixture was washed with brine, extracted with CH2Cl2 (3 x 10 mL), dried over Na2S04, and concentrated. The product was purified by column chromatography on silica gel to give compound 34 (0.003 g) as a tan solid: 'H NMR (300 MHz, CDCl3) 5 7.51 (s, 1 H), 7.43-7. 35 (m, 6 H), 7.25-7. 22 (m, 2 H), 6.51 (s, 1 H), 4.98 (d, 1 H), 4.35-4. 25 (m, 2 H), 4.64-4. 54 (m, 1 H), 3.93-3. 80 (m, 3 H), 3.74-3. 59 (m, 3 H), 1.68-1. 58 (m, 2 H), 1.52 (d, 6 H), 1.06-0. 99 (t, 3 H).

Example 30-Preparation of Compound 36 To compound 4 (0.12 g, 0.27 mmol) was added 3-aminophenylboronic acid hydrochloride (0.12 g, 0.69 mmol), and Pd (PPh3) 4 (0. 09 g, 0.75 mmol) in a sealed tube filled with argon. To this mixture was added toluene (5 mL) and Na2C03 (2M, 0.55 mL). The resulting solution was degassed with argon for 5 min and placed in a 130 °C oil bath for 6 h. The cooled solution was diluted with water and extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were washed with brine, dried over Na2SO4, and concentrated. The solution was purified by column chromatography on silica gel to yield 36 (0.04 g, 36%) :'H NMR (300 MHz, CDC13) 8 7.52-7. 46 (m, 3 H), 7.39 (d, 2 H), 7.23-7. 18 (m, 1 H), 6.96 (d, 1 H), 6.88 (t, 1 H), 6.68-6. 66 (m, 1 H), 6.12 (s, 1 H), 4.90 (d, 1 H), 4.79 (s, 2 H), 4.62-4. 57 (m, 1 H),

3.92-3. 76 (m, 4 H), 3.66-3. 60 (m, 1 H), 1.65-1. 48 (m, 8 H), 1.04-0. 99 (t, 3 H); CI MS m/z = 446 [C25H3iN70+H] +.

Example 31-Preparation of Compound 38 To a suspension of Pd (PPh3) 4 (0.02 g, 0.01 mmol) in anhydrous DME (8 mL) was added 4 (0.12 g, 0.27 mmol) and the mixture stirred at room temperature for 10 min. To this solution was added 3- (trifluoromethyl) phenylboronic acid (37; 0.12 g, 0.65 mmol) in a minimum of EtOH, followed by Na2CO3 (2M, 0.27 mL), and the resulting mixture was heated at reflux for 20 h. The cooled reaction mixture was diluted with water and extracted with CHZC12 (3 x 50 mL). The combined organic layers were washed with brine, dried over Na2S04, and concentrated. The reaction mixture was purified by column chromatography on normal phase silica gel followed by reversed phase column chromatography to obtain 38 (0.04 g, 33%) as an off white solid: mp 60-67 °C ;'H NMR (300 MHz, CDC13) 8 7.81 (s, 1 H), 7.74 (d, 1 H), 7.58- 7.45 (m, 7 H), 5.98 (s, 1 H), 4.90-4. 83 (m, 3 H), 4.63-4. 59 (m, 1 H), 3.90-3. 81 (m, 2 H), 3.66-3. 60 (m, 1 H), 1.68-1. 51 (m, 8 H), 1. 05-1. 00 (t, 3 H); IR (KBr) 3406,2969, 1602,1489, 1335 cm-1 ; CI MS m/z = 499 [C26H29FN70+H] +.

Example 32-Preparation of Compound 40 A mixture of 4 (0.13 g, 0.31 mmol), 2-naphthaleneboronic acid (39; 0.11 g, 0.62 mmol) and Pd (PPh3) 4 (0.09 g, 0.08 mmol) was placed in a sealed tube that was filled with argon. To the mixture was added toluene (5 mL) and Na2C03 (2M, 0.62 mL). The tube was quickly sealed and heated at 125 °C in an oil bath for 6 h. The cooled solution was diluted with water and extracted with CH2C12 (3 x 50 mL). The organic layers were washed with brine, dried over Na2S04, and concentrated. The reaction mixture was purified by column chromatography on normal phase silica gel, followed by reversed phase chromatography to give 40 (0.04 g, 28%): mp 70-75 °C ;'H NMR (300 MHz, CDC13) 8 8.02 (s, 1 H), 7.92-7. 84 (m, 3 H), 7.74-7. 67 (m, 3 H), 7.51-7. 44 (m, 5 H), 5.96 (s, 1 H), 4.89-4. 84 (m, 3 H), 4.66- 4.57 (m, 1 H), 3.93-3. 82 (m, 2 H), 3.67-3. 61 (m, 1 H), 1.76-1. 50 (m, 8 H), 1.06-1. 01 (t, 3 H); IR (KBr) 3422,2927, 1601,1491, 1388 cm''.

Example 33-Preparation of Compound 43 To compound 4 (0.14 g, 0.33 mmol) was added 4- methoxyphenylboronic acid (42,0. 11 g, 0.71 mmol), Pd (PPh3) 4 (0. 10 g, 0.087 mmol), Na2C03 (2M, 0.66 mL), and toluene (7 mL). The solution was degassed for 8 min with argon and heated in an oil bath at 125 °C for 6 h. The cooled solution was diluted with water and extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered, and concentrated. The reaction mixture was purified by normal phase column chromatography followed by reversed phase chromatography to give 43 (0.05 g, 28%) as a white solid: mp 128- 130 °C ;'H NMR (300 MHz, CDC13) 8 7.52-7. 50 (m, 5 H), 7.41 (d, 2 H), 6.97 (d, 2 H), 5.93 (s, 1 H), 4.89-4. 79 (m, 3 H), 4.63-4. 56 (m, 1 H), 3.92-3. 81 (m, 5 H), 3.67- 3.60 (m, 1 H), 1.68-1. 49 (m, 8 H), 1. 05-1. 00 (t, 3 H); IR (KBr) 3417,2931, 1610, 1499, 1389 cm~'; CI MS m/z = 461 [C26H32N602+H] +.

Example 34-Preparation of Compound 45 To a solution of s-BuLi (5 mL, 6.24 mmol) and TMEDA (1 mL) in anhydrous THF (35 mL) at-75 °C under argon was added dropwise a solution of N, N- diethylbenzamide (0.98 g, 5.57 mmol) in THF (5 mL). The mixture was stirred for 50 min and then treated with trimethylborate (2 mL, 17 mmol). The solution was allowed to warm to room temperature overnight. The colorless solution was cooled to 0 °C and acidified to pH = 6 with 2N HCI. The THF was removed in vacuo and the residue was diluted with water. This was extracted with CH2C12 (3 x 50 mL) and the combined organic layers were washed with brine, dried over Na2S04, concentrated in vacuo, followed by removal of trace solvent on the vacuum pump to give 45 as an off- white foamy solid :'H NMR (300 MHz, CD30D) 8 7.67-7. 39 (m, 4 H), 3.88-3. 69 (q, 4 H), 1.41-1. 30 (t, 6 H).

Example 35-Preparation of Compound 46 To compound 4 (0.14 g, 0.31 mmol) was added 2- (diethylcarbamoyl) phenylboronic acid (45,0. 29 g, 1.31 mmol), Pd (PPh3) 4 (0. 1 g, 0.09

mmol), Na2C03 (2M, 0.63 mL), toluene (5 mL), and the mixture degassed with argon for 10 min. The mixture was heated in an oil bath for 5 h at 135 °C. The cooled solution was diluted with water and extracted with CHzC (3 x 50 mL). The organic layers were combined, washed with brine, dried over Na2C03, and concentrated. The reaction mixture was purified by normal phase column chromatography on silica gel, followed by reversed phase chromatography to give 46 (0.03 g, 18%) as a yellow solid :'H NMR (300 MHz, CDC13) 6 7.49-7. 36 (m, 9 H), 6.18 (s, 1 H), 4.93 (d, 1 H), 4.78 (s, 2 H), 4.64-4. 55 (m, 1 H), 3.92-3. 60 (m, 4 H), 3.06-2. 92 (m, 2 H), 2.69-2. 64 (m, 1 H), 1.68-1. 51 (m, 8 H), 1.04-0. 99 (t, 3 H), 0.91-0. 86 (t, 3 H), 0.77-0. 72 (t, 3 H) ; CI MS m/z= 530 [C3oH39N702+H] +.

Example 36-Preparation of Compound 48 To a suspension of Pd (PPh3) 4 (0. 08 g, 0.69 mmol) in DME was added 4 (0.129 g, 0.30 mmol) and the mixture stirred for 10 min at room temperature. To this was added 3-nitrophenylboronic acid (47,0. 157 g, 0.94 mmol) and Na2C03 (2 M, 0.59 mL). The solution was heated at reflux under argon overnight. The cooled solution was diluted with water and extracted with CH2Cl2 (3 x 50 mL). The organic layers were combined, washed with brine, dried over Na2S04, and concentrated in vacuo. The solution was purified by flash column chromatography on silica gel to give 48 (0.04 g, 29%) as a bright yellow solid: mp 73-77 °C ;'H NMR (300 MHz, CDC13) 8 8.43 (s, 1 H), 8.20 (d, 1 H), 7.89 (d, 1 H), 7.63-7. 43 (m, 6 H), 6.01 (s, 1 H), 4.95-4. 76 (m, 3 H), 4.68-4. 58 (m, 1 H), 3.98-3. 80 (m, 2 H), 3.68-3. 60 (m, 1 H), 1.71- 1.40 (m, 8 H), 1.02-0. 98 (t, 3 H); IR (KBr) 3405,2930, 1713,1602, 1490,1351 cm-1 ; CI MS m/z = 476 [C25H29N703+H] +.

Example 37-Preparation of Compound 50 To a suspension of Pd (PPh3) 4 (0. 09 g, 0.08 mmol) in DME (5 mL) was added 4 (0.14 g, 0.32 mmol) and the mixture stirred at room temperature for 15 min.

To this was added benzo [b] furan-2-boronic acid (49,0. 153 g, 0.94 mmol) and Na2C03 (2 M, 0.63 mL). The solution was heated at reflux under argon overnight.

The reaction mixture was cooled, diluted with water, extracted with CH2Cl2 (3 x 50

mL). The organic layers were combined, washed with brine, dried over Na2S04, and concentrated in vacuo. The solution was purified by flash column chromatography on silica gel followed by flash column chromatography on reversed phase silica to give 50 (0.09 g, 60%) as a white solid :'H NMR (300 MHz, CDCl3) 8 7.82 (d, 2 H), 7.58- 7.42 (m, 5 H), 7.30-7. 19 (m, 2 H), 7.01 (s, 1 H), 6.11 (s, 1 H), 4.91 (d, 1 H), 4.81 (s, 2 H), 4.62-4. 58 (m, 1 H), 3.92-3. 80 (m, 2 H), 3.66-3. 60 (m, 1 H), 1.66-1. 48 (m, 8 H), 1.04-0. 99 (t, 3 H); CI MS m/z= 471 [C27H3oN602+H] +.

Example 38-Preparation of Compound 52 To compound 4 (0.46 g, 1.20 mmol) was added 1-amino-1- cyclopentanemethanol (51,1. 0 g, 8.61 mmol) and EtOH (2 mL) and the mixture was heated in an oil bath at 150 °C for 60 h. The brown solution was cooled and heated again at 150 °C for 48 h. The reaction mixture was cooled and concentrated in vacuo.

The reaction mixture was purified by flash column chromatography on silica gel to give 52 (0.39 g, 71%) as a tan solid : lH NMR (300 MHz, CDCI3) 6 7. 48-7.40 (m, 3 H), 7.29-7. 20 (m, 2 H), 6.88 (s, 1 H), 6.25 (s, 1 H), 5.10 (s, 1 H), 4.72 (s, 2 H), 4.63- 4.51 (m, 1 H), 3.78 (s, 2 H), 2.10-1. 65 (m, 8 H), 1.54 (d, 6 H); CI MS m/z = 459 [C2lH27BrN60+H] +.

Example 39-Preparation of Compound 53 To a suspension of Pd (PPh3) 4 (0.07 g, 0.06 mmol) in DME (5 mL) was added 52 (0.102 g, 0.22 mmol) and stirred at room temperature for 15 min. To this was added phenylboronic acid (0.098 g, 0.80 mmol) and Na2C03 (2 M, 0.44 mL).

The solution was heated at reflux under argon for 18 h. The reaction mixture was diluted with water, extracted with CH2Cl2 (3 x 50 mL), washed with brine, and dried over Na2S04. The solution was purified by flash column chromatography on silica gel followed by flash column chromatography on reversed phase silica gel to give 53 (0.02 g, 20%) :'H NMR (300 MHz, CDC13) 8 7.59-7. 31 (m, 10 H), 6.95 (s, 1 H), 5.95 (s, 1 H), 5.10 (s, 1 H), 4.79 (s, 2 H), 4.61-4. 52 (m, 1 H), 3.76 (s, 2 H), 2.01-1. 61 (m, 8 H), 1.54 (d, 6 H); CI MS m/z = 457 [C27H32N60+H] +.

Example 40-Preparation of Compound 54 To compound 3 (0.26 g, 0.67 mmol) was added trans-4- aminocyclohexanol hydrochloride (0.62 g, 4.11 mmol), Et3N (0.58 mL, 4.16 mmol), and ethanol (5 mL). The mixture was heated for 5 h at 135 °C in an oil bath. The temperature increased to 150 °C and heating was continued for a further 48 h. The solution was cooled and evaporated to give a yellow oil: CI MS m/z = 459 [C2lH27BrN60+H] +.

Example 41-Preparation of Compound 55 To compound 3 (0.50 g, 1.31 mmol) was added cis-1, 2- diaminocyclohexane (1.57 mL, 13.1 mmol) and EtOH (4 mL). The mixture was heated in an oil bath at 150 °C for 6 h. The reaction mixture was concentrated in vacuo. The reaction mixture was purified by column chromatography on silica gel to give 55 (0.49 g, 82%) as a yellow solid :'H NMR (300 MHz, CDC13) 8 7.43-7. 40 (m, 3 H), 7.23 (d, 2 H), 6.21 (s, 1 H), 5.04 (d, 1 H), 4.72 (s, 2 H), 4.67-4. 58 (m, 1 H), 4.08-4. 05 (m, 1 H), 3.17-3. 15 (m, 1 H), 2.08 (s, 2 H), 1.65-1. 38 (m, 14 H); CI MS m/z = 458 [C2iH2sBrN7+H] +.

Example 42-Preparation of Compound 56 To compound 55 (0.10 g, 0.22 mmol) was added 2- (tributylstannyl) pyridine (0.10 g, 0.27 mmol), Pd (PPh3) 4 (0.05 g, 0.04 mmol), and toluene (5 mL). The solution was degassed with argon for 8 min and heated at 135 °C for 3 h. The cooled solution was diluted with water, extracted with CH2C12 (3 x 50 mL), and the combined organic extracts were washed with brine, dried over Na2S04, filtered, and concentrated. The solution was followed by flash column chromatography (2 x) to give the desired product 56 (0.03 g, 36%) yellow crystalline solid :'H NMR (300 MHz, CDC13) 8 8.68 (d, 1 H), 7.96 (d, 2 H), 7.78-7. 69 (m, 2 H), 7.49 (s, 1 H), 7.44 (d, 2 H), 7.23-7. 18 (m, 1 H), 6.10 (s, 1 H), 5.10-5. 00 (m, 1 H), 4.83 (s, 2 H), 4.69-4. 60 (m, 1 H), 4.20-4. 10 (m, 1 H), 3.27-3. 13 (m, 1 H), 2.48 (s, 2 H), 1.78-1. 42 (m, 14 H); CI MS milz = 457 [C26H32N8+H] +.

Example 43-Preparation of Compound 57 To compound 1 (0.50 g, 1.31 mmol) was added trans-1, 2- diaminocyclohexane (2.52 mL, 21 mmol), and EtOH (6 mL). The reaction mixture was placed in an oil bath and heated to 190 °C for 25 h. The reaction mixture was removed from the heat and cooled to room temperature, concentrated for purification.

The reaction mixture was purified by column chromatography on silica gel to yield 57 (520 mg, 87%) as an off white foam :'H NMR (300 MHz, DMSO) 8 7.95 (bs, 1 H), 7.85 (s, 1 H), 7.50 (d, 2 H), 7.34 (d, 2 H), 6.17 (d, 1 H), 4.70-4. 40 (m, 1 H), 2.00-1. 71 (m, 4 H), 1.70-1. 52 (m, 2 H), 1.41 (d, 6 H), 1.30-0. 92 (m, 4 H); API MS m/z = 460 [C2lH28N7Br+H] +.

Example 44-Preparation of Compound 58 Compound 57 (0.15 g, 0.32 mmol) was added to a suspension of Pd (PPh3) 4 (0.11 g, 0.1 mmol) in DME (7 mL) and stirred at room temperature for 15 min. Phenylboronic acid (0.14 g, 1.14 mmol) was added followed by the Na2C03 (2M, 0.62 mmol). The reaction mixture was refluxed under argon for 18 h and allowed to stir at room temperature for 51 h. It was then diluted with water, extracted with CH2CI2, washed with brine, and then extracted with CH2CI2. The organic layer was evaporated, dried over anhydrous Na2S04, purified by column chromatography, and placed in vacuo for 18 h to give 58 (0.10 g, 72%) as a white solid : 1H NMR (300 MHz, CDC13) 8 7.62-7. 35 (m, 10 H), 5.92 (br, 1 H), 4.83 (br, 2 H), 4.74-4. 56 (m, 2 H), 3.77-3. 55 (m, 1 H), 2.55-2. 43 (m, 1 H), 2.16-1. 91 (m, 2 H), 1.73 (br, 2 H), 1.52 (d, 6 H), 1.37-1. 09 (m, 6 H); API MS m/z = 456 [C27H33N7+H] +.

Example 45-Preparation of Compound 59 To compound 57 (460 mg, 1.0 mmol) in solution with CH2CI2 (2 mL) was added acetic anhydride (0.44 mL, 4.6 mmol), catalytic DMAP, and pyridine (0.5 mL). The mixture was stirred at room temperature for 2.5 h. The mixture was diluted with CH2CI2, washed with 2N HCI, and the combined organics were then washed with NaHC03. The organics were then washed with brine, dried over Na2S04, filtered, and concentrated to give 59 (472 mg, 94%) as an off white solid : lH NMR

(300 MHz, DMSO-d6) 8 7.76 (s, 1 H), 7.42 (d, 2 H), 7.29 (d, 2 H), 4.68-4. 40 (m, 1 H), 4.10 (s, 3 H), 3.61-3. 40 (m, 2 H), 2.15-1. 80 (m, 2 H), 1.74-1. 55 (m, 4 H), 1.45 (d, 6 H), 1.35-1. 05 (m, 4 H); API MS mlz = 500 [C23H3oBrN70+H] +.

Example 46-Preparation of Compound 60 To a suspension of Pd (PPh3) 4 (0. 11 g, 0.1 mmol) in DME (7 mL) was added compound 59 (0. 15 g, 0.3 mmol) and stirred at room temperature for 15 min under argon. Phenylboronic acid (0.13 g, 1.06 mmol) was added, followed by Na2C03 (2M, 0.62 mL). The reaction mixture was refluxed under argon for 18 h.

The reaction mixture was then diluted with H2O, extracted with CH2C12, washed with brine, and extracted with CH2C12. The organic layer was dried over anhydrous Na2S04, purified by column chromatography, concentrated in vacuo for 18 h to yield 60 (61 mg, 42%) :'H NMR (300 MHz, DMSO-d6) 8 7.96 (s, 1 H), 7.72 (s, 1 H), 7.51 (t, 3 H), 7.40-7. 28 (m, 3 H), 7.28-7. 13 (m, 2 H), 5.84 (br, 1 H), 4.46 (br, 3 H), 3.47 (br, 2 H), 1.83 (br, 1 H), 1.62 (s, 4 H), 1.43 (d, 6 H), 0.12 (s, 3 H); API MS m/z = 498 [C29H35N70+H] +.

Example 47-Preparation of Compound 61 To compound 3 (0. 58 g, 1. 53 mmol) was added trans-1, 4- diaminocyclohexane (1.78 g, 15.6 mmol), and EtOH (4 mL). The mixture was heated in an oil bath at 150 °C for ca. 60 h. The reaction mixture was purified by column chromatography on silica gel to yield 61 (0.48 g, 68%) as an off white solid: mp 122- 125 °C ; H NMR (300 MHz, CDC13) 8 7.43 (s, 1 H), 7.40 (d, 2 H), 7.20 (d, 2 H), 6.27 (s, 1 H), 4.75-4. 68 (m, 2 H), 4.67-4. 58 (m, 2 H), 3.81-3. 68 (m, 1 H), 3.45 (s, 2 H), 2.88-2. 75 (m, 1 H), 2.18-2. 05 (m, 2 H), 2.05-1. 89 (m, 2 H), 4.52 (d, 6 H), 1.45-1. 13 (m, 4 H); CI MS m/z = 459 [C2tH28BrN7+H] +.

Example 48-Preparation of Compound 62 Amine 61 (53 mg, 0.12 mmol) was dissolved in CH2C12 (2 mL) and pyridine (5 mL). Acetic anhydride (0.05 g, 0.53 mmol) and DMAP (few crystals) were added. The reaction mixture was allowed to stir at room temperature for 2.25 h.

The reaction mixture was diluted with CH2Cl2, washed with 2N HCI, NaHC03, dried over MgS04, filtered, and evaporated to yield 62 (0.05 g, 78%) as a white solid : lH NMR (300 MHz, CDC13) 8 7.50-7. 20 (m, 5 H), 6.02 (br, 1 H), 5.29-5. 20 (m, 1 H), 4.72 (d, 2 H), 4.66-4. 54 (m, 2 H), 3.72 (br, 2 H), 2.18-2. 06 (m, 2 H), 2.06-1. 91 (m, 2 H), 1.97 (s, 3 H), 1.54 (d, 6 H), 1.36-1. 15 (m, 4 H); API MS m/z = 500 [C23H3oBrN70+H] +.

Example 49-Preparation of Compound 64 Compound 61 (0.05 g, 0.11 mmol) was dissolved in CH2C12 (3 mL) and Et3N (2 mL) and placed in an ice bath for 10 min. Compound 63 (0.06 g, 0.22 mmol) was dissolved in CHzC (2 mL), added dropwise, and rinsed with CHsCIs (1.5 mL). The ice bath was removed after 20 min and the reaction was allowed to stir for 7 d. The reaction mixture was diluted with CH2C12, washed with 2N HCl until the aqueous layer was acidic, washed with NaHC03, dried over MgS04, and evaporated.

The desired product was isolated by column chromatography and dried in vacuo to yield 64 (0.04 g, 50%) as a green solid :'H NMR (300 MHz, CDC13) 8 8.53 (d, 1 H), 8.32-8. 20 (m, 2 H), 7.59-7. 35 (m, 4 H), 7.23-7. 11 (m, 4 H), 6.02 (br, 1 H), 4.69-4. 45 (m, 5 H), 3.57 (br, 1 H), 3.12 (br, 1 H), 2.87 (s, 1 H), 1.97 (br, 2 H), 1.75 (br, 2 H), 1.48 (d, 6 H), 1.27-0. 97 (m, 4 H); API MS mlz = 693 [C33H39BrN802S+H] +.

Example 50-Preparation of Compound 65 Compound 61 (0.05 g, 0.11 mmol) was dissolved in CH2Cl2 (3 mL) and Et3N (2 mL) and placed in an MeOH/ice bath. Methanesulfonyl chloride (0.012 mg, 0.11 mmol) in CH2C12 (2.3 mL) was slowly added. The reaction mixture and ice bath was allowed to come to room temperature. After 1.5 h, the reaction mixture was diluted with CH2C12, washed with 2N HCl until the aqueous layer was acidic. The organic layer was washed with NaHC03, dried over MgS04, filtered, and evaporated.

The product was purified by column chromatography, and dried in vacuo for 14 h to yield 65 (13 mg, 24%) as an off-white solid :'H NMR (300 MHz, CDC13) 8 7.50-7. 17 (m, 5 H), 5.90 (br, 1 H), 4.75-4. 57 (m, 3 H), 4. 11 (d, 1 H), 3.69 (br, 1 H), 3.30 (br, 1

H), 2.99 (s, 3 H), 2.18-2. 03 (m, 4 H), 1.69 (d, 6 H), 1.42-1. 15 (m, 5 H); API MS m/z = 538 [C22H3oBrN702S+H] +.

Example 51-Preparation of Compound 66 Compound 61 (0.05 g, 0.11 mmol) was dissolved in toluene (4 mL).

2-Acetylphenylisocyanate (0.024 g, 0.15 mmol) diluted with toluene (1 mL) and added to compound 61. Toluene (6 mL) was added to the reaction mixture. The reaction mixture was placed under reflux for 19 h. The product was purified by column chromatography, concentrated, and dried in vacuo for 23 h to yield 66 (42 mg, 62%) as an off-white solid :'H NMR (300 MHz, CDCl3) 8 7.87-7. 20 (m, 9 H), 6.41 (s, 1 H), 5.86 (br, 1 H), 4.75-4. 54 (m, 4 H), 3.69 (br, 1 H), 2.60 (s, 3 H), 2.12 (br, 4 H), 1.51 (d, 6 H), 1.42-1. 15 (m, 5 H); API MS m/z = 619 [C3oH3sBrN802+H] +.

Example 52-Preparation of Compound 67 Compound 61 (0.04 g, 0.10 mmol) was dissolved in CH2C12 (2 mL) and pyridine (0.5 mL). Cyclopropanecarbonyl chloride (0.05 g, 0.44 mmol) was added along with DMAP (small amount). The reaction mixture was allowed to stir at room temperature for 2.25 h. The reaction mixture was diluted with CH2C12, washed with 2N HCI, saturated NaHC03, dried over MgS04, filtered, and evaporated. The product was isolated by column chromatography to yield 67 (0.03 g, 63%) as a white solid : lH NMR (300 MHz, CDC13) 8 7.50-7. 20 (m, 5 H), 5.96 (br, 1 H), 5.41 (d, 1 H), 4.72 (d, 2 H), 4.66-4. 54 (m, 2 H), 3.72 (br, 2 H), 2.18-1. 97 (m, 4 H), 1.51 (d, 6 H), 1.36-1. 15 (m, 5 H), 1.06-0. 88 (m, 2 H), 0.79-0. 67 (m, 2 H); API MS m/z= 526 [C25H32BrN70+H] +.

Example 53-Preparation of Compound 69 To a solution of 4-biphenylcarboxaldehyde (1.0 g, 5.49 mmol) in MeOH (20 mL) was added NaBH3CN (0.69 g, 11.0 mmol), and NH40H (15 mL) and the mixture was stirred at room temperature overnight. To this added HCl and extracted with CHCl3. The resulting aqueous layer was brought to pH > 7 with sodium bicarbonate and then extracted with CHC13. The solution was dried with

MgS04, filtered, and evaporated to give 69 (200 mg) as a white solid: EI MS m/z = 183 [CuHN]".

Example 54-Preparation of Compound 69 To compound 70 (2.75 g, 13.9 mmol) was added anhydrous THF (60 mL), heated to reflux, and kept under nitrogen. 1M Borane-THF (69.7 mL) was added dropwise to 70 through an addition funnel resulting in a homogeneous solution.

The solution was refluxed for 18 h. The reaction mixture was cooled in an ice water bath and quenched with H20, 2N HCl (20 mL), followed by 3N NaOH (60 mL). The reaction mixture was extracted with EtOAc (3 x). The organic extracts were washed with brine, and dried over sodium sulfate. The crude product was concentrated, dissolved in MeOH, and HC1 gas was bubbled through the solution. The solution was filtered in vacuo to give 69 as a white solid :'H NMR (300 MHz, CD30D) 8 7.71 (d, 2 H), 7.63 (d, 2 H), 7.52 (d, 2 H), 7.47-7. 30 (m, 3 H), 4.13 (s, 2 H).

Example 55-Preparation of Compound 71 To compound 1 (6.8 g, 36.0 mmol) and 69 (8.0 g, 36.5 mmol) was added H20 (60 mL) and Hunigs base (9.0 g, 70.0 mmol). The mixture was stirred and heated to reflux for 5 h during which time H20 (50 mL) was added as the reaction continued to thicken. The crude product was collected by filtration, washed with H20 (500 mL) and EtOH (2 x 30 mL), air dried, and dried in vacuo to give 71 (11.1 g, 92%): mp 267-269 °C.

Example 56-Preparation of Compound 72 Compound 71 (4.7 g, 14.0 mmol), K2C03 (15.0 g, 109 mmol), DMSO (80 mL), and 2-iodopropane (9.4 g, 55.0 mmol) were combined and stirred overnight.

H20 and EtOAc were added. The EtOAc layer was separated and washed with brine (3 x). The EtOAc solution was dried with MgS04, concentrated, and crystallized from EtOAc to give 72 (3.5 g, 66%): mp 139-140 °C.

Example 57-Preparation of Compound 73 Compound 72 (2.00 g, 5.30 mmol) and (R)-(-)-2-amino-1-butanol (10.8 g, 121 mmol) were combined in a sealed tube, and heated in an oil bath at 190 °C for 2 h. The solution was cooled to 60 °C, diluted in EtOAc, washed with brine (4 x), dried with Na2S04, and concentrated. Purification by column chromatography on Si02 gave the desired product 73 (1.72 g, 75%) as a foam : 1H NMR (300 MHz, CDC13) 8 7.65-7. 10 (m, 9 H), 6.40-6. 10 (bs, 1 H), 5.05-4. 85 (m, 1 H), 4.85-4. 67 (m, 1 H), 4.60 (heptuplet, 1 H), 4.00-3. 70 (dd, 2 H), 3.76-3. 50 (m, 1 H), 1.95 (bs, 1 H), 1.80-1. 55 (m, 2 H), 1.51 (d, 6 H), 1.03 (t, 3 H); IR (CH2C12) 3301,2969, 1601,1488, 1389,1255, 762,698 cm-1 ; API MS mlz = 431 [C25H3oN60+H] +.

Example 58-Preparation of Compound 74 Compound 72 (0.23 g, 0.60 mmol), cis-1, 2-diaminocyclohexane (0.72 mL, 6.0 mmol), and ethanol (2 mL) were combined in a sealed tube and heated in an oil bath at 155 °C for 5 d. The ethanol was removed in vacuo and the crude reaction mixture was filtered through a silica plug. The reaction mixture was chromatographed on silica gel, the resulting orange solid was dissolved in CH2C12 and a portion of activated charcoal was added. The solution was filtered through a pad of celite and concentrated to give 74 as a yellow solid (0.04 g, 27%) :'H NMR (300 MHz, CDC13) 7.59-7. 31 (m, 10 H), 6.00 (s, 1 H), 5.09 (d, 1 H), 4.83 (s, 2 H), 4.68- 4.62 (m, 1 H), 4.11 (s, 1 H), 3.70-3. 65 (m, 2 H), 3.18-3. 16 (m, 1 H), 2.02 (s, 2 H), 1.67-1. 42 (m, 12 H); CI MS m/z= 456 [C27H33N7+H] +.

Example 59-Preparation of Compound 75 Compound 72 (0.17 g, 0.45 mmol), trans-1, 4-diaminocyclohexane (0.53 g, 4.69 mmol), and EtOH (5 mL) were combined in a sealed tube and heated at 155 °C for 5 d. The EtOH was removed in vacuo and the crude mixture was subjected to flash chromatography on silica gel. Recrystallization from CHCl3/MeOH gave 75 (5.8 mg) as an off-white crystalline solid: mp 110-112 °C ;'H NMR (300 MHz, CDC13) 6 7.58-7. 31 (m, 10 H), 5.95 (s, 1 H), 4.88-4. 78 (m, 2 H), 4.69-4. 60 (m,

2 H), 3.88-3. 78 (m, 1 H), 3.07-2. 98 (m, 1 H), 2.26-2. 10 (m, 4 H), 1.62-1. 52 (m, 8 H), 1.29-1. 15 (m, 4 H); CI MS milz = 456 [C27H33N7+H] Example 60-Preparation of Compound 76 Compound 75 (0.05 g, 0.11 mmol) was dissolved in CH2C12 and the solution cooled to 0 °C under an argon atmosphere. A catalytic amount of DMAP, triethylamine (50 L, 0.36 mmol), followed by the acetyl chloride (25 L, 0.36 mmol) were added to the reaction mixture. The solution was warmed to room temperature and washed with NaHC03 (5%), water, and brine. The solution was dried over Na2S04 and concentrated. Purification by flash chromatography on silica gel gave 76 (0.028 g, 53%) as a pale yellow solid: mp 224-225 °C ; lH NMR (300 MHz, CDCl3) 8 7.59-7. 31 (m, 10 H), 5.93 (s, 1 H), 5.26 (d, 1 H), 4.81 (s, 2 H), 4.65-4. 58 (m, 1 H), 3.78-3. 75 (m, 2 H), 2.18-1. 99 (m, 4 H), 1.95 (s, 3 H), 1.77 (s, 1 H), 1.53 (d, 6 H), 1.32-1. 22 (m, 4 H); CI MS m/z= 498 [C29H3sN7O+H] +.

Example 61-Preparation of Compound 77 Compound 72 (0. 15 g, 0.40 mmol), trans-4-aminocyclohexanol hydrochloride (0.31 g, 1.99 mmol), Et3N (0.11 mL, 0.8 mmol), and EtOH (5 mL) were combined and heated in a sealed tube at 155 OC for 4 d. Additional trans-4- aminocyclohexanol hydrochloride (0.34 g, 2.2 mmol) and triethylamine (0.60 mL, 4.3 mmol) were added and the heat was resumed at 155 °C overnight. The crude product was purified by flash column chromatography to give 77 (0.036 g, 20%) as an off- white solid: mp 196-200 °C ;'H NMR (300 MHz, CDC13) 8 7.58-7. 30 (m, 10 H), 5.97 (s, 1 H), 4.83-4. 81 (m, 2 H), 4.66-4. 60 (m, 2 H), 3.82-3. 77 (m, 1 H), 3.69-3. 62 (m, 1 H), 2.17-2. 13 (m, 2 H), 2.01-1. 97 (m, 2 H), 1.68 (s, 1 H), 1.53 (d, 6 H), 1.49-1. 20 (m, 4 H); CI MS mlz = 457 [C27H33N60+H] +.

Example 62-Preparation of Compound 78 To compound 61 (0.12 g, 0.26 mmol), was added compound 16 (0.12 g, 0.33 mmol), and Pd (PPh3) 4 (0.06 g, 0.056 mmol) and toluene (5 mL). The resulting mixture was degassed for 10 min with argon. The mixture was heated at 140 °C for 3

h. The cooled solution was diluted with saturated NaHC03 and extracted with CH2C12 (3 x 50 mL). The combined organic extracts were washed with brine, dried over Na2S04, filtered, and concentrated to give a pale yellow oil which crystallized upon standing at room temperature. The crude product was purified by column chromatography and concentrated to give a white solid. The solid was precipitated with acetonitrile, filtered, washed with ether and hexane to give 78 (0.02 g, 18%) :'H NMR (300 MHz, DMSO-d6) 8 8.63 (d, 1 H), 8.01 (d, 1 H), 7.93-7. 83 (m, 2 H), 7.59- 7.44 (m, 4 H), 7.34-7. 29 (m, 1 H), 6.25 (s, 1 H), 4.70-4. 60 (m, 2 H), 4.57-4. 49 (m, 2 H), 3.65-3. 52 (m, 1 H), 2.98-2. 88 (m, 1 H), 1.98-1. 90 (m, 4 H), 1.48 (d, 6 H), 1.42- 1.18 (m, 6 H); CI MS m/z = 457 [C26H32N8+H]- Example 63-Preparation of Compound 78 To compound 24 (200 mg, 0.53 mmol) was added trans-1, 4- diaminocyclohexane (2.00 g, 17 mmol) and EtOH (4 mL). The reagents were heated in a sealed tube in an oil bath at 170 °C for 18 h. The mixture was cooled to 60 °C and partitioned between EtOAc and brine. The EtOAc layer was separated, washed with brine (3 x), dried with Na2SO4, and concentrated to give 78 (0.12 g, 50%): mp 135-138 °C ;'H NMR (300 MHz, CDC13) 8 8.03-7. 82 (m, 2 H), 7.80-7. 58 (m, 3 H), 7.57-7. 30 (m, 3 H), 7.30-7. 05 (m, 1 H), 6.20 (bs, 1 H), 5.95-4. 73 (m, 2 H), 4.73-4. 45 (m, 2 H), 3.90-3. 60 (m, 1 H), 2.80-2. 52 (m, 1 H), 2.25-1. 80 (m, 4 H), 1.80-1. 60 (bs, 3 H), 1.52 (d, 6 H), 1.38-1. 05 (m, 4 H); IR (KBr) 3422,2927, 1599,1489, 1253,779 cm-1 ; API MS m/z = 457 [C26H32N8+H]- Example 64-Preparation of Compound 79 Compound 78 (50 mg, 0.11 mmol) was dissolved in CH2C12 (2 mL) and stirred at room temperature. Pyridine (0.5 mL), Ac20 (0.5 mL, 4.9 mmol), and DMAP (few crystals) were added to the reaction mixture and stirred for 2 h. The solution was diluted in CH2CI2 and washed in 2N HCI. The HCI layer was concentrated, CH2CI2 was added and the aqueous phase neutralized with saturated NaHCO3. The CH2CI2 layer was separated, dried (MgS04), and concentrated to give 79 (0.03 g, 55%) as a white solid :'H NMR (300 MHz, CDC13) 8 8.00-7. 80 (m, 2 H),

7.81-7. 57 (m, 2 H), 7.56-7. 33 (m, 3 H), 7.30-7. 05 (m, 2 H), 6.15-5. 90 (bs, 1 H), 5.47- 5.28 (m, 1 H), 4.96-4. 72 (m, 2 H), 4.73-4. 45 (m, 2 H), 2.25-1. 82 (m, 4 H), 2.00 (s, 3 H), 1.54 (d, 6 H), 1.40-1. 00 (m, 4 H); API MS m/z = 499 [C28H34Ng0+H] +.

Example 65-Preparation of Compound 80 Compound 74 (0.02 g, 0.05 mmol) was dissolved in dry benzene (5 mL) and stirred under a blanket of argon. The solution was cooled in an ice bath and phenylisocyanate (25 L, 0.23 mmol) was added dropwise. The ice bath was removed and the mixture stirred at room temperature for 0.5 h. The solvent was evaporated in vacuo to give a yellow oil. The crude product was purified by flash column chromatography on silica gel to give 80 (0.008 g) : 1H NMR (300 MHz, CDC13) 6 7.53-7. 30 (m, 10 H), 7.13-7. 06 (m, 4 H), 6.98-6. 88 (m, 1 H), 6.62 (s, 1 H), 6.02 (s, 1 H), 5.65 (s, 1 H), 5.02 (d, 1 H), 4.85-4. 70 (m, 2 H), 4.60-4. 52 (m, 1 H), 4.45-4. 40 (m, 1 H), 4.36-4. 22 (m, 2 H), 4.00 (s, 1 H), 1.91-1. 60 (m, 6 H), 1.48-1. 43 (m, 6 H).

Example 66-Preparation of Compound 82 A mixture of 6-chloronicotinamide (2.96 g, 18.9 mmol), phenylboronic acid (2.54 g, 20.8 mmol), and Pd (PPh3) 4 (643 mg, 0.565 mmol) in toluene (47 mL), ethanol (7 mL) and 2M aqueous sodium carbonate solution (21 mL, 43 mmol) was stirred and heated at 90-100 °C under nitrogen for 16 h. The mixture was cooled to room temperature and filtered. The resulting solid was washed with water (2 x 20 mL) and dried in vacuo. To the dried solid was added methanol (50 mL). The mixture was stirred at reflux, cooled to room temperature, and filtered to give the product (90%) as a powder: mp 218-220 °C ;'H NMR (500 Hz, DMSO-d6) 8 9.23 (d, J = 2. 5 Hz, 1 H), 8.41 (dd, J, = 2.2 Hz, J2 = 8.3 Hz, 1 H), 8.32 (s, I H), 8.27 (d, J = 7.1 Hz, 2 H), 8.20 (d, J = 8.5 Hz, 1 H), 7.74 (s, 1 H), 7.66-7. 60 (m, 3 H).

Example 67-Preparation of Compound 83 To NaBH4 (0.19 g, 5 mmol) in 1,4-dioxane (4 mL) was added HOAc (0.3 g, 5 mmol) in 1,4-dioxane (2 mL) slowly while the flask was cooled with ice.

Compound 82 (0.2 g, 1 mmol) was then added. The mixture was stirred at reflux at

100-110 °C for 4 h and the solvent was evaporated. To this mixture was added water (2 mL) slowly. The mixture was extracted with CHzC (4 x 10 mL), washed with water (3 x 5 mL), dried with anhydrous sodium sulfate, concentrated, and purified by flash chromatography on silica gel to provide the product as a yellow liquid. This was triturated with ethanol (1 mL) to provide a white solid which was collected (60%) and dried: mp 97-99 °C ;'H NMR (500 Hz, CDC13) 8 8.60 (d, J = 2 Hz, 1 H), 7.97-7. 95 (m, 2 H), 7.72-7. 67 (m, 2 H), 7.47-7. 37 (m, 3 H), 3.90 (s, 2 H), 1.77 (bs, 2 H).

Example 68-Preparation of Compound 84 A mixture of 2, 6-dichloropurine (1,0. 19 g, 1 mmol), amine 83 (0. 39 g, 2.15 mmol) in ethanol (13 mL), and water (3 mL) was heated at 100-110 °C under nitrogen for 24 h and then cooled to room temperature. The mixture was concentrated and water (5 mL) was added. A solid was filtered and washed with water (2 x 5 mL) and dried under vacuum to give the product (80%) as a yellow solid: mp 260 °C (dec) ;'H NMR (500 Hz, DMSO-d6) 8 13.26 (s, 1 H), 8.79 (s, 1 H), 8.27 (s, 1 H), 8.16 (d, J = 7. 1 Hz, 2 H), 8.34 (d, J = 7.3 Hz, 1 H), 7.96 (d, J = 7.6 Hz, 1 H), 7.63-7. 52 (m, 3 H), 4.81 (s, 2 H).

Example 69-Preparation of Compound 85 To a solution of compound 84 (0.34 g, 1 mmol) in DMSO (5 mL), was added potassium carbonate (0.7 g, 5 mmol) and 2-iodopropane (0.5 g, 3 mmol). The mixture was stirred at ambient temperature under nitrogen for 24 h and poured into ice water (30 mL). After filtration, the solid was washed with water (4 x 5 mL), dried under vacuum to give the crude product as a yellow solid. Flash column chromatography of the crude product on silica gel and recrystallization provided the pure product (63%) as ivory colored crystals: mp 138-139 °C ;'H NMR (500 Hz, CDC13) 8 8.70 (d, J =1.5 Hz, 1 H), 7.97 (m, 2 H), 7.79 (dd, J = 1.7 Hz, J2 = 8.1 Hz, 1 H), 7.71 (s, 1 H), 7.69 (d, J = 8.1 Hz, 1 H), 7.48-7. 39 (m, 3 H), 4.87 (s, 2 H), 4.80 (m, 1 H), 1.55 (d, J = 6. 8 Hz, 6 H); CI MS m/z = 379 [C2oHl9ClN6+H] +. Anal. Calcd. for C20HlgCIN6 : C, 63.41 ; H, 5. 05 ; N, 22.18. Found: C, 63.75 ; H, 5.09 ; N, 21.87.

Example 70-Preparation of Compound 86 To compound 85 (0.1 g, 0.26 mmol) was added trans-1, 4- diaminocyclohexane (1 g, 8.8 mmol) and EtOH (2 mL). The reaction mixture was heated in a sealed tube in an oil bath at 120 °C. The crude product was purified by column chromatography to give 86 (0.08 g, 67%) :'H NMR (300 MHz, CDCI3) 8 8.68 (d, 1 H), 7.83-7. 97 (m, 2 H), 7.70-7. 83 (m, 1 H), 7.55-7. 73 (m, 1 H), 7.30-7. 55 (m, 4 H), 6.35 (bs, 1 H), 4. 72-4. 95 (m, 2 H), 4.50-4. 72 (m, 2 H), 3.63-3. 85 (m, 1 H), 2.65-2. 90 (m, 1 H), 2.37-2. 63 (bs, 2 H), 1.80-2. 20 (dd, 4 H), 1.53 (d, 6 H), 0.72-1. 42 (m, 4 H); API MS m/z= 457 [C26H22N8+H] +.

Example 71-Preparation of Compound 87 Compound 86 (0.08 g, 0.18 mmol) was stirred at room temperature in CH2C12 (3 mL). Pyridine (100 mg, 0.82 mmol) was added followed by Ac20 (100 mg, 0.98 mmol) and DMAP (few crystals). After 2 h, more CH2C12 (3 mL) was added and the mixture was washed carefully with 2N HCl (10 drops), and saturated NaHCO3. After separation of the CH2CI2 layer, the organic phase was then dried with Na2S04 and concentrated to give 87 (80 mg, 92%) :'H NMR (300 MHz, CDC13) 8 8.72 (s, 1 H), 8.30-7. 03 (m, 9 H), 5.75-5. 38 (m, 1 H), 5.02 (bs, 1 H), 4.83 (bs, 2 H), 4.72-4. 40 (m, 1 H), 3.73 (bs, 2 H), 2.52-1. 83 (m, 4 H), 1.98 (s, 3 H), 1.52 (d, 6 H), 1.50-1. 00 (m, 4 H); API MS m/z = 499 [C28H34N8O+H] +.

Example 72-Preparation of Compound 88 Compound 85 (0.05 g, 0.13 mmol) and (R)-(-)-2-amino-1-butanol (0.50 g, 5.6 mmol) were combined in a sealed tube and heated in an oil bath at 190 °C for 2 h then cooled to room temperature. The mixture was partitioned between EtOAc and brine, washed with brine (3 x), dried with Na2S04, and concentrated. The mixture was allowed to stand over the weekend and then purified by column chromatography on Si02 to give 88 (0.01 g, 17%) as a foam :'H NMR (300 MHz, CDC13) 8 8.70 (s, 1 H), 8.05-7. 82 (m, 2 H), 7.82-7. 55 (m, 2 H), 7.57-7. 30 (m, 4 H), 6.55 (bs, 1 H), 5.00-4. 88 (s, 1 H), 4.78 (s, 2 H), 4.60 (heptuplet, 1 H), 3.98-3. 83 (m, 1

H), 3.84-3. 70 (m, 1 H), 3.70-3. 50 (m, 1 H), 2.90 (bs, 1 H), 1.75-1. 55 (m, 2 H), 1.53 (d, 6 H), 1.00 (t, 3 H); API MS m/z = 432 [C24H29N70+H] +.

Example 73-Preparation of Compound 89 A mixture of 6-chloronicotinamide (2.5 g, 16 mmol), crude 2- trimethylstannylpyridine (5.8 g, 24 mmol), and PdCl2 (PPh3) 2 (560 mg, 0.8 mmol) in DMF (35 mL) was heated at 150-160 °C in a pressure tube for 17 h. The DMF was distilled off under reduced pressure and the residue was extracted with ethyl acetate (6 x 30 mL) and concentrated. The residue was treated with methanol (15 mL) and a solid separated which was filtered and dried to give the product (40%) as a powder: mp 237-240 °C ; lH NMR (500 Hz, DMSO-d6) 9.22 (d, J = 2. 2 Hz, 1 H), 8.83 (m, 1 H) 8.57-8. 53 (m, 2 H), 8.48-8. 46 (m, 1 H), 8.38 (s, 1 H), 8.11-8. 07 (m, 1 H), 7.78 (s, 1 H), 7.63-7. 60 (m, 1 H).

Example 74-Preparation of Compound 90 To NaBH4 (0.2 g, 5 mmol) in 1,4-dioxane (4 mL) was added HOAc (0.29 g, 5 mmol) in 1,4-dioxane (2 mL) slowly while the flask was cooled with ice.

Compound 89 (0.199 g, 1 mmol) was then added. The mixture was stirred at reflux at 100-110 °C for 4 h and the solvent was evaporated. To this mixture was added water (2 mL) slowly. The mixture was extracted with CH2C12 (4 x 10 mL), washed with water (3 x 5 mL), dried with anhydrous sodium sulfate, filtered, concentrated, and purified by flash chromatography on silica gel to provide the product as a yellow liquid. This was triturated with ethanol (1 mL) and a white solid (32%) was collected and dried: mp 109-112 °C ;'H NMR (500 Hz, CDC) 8 8.63 (m, 1 H), 8.58 (s, 1 H), 8.32 (m, 2 H), 7.77 (m, 2 H), 7.25 (m, 1 H), 3.91 (s, 2 H), 1.94 (s, 2 H).

Example 75-Preparation of Compound 91 A mixture of 2, 6-dichloropurine (1,0. 2 g, 1 mmol), compound 90 (0.4 g, 2.2 mmol) in ethanol (13 mL), and water (3 mL) was heated at 100-110 °C under nitrogen for 24 h and then cooled to room temperature. The mixture was concentrated and water (5 mL) was added. A solid was filtered and washed with water (2 x 5 mL)

and dried under vacuum to give the product (83%) as a yellow solid: mp 248 °C (dec) ;'H NMR (500 Hz, DMSO-d6) 8 13.27 (s, 1 H), 8.81 (s, 1 H), 8.78 (d, J = 4. 1 Hz, 1 H), 8.47 (m, 2 H), 8.28 (s, 1 H), 8.06-8. 01 (m, 2 H), 7.50 (m, 1 H), 4.84 (s, 2 H).

Example 76-Preparation of Compound 92 To the solution of compound 91 (0.35 g, 1 mmol) in DMSO (5 mL), added potassium carbonate (0.68 g, 5 mmol) and 2-iodopropane (0.49 g, 3 mmol).

The mixture was stirred at ambient temperature under nitrogen for 24 h and poured into ice water (30 mL). After filtration, the solid was washed with water (4 x 5 mL), dried under vacuum to give the crude product as a yellow solid. Flash column chromatography of the crude product on silica gel and recrystallization provided the pure product (64%) as white crystals: mp 150-151 °C ;'H NMR (500 Hz, CDC13) 8 8.71 (d, J = 1.9 Hz, 1 H), 8.67 (m, 1 H), 8.38-8. 36 (m, 2 H), 7.86-7. 79 (m, 2 H), 7.75 (s, 1 H), 7.30 (m, 1 H), 4.91 (s, 2 H), 4.82 (m, 1 H), 1.57 (d, J = 6.8 Hz, 6 H); CI MS m/z = 380 [ClgHl8ClN7+H] +. Anal. Calcd. for Cl9Hl8CIN7 : C, 60.08 ; H, 4.78 ; N, 25. 81. Found: C, 59.76 ; H, 4.72 ; N, 25.57.

Example 77-Preparation of Compound 93 Compound 92 (150 mg, 0.39 mmol), trans-1, 4-diaminocyclohexane (1.50 g, 13.1 mmol), and EtOH (30 mL) were heated to 120 °C for 26 h in a sealed tube. The mixture was cooled, additional EtOAc was added, washed with brine, dried over Na2S04, and concentrated to give 93 (170 mg, 94%) as a waxy solid :'H NMR (300 MHz, CDC13) 8 8.77-8. 60 (m, 1 H), 8.44-8. 27 (m, 2 H), 7.90-7. 75 (m, 2 H), 7.50 (s, 1 H), 7.36-7. 22 (m, 2 H), 6.27 (bs, 1 H), 4.96-4. 73 (m, 2 H), 4.73-4. 52 (m, 2 H), 3.84-3. 60 (m, 1 H), 2.80-2. 57 (m, 1 H), 2.22-2. 00 (m, 2 H), 2.00-1. 67 (m, 5 H), 1.54 (d, 6 H), 1.38-1. 05 (m, 4 H); API MS m/z = 458 [C25H3lN9+H] +.

Example 78-Preparation of Compound 94 Compound 93 (0.15 g, 0.33 mmol) was dissolved in CH2CI2 (6 mL) and then pyridine (0.200 g, 1.64 mmol) followed by AczO (0.200 g, 1.96 mmol) and DMAP (few crystals) were added. The reaction mixture was stirred for 2 h, washed

with 2N HCl and NaHC03, extracted with CH2C12, dried with Na2S04, and concentrated to give 94 (0.17 g, 69%) as a solid: mp 141-145 °C ;'H NMR (300 MHz, CDCl3) 6 8.80-8. 63 (m, 1 H), 8.45-8. 25 (t, 2 H), 7.95-7. 73 (m, 1 H), 7.52 (s, 1 H), 7.35-7. 20 (m, 2 H), 6.20 (bs, 1 H), 5.50-5. 30 (m, 1 H), 4.98-4. 75 (m, 2 H), 4.75- 4.50 (m, 2 H), 3.84-3. 60 (m, 2 H), 2.27-1. 87 (m, 4 H), 2.00 (s, 3 H), 1.52 (d, 6 H), 1. 40-1. 10 (m, 4 H); API MS m/z = 499 [C27H33N90+H].

Example 79-Preparation of Compound 95 DME (3 mL), tris (dibenzylideneacetone) dipalladium (0.01 g, 0.01 mmol), and PPh3 (0.04 g, 0.15 mmol) were added to a round bottomed flask equipped with a condensor and maintained under an argon atmosphere. To the solution was added compound 11 (0.13 g, 0.25 mmol). 3-Fluorobenzene boronic acid (0.123 g, 0.9 mmol) was dissolved in a solution of 2M Na2C03 (0.6 mL) and DME (1 mL), and added to the reaction mixture. The mixture was stirred under argon and refluxed for 19 h then stirred at room temperature for 22 h. The reaction mixture was diluted with H20, extracted with CH2Cl2, washed with brine. The organic layer was dried over Na2S04 and evaporated. The reaction mixture was purified twice by column chromatography and dried under high vacuum to give a white solid (17 mg, 14%) :'H NMR (300 MHz, CDCl3) 5 7.56-7. 32 (m, 8 H), 7.08-6. 99 (m, 1 H), 5.86 (br, 1 H), 4.83 (d, 2 H), 4.71-4. 56 (m, 1 H), 3.77 (br, 2 H), 2.70 (br, 1 H), 2.12 (d, 1 H), 1.88 (d, 1 H), 1.51 (d, 6 H), 1.22 (d, 5 H), 0.94-0. 70 (m, 3 H); API MS m/z = 474 [C27H32FN7+H] +.

Example 80-Preparation of Compound 96 A stock solution of acetic anhydride was made by mixing CH2C12 (16 mL), pyridine (4 mL), and Ac2O (0.16 mL). To this stock solution (1.5 mL) was added compound 95 (0.01 g, 0.02 mmol) followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 26 h. The reaction mixture was then diluted with CH2C12, washed with 2N HCl until the aqueous layer was acidic, washed with NaHC03, dried over MgS04, evaporated, and dried in vacuo for 15 h to give a white solid (11 mg, 92%) :'H NMR (300 MHz, CDC13) 8 8.65 (br,

1 H), 7.77-7. 17 (m, 8 H), 7.11-6. 99 (m, 1 H), 5.14 (br, 2 H), 4.90 (br, 1 H), 4.69 (br, 1 H), 3.78 (br, 2 H), 2.09 (br, 3 H), 1.94 (s, 2 H), 1.57 (d, 6 H), 1.42 (br, 4 H), 1.24 (s, 2 H), 0.94-0. 76 (m, 1 H); CI MS milz = 516 [C29H34FN70+H] +.

Example 81-Preparation of Compound 97 A stock solution of acetic anhydride was made by mixing CH2C12 (16 mL), pyridine (4 mL), and Ac20 (0.16 mL). To this stock solution (1.5 mL) was added compound 13 (0.01 g, 0.02 mmol) followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 2 h. The reaction mixture was then diluted with CH2Cl2, washed with 2N HCl until it was acidic, washed with NaHC03, dried over MgS04, and evaporated to give a white solid (8 mg, 89%) :'H NMR (300 MHz, CDC13) 8 8.78 (d, 1 H), 8.44 (t, 1 H), 7.95 (t, 2 H), 7.69- 7.45 (m, 5 H), 5.30 (br, 2 H), 4.84 (br, 1 H), 4.68 (br, 1 H), 3.78 (br, 2 H), 2.39 (s, 3 H), 2.10 (br, 4 H), 1.96 (s, 2 H), 1.57 (br, 10 H), 1.25 (s, 2 H), 0.88 (br, 1 H); API MS m/z = 512 [C3oH37N70+H] +.

Example 82-Preparation of Compound 98 DME (3 mL), tris (dibenzylideneacetone) dipalladium (0.01 g, 0.01 mmol), and PPh3 (0.04 g, 0. 15 mmol) were added to a round bottom flask equipped with condensor and maintained under an argon atmosphere. Iodide 11 (0.13 g, 0.26 mmol), and 3-chlorobenzene boronic acid (0.15 g, 0.93 mmol) was dissolved in 2M Na2C03 (0.6 mL) and DME (1 mL). This was then added to the reaction mixture and refluxed for 19.5 h then stirred at room temperature for 30 h. The reaction mixture was then diluted with H20, extracted with CH2C12, washed with brine, dried over Na2S04, filtered, and evaporated. The reaction mixture was purified by column chromatography (3 x) and evaporated. The product was triturated in hexanes, filtered, and dried in vacuo for 1 h to give a white solid (16 mg) :'H NMR (300 MHz, CDC13) 8 7.56-7. 38 (m, 9 H), 6.01 (br, 1 H), 4.80 (d, 2 H), 4.71-4. 62 (m, 1 H), 3.77 (br, 2 H), 2.73 (br, 1 H), 2.19-2. 04 (m, 1 H), 1.94-1. 85 (m, 1 H), 1.51 (d, 6 H), 1.24 (d, 5 H), 0.91-1. 76 (m, 3 H); API MS m/z = 490 [C27H32ClN7+H] +.

Example 83-Preparation of Compound 99 A stock solution of acetic anhydride was made by mixing CH2Cl2 (16 mL), pyridine (4 mL), and Ac20 (0.16 mL). To this solution (1.5 mL) was added compound 98 (0.01 g, 0.02 mmol), followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 2 h. The reaction mixture was diluted with CH2Cl2, washed with 2N HCl until the aqueous layer was acidic, washed with NaHC03, dried over MgS04, filtered, and evaporated to give a white solid (0.01 g, 83%) :'H NMR (300 MHz, CDCl3) 6 7.65-7. 35 (m, 8 H), 7.26-7. 14 (m, 1 H), 5.23 (br, 1 H), 4.66 (br, 1 H), 3.78 (br, 2 H), 2.18-2. 00 (m, 4 H), 1.94 (s, 3 H), 1.54 (d, 6 H), 1.24 (s, 5 H), 0.94-0. 69 (m, 3 H); API MS m/z = 532 [C29H34Cl N70+H] +.

Example 84-Preparation of Compound 100 A stock solution of acetic anhydride was made by mixing CH2C12 (16 mL), pyridine (4 mL), and Ac20 (0.16 mL). To compound 14 (0.02 g, 0.03 mmol) was added this solution (2 mL), followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 3 h. The reaction mixture was diluted with CH2C12, washed with 2N HCl until the aqueous layer was acidic, washed with NaHC03, filtered, and evaporated to give a white solid (8 mg, 44%) :'H NMR (300 MHz, CDC13) 8 7.41-7. 32 (m, 7 H), 7.26-7. 14 (m, 1 H), 5.96 (br, 1 H), 5.23 (d, 1 H), 4.84 (br, 2 H), 4.69-4. 54 (m, 1 H), 3.75 (br, 1 H), 2.21-2. 12 (m, 1 H), 2.09-1. 96 (m, 1 H), 1.97 (s, 3 H), 1.54 (d, 6 H), 1.36-1. 15 (m, 5 H), 0.85 (br, 3 H); API MS m/z = 550 [C29H33ClFN7O+H] +.

Example 85-Preparation of Compound 101 DME (3 mL), tris (dibenzylideneacetone) dipalladium (0.01 g, 0.01 mmol), and PPh3 (0.04 g, 0. 15 mmol) were added to a round bottomed flask equipped with a condensor and maintained under an argon atmosphere. Compound 10 (0.13 g, 0.26 mmol) and 4-fluorobenzene boronic acid (0.13 g, 0.95 mmol) was dissolved in 2M Na2C03 (0.6 mL) and DME (1 mL). This was then added to the reaction mixture and refluxed for 19 h then stirred at room temperature for 72 h. The reaction mixture was then diluted with H20, extracted with CH2CI2, washed with brine, dried over

Na2S04, filtered, and evaporated. The reaction mixture was purified by column chromatography on silica gel to give a white solid (17 mg, 14%) :'H NMR (300 MHz, CDCl3) 8 7.56-7. 38 (m, 8 H), 7.11 (t, 1 H), 5.81 (br, 1 H), 4.81 (d, 2 H), 4.69- 4.57 (m, 1 H), 3.78 (br, 2 H), 2.69 (br, 1 H), 2.12 (br, 1 H), 1.88 (br, 1 H), 1.54 (d, 6 H), 1.33-1. 12 (m, 5 H), 0.85 (br, 3 H); API MS m/z = 474 [C27H32FN7+H] +.

Example 86-Preparation of Compound 102 A stock solution of acetic anhydride was made by mixing CH2Cl2 (16 mL), pyridine (4 mL), and Ac20 (0.16 mL). To the solution (1.4 mL) was added compound 101 (0.01 g, 0.02 mmol), followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 2.5 h. The reaction mixture was diluted with CH2C12, washed with 2N HCl until the aqueous layer was acidic, and washed with saturated NaHC03. The organic layer was dried over MgS04 and evaporated to give a product (3 mg). The NaHC03 layer was further extracted with EtOAc (2 x), the organic layers were combined, dried over MgS04, evaporated to give product 102 (2 mg). The products were combined using EtOAc, evaporated, and dried in vacuo for 15 h to give product 102 (5 mg, 50%) :'H NMR (300 MHz, CDCl3) 8 7.71-7. 08 (m, 9 H), 5.29 (br, 2 H), 4.84 (br, 1 H), 4.66 (br, 1 H), 3.78 (br, 2 H), 2.09 (br, 4 H), 1.97 (s, 1 H), 1.57 (br, 3 H), 1.24 (d, 6 H), 0.87 (br, 5 H); API MS milz = 516 [C29H34FN70+H] +.

Example 87-Preparation of Compound 103 Compound 30 (0.10 g, 0.27 mmol) and trans-1, 4-diaminocyclohexane (0.48 g, 4.2 mmol) were combined with EtOH (2 mL) in a sealed tube and heated at 190 °C for 24 h, and then stirred at room temperature for 46 h. The reaction mixture was purified by column chromatography and dried in vacuo to give 103 as a white solid (0.10 g, 81%) :'H NMR (300 MHz, CDC13) 8 8.83 (d, 1 H), 8.58 (t, 1 H), 7.87- 7.83 (m, 1 H), 7.55-7. 47 (m, 5 H), 7.38-7. 33 (m, 1 H), 5.96 (br, 1 H), 4.82 (d, 2 H), 4.68-4. 59 (m, 1 H), 3.75 (br, 2 H), 2.69 (br, 1 H), 2.14 (d, 2 H), 1.86 (d, 2 H), 1.54 (d, 6 H), 1.31-1. 18 (m, 5 H); API MS m/z = 457 [C26H32N8+H] +.

Example 88-Preparation of Compound 104 A stock solution of acetic anhydride was made by mixing CH2C12 (16 mL), pyridine (4 mL), and Ac2O (0.16 mL). To the solution (3.1 mL) was added compound 103 (0.02 g, 0.04 mmol), followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 2.5 h. The reaction mixture was evaporated, dried in vacuo for 19 h, and purified by column chromatography to give a white solid (0.02 g) :'H NMR (300 MHz, CDCl3) 8 8.83 (d, 1 H), 8.59 (t, 1 H), 7.85 (d, 1 H), 7.55-7. 47 (m, 5 H), 7.38-7. 34 (m, 1 H), 5.89 (br, 1 H), 5.25 (d, 2 H), 4.85 (br, 1 H), 4.66-4. 61 (m, 1 H), 3.77 (br, 2 H), 2.15 (br, 2 H), 2.05 (br, 2 H), 1.97 (s, 2 H), 1.54 (d, 6 H), 1.33-1. 25 (m, 5 H), 0.88 (br, 1 H); API MS m/z = 499 [C28H34NgO+H] .

Example 89-Preparation of Compound 106 Compound 72 (0.30 g, 0.80 mmol) and compound 105 (1.15 g, 6.50 mmol) (Gardiner, J. M. , et al. Tetrahedron, 42 (11): 515 (1995), which is hereby incorporated by reference, were combined with EtOH (7 mL) and allowed to reflux for 23 h. Triethylamine (1 mL) was added and the reaction was refluxed further for another 21 h. The reaction mixture was then transferred to a sealed tube and EtOH (3 mL) was added. The reaction mixture was heated further at 100 °C for 3 h. The mixture was purified by column chromatography to give 105 (0.13 g) : H NMR (300 MHz, CDCl3) 8 7.57-7. 26 (m, 10 H) 5.58 (br, 1 H), 5.10 (br, 1 H), 4.83 (br, 1 H), 4.69-4. 62 (m, 2 H), 3.36-2. 91 (m, 5 H), 2.82-2. 65 (m, 2 H), 1.53 (d, 2 H), 1.44 (s, 9 H), 1.25 (d, 1 H), 1.13 (d, 3 H); CI MS m/z = 416 [C29H39N7O-Boc+H] +.

Example 90-Preparation of Compound 107 To compound 106 (0.10 g, 0.18 mmol) was added Et2O (2 mL), CH2Cl2 (1 mL) and MeOH (1 mL). During 16 h HCl/ether (1M, 5 mL) was added while stirring. The resulting precipitate was collected by filtration and dried in vacuo for 30 min to provide 106 as an off-white solid (60 mg, 81%) :'H NMR (300 MHz, DMSO) 8 8.48 (br, 2 H), 8.15 (br, 1 H), 7.67-7. 27 (m, 10 H), 4.79 (br, 1 H), 3.60-3. 42

(m, 3 H), 3.18-3. 06 (m, 2 H), 3.03-2. 91 (m, 2 H), 1.52 (d, 2 H), 1.27 (d, 6 H); CI MS =416 [C24H29N7+H] .

Example 91-Preparation of Compound 108 A stock solution of acetic anhydride was made by mixing CHzClz (16 mL), pyridine (4 mL), and Ac20 (0.16 mL). To this solution (5.6 mL) was added compound 107 (0.04 g, 0.09 mmol), followed by DMAP (few crystals). The reaction mixture was allowed to stir at room temperature for 2 h. The reaction mixture was diluted with CH2Cl2, washed with 2N HCl until acidic, the organic layer was washed with NaHC03, dried over MgS04, filtered, and evaporated to give a white solid (16 mg). The product was purified by column chromatography to provide 108 as a white solid (0.01 g, 18%) :'H NMR (300 MHz, CDC13) 8 7.58-7. 43 (m, 10 H), 6.60 (br, 1 H), 5.91 (br, 1 H), 5.04 (t, 1 H), 4.84 (br, 2 H), 4.72-4. 59 (m, 1 H), 4.10-4. 02 (m, 1 H), 3.59-3. 47 (m, 2 H), 1.80 (s, 3 H), 1.57 (d, 6 H), 1.19 (d, 3 H); CI MS m/z = 458 [C26H3lN70+H] Example 92-Preparation of Compound 109 Compound 61 (1.0 g, 2.18 mmol), 3-chlorophenylboronic acid (1.3 g, 8.16 mmol), PPh3 (0.3 g, 1.26 mmol), 2M Na2C03 (5.0 mL), and DME (54 mL) were added to a three-necked round-bottomed flask. The mixture was degassed with argon and heated to reflux for 40 min, cooled to room temperature, and then Pd2 (dba) 3 (0.08 g, 0.08 mmol) was added. The reaction mixture was heated at reflux for 7 h. 3- Chlorophenylboronic acid (0.6 g) and Pd2 (dba) 3 (0.08 g) was then added and reflux continued for 12 h. The reaction mixture was cooled to room temperature, diluted with H20 (50 mL), and extracted with CH2Cl2 (3 x 50 mL). The combined organic phases were washed with H20 (50 mL), brine (50 mL), dried over Na2S04, filtered, and concentrated in vacuo. The residue was purified by silica gel chromatography and concentrated in vacuo to obtain compound 109 (950 mg, 89%): mp 178-181 °C ; 'H NMR (500 MHz, CDCl3) 6 7.56 (s, 1 H), 7.42-7. 54 (m, 6 H), 7.26-7. 35 (m, 2 H), 6.08 (bs, 1 H), 4.81 (bs, 2 H), 4.59-4. 64 (m, 2 H), 3.75-3. 81 (m, 1 H), 2.65-2. 72 (m, 1

H), 2.12 (d, 2 H), 1.88 (d, 2 H), 1.53 (d, 6 H), 1.18-1. 27 (m, 4 H); CI MS m/z = 490 [C27H32C1N7+H] +.

Example 93-Preparation of Compound 110 Compound 109 (500 mg, 1.02 mmol) was dissolved in anhydrous CH2C12 (30 mL), cooled with an ice-water bath, followed by the addition of DMAP (12.2 mg, 0.1 mmol), pyridine (124 pL, 1.53 mmol), and Ac20 (106 1L, 1.12 mmol).

The reaction mixture was stirred for 30 min at 0 °C an ice-water bath then stirred another 2 h at room temperature. The reaction mixture was then concentrated in vacuo and the residue was purified by column chromatography on silica gel. After removal of the solvent, the residue was dried in vacuo to give 110 (339 mg, 63%): mp 198-200 °C ; IH NMR (500 MHz, CDC13) 8 7.57 (s, 1 H), 7.39-7. 53 (m, 6 H), 7.27-7. 37 (m, 2 H), 6.31 (bs, 1 H), 5.28 (d, 1 H), 4.78 (bs, 2 H), 4.70 (d, 1 H), 4.58- 4.67 (m, 1 H), 3.72-3. 83 (m, 1 H), 2.18 (d, 2 H), 2.00 (d, 2 H), 1.90 (s, 3 H), 1.51 (d, 6 H), 1.18-1. 31 (m, 4 H) ; CI MS m/z= 532 [C29H34ClN70+H] +.

Example 94-Preparation of Compound 111 Compound 61 (1.0 g, 2.18 mmol), 2-thiopheneboronic acid (1.0 g, 8.16 mmol), PPh3 (0.3 g, 1.26 mmol), 2M Na2C03 (5. 0 mL), Pd2 (dba) 3 (0. 08 g, 0.08 mmol), and DME (54 mL) were added to a round-bottomed flask and purged with argon. The reaction mixture was heated at reflux for 24 h. 2-Thiopheneboronic acid (0.5 g), Pd2 (dba) 3 (0. 1 g), and 2M Na2C03 (2 mL) were added and heated to reflux for another 24 h. The reaction mixture was cooled to room temperature, diluted with H20 (50 mL) and extracted with CH2Cl2 (3 x 50 mL). The organic phase was washed with H20 (50 mL) and brine (50 mL), dried over Na2S04, filtered, and concentrated in vacuo. The residue was repeatedly chromatographed on silica gel to obtain 111 (574 mg, 59%): mp 109-110 °C ; tH NMR (500 MHz, CDC13) 8 7.56 (d, 2 H), 7.54 (s, 1 H), 7.46 (d, 2 H), 7.24-7. 37 (m, 2 H), 7.06 (t, 1 H), 6.04 (bs, 1 H), 4.78 (bs, 2 H), 4.59-4. 69 (m, 2 H), 3.75-3. 81 (m, 1 H), 2.67-2. 74 (m, 1 H), 2.14 (d, 2 H), 1.87 (d, 2 H), 1.52 (d, 6 H), 1.17-1. 29 (m, 4 H); CI MS m/z = 462 [C25H3, N7S+H] +.

Example 95-Preparation of Compound 112 Compound 111 (410.0 mg, 0.89 mmol) was dissolved in anhydrous CH2C12 (30 mL) and purged with N2 and cooled with an ice-water bath. Pyridine (108 mg, 1.34 mmol) and DMAP (10.9 mg, 0.09 mmol) followed by Ac20 (92 uL, 0.98 mmol) were added slowly. The reaction mixture was stirred for 30 min in an ice- water bath followed by 2 h at room temperature. The reaction mixture was concentrated in vacuo. The residue was chromatographed on silica gel to give 112 (325 mg, 73%): mp 237-244 °C ;'H NMR (500 MHz, CDCl3) 8 7.54 (d, 2 H), 7.50 (s, 1 H), 7.36 (d, 2 H), 7.24-7. 37 (m, 2 H), 7.08 (t, 1 H), 6.06 (bs, 1 H), 5.34 (s, 1 H), 4.78 (bs, 2 H), 4.58-4. 70 (m, 2 H), 3.78 (bs, 2 H), 2.17 (d, 2 H), 2.04 (d, 2 H), 1.96 (s, 3 H), 1.56 (d, 6 H), 1.18-1. 32 (m, 4 H); CI MS m/z= 504 [C27H33N70S+H] +.

Example 96-Preparation of Compound 113 Compound 12 (600 mg, 1.30 mmol) was dissolved in anhydrous CH2Cl2 (40 mL), purged with N2, and cooled to 0 °C followed by an addition of DMAP (15.9 mg, 0.13 mmol), pyridine (165.3 mg, 1.95 mmol), and Ac20 (135 mg, 1.43 mmol). The mixture was stirred 30 min at 0 °C then 2 h at room temperature.

The reaction mixture was concentrated in vacuo. The residue was chromatographed on silica gel to give 113 (495 mg, 76%): mp 248-253 °C ;'H NMR (500 MHz, CDCl3) 8 7.54 (d, 2 H), 7.46 (s, 1 H), 7.35-7. 41 (m, 5 H), 6.13 (bs, 1 H), 5.28 (d, 1 H), 4.78 (br, 2 H), 4.61-4. 63 (m, 2 H), 3.75 (bs, 2 H), 2.14 (d, 2 H), 1.97 (d, 2 H), 1.95 (s, 3 H), 1.52 (d, 6 H), 1.15-1. 37 (m, 4 H); CI MS mlz = 504 [C27H33N70S+H] +.

Example 97-Preparation of Compound 114 To compound 61 (1.0 g, 2.18 mmol) was added PPh3 (330 mg, 1.26 mmol), 2M Na2C03 (5 mL), DME (54 mL), and 4-carboxyphenylboronic acid (1.0 g, 6.03 mmol). The mixture was purged with N2 for 45 min then Pd2 (dba) 3 (366 mg, 0.4 mmol) was added and the mixture was heated at reflux for 3 d. The reaction mixture was diluted with H20 (100 mL). The aqueous layer was separated, and washed with CH2Cl2 (3 x 40 mL). The aqueous layer was adjusted the pH to 5.8 by using IN HCI.

Some precipitate appeared. The mixture was stored in a freezer overnight. The

precipitate was collected and dried to obtain 114 (450 mg, 41%): mp 246-249 °C <BR> <BR> <BR> (dec. ) ;'H NMR (500 MHz, CD30D+NaOD) 8 7.84 (s, 2 H), 7.64 (s, 1 H), 7.54-7. 63 (m, 4 H), 7.39 (s, 2 H), 6.08 (bs, 1 H), 4.85 (bs, 2 H), 4.73 (s, 1 H), 3.76 (m, 1 H), 2.74 (m, 1 H), 1.99 (s, 2 H), 1.88 (s, 2 H), 1.63 (d, 6 H), 1.21-1. 36 (m, 4 H); CI MS m/z= 500 [C28H33N702+H].

Example 98-Preparation of Compound 115 To a cooled MeOH (20 mL) solution was slowly added TMSCI (253 uL, 2.0 mmol). The solution was stirred 20 min, followed by the addition of 114 (100 mg, 0.2 mmol). The reaction mixture was stirred at room temperature for 24 h. The reaction mixture was cooled with an ice-water bath then Et3N (557 mL) was added.

The mixture was concentrated in vacuo, to provide the crude product, which was washed with water (2 x 20 mL). The residue was purified by chromatography on a silica gel. After removal of the solvent and drying in vacuo, the residue was dissolved in MeOH (5 mL), followed by the addition of ether (10 mL). The precipitate was collected and dried to provide 115 (75 mg, 73%): mpl94-197 °C ;'H NMR (500 MHz, CD30D) 8 8.07 (d, 2 H), 7.80 (s, 1 H), 7.72 (d, 2 H), 7.63 (d, 2 H), 7.46 (d, 2 H), 4.63-4. 79 (m, 1 H), 3.91 (s, 3 H), 3.65-3. 77 (m, 1 H), 3.07 (bs, 1 H), 2.12 (d, 2 H), 2.01 (d, 2 H), 1.55 (d, 6 H), 1.29-1. 49 (m, 4 H); API MS m/z= 514 [C29H3sN702+H] +.

Example 99-Preparation of Compound 117 To a suspension of compound 114 (250 mg, 0.50 mmol), pyridine (60 1L, 0.75 mmol), and DMAP (6.1 mg, 0.05 mmol) in H20-dioxane (2: 1,40 mL) was added Ac2O (57 pL, 0.60 mmol). After stirring 4 h at room temperature, K2CO3 (100 mg) was added followed by additional Ac20 (100 1L). The reaction mixture was stirred 2 h at room temperature. Water (50 mL) was added and the pH was adjusted to 5. The precipitate was collected, washed with water and ether, and dried in vacuo.

The precipitate (200 mg) was added to a solution of TMSCI (500 pL, 3.94 mmol) in MeOH (25 mL). The reaction mixture was stirred 24 h at room temperature. The mixture was concentrated in vacuo. The product was purified by silical gel chromatography to provide 117 (145 mg, 52%): mp 247-250 °C ;'H NMR (500 MHz,

CDCl3) 8 8.09 (d, 2 H), 7.64 (d, 2 H), 7.58 (d, 2 H), 7.49 (s, 1 H), 7.45 (d, 2 H), 5.91 (bs, 1 H), 5.18 (d, 1 H), 4.83 (bs, 2 H), 4.61-4. 68 (m, 2 H), 3.93 (s, 3 H), 3.67-3. 78 (m, 2 H), 3.07 (bs, 1 H), 2.16 (d, 2 H), 2.02 (d, 2 H), 1.95 (s, 3 H), 1.54 (d, 6 H), 1.23- 1.32 (m, 4 H); API MS m/z = 556 [C31H37N703+H] +.

Example 100-Preparation of Compound 116 To a solution of compound 117 (90 mg, 0.16 mmol) in MeOH-H2O (6: 1,23 mL) was added KOH (11 mg, 0.19 mmol) in 5 mL MeOH. The reaction mixture was refluxed for 24 h. After removal of the solvent the residue was dissolved in 15 mL of water and washed with CH2C12. The aqueous layer was separated and adjusted pH to 4.5 by using IN HC1. The precipitate was collected and dried to obtain 116 (60 mg, 68%): mp 344-347 °C ;'H NMR (500 MHz, DMSO-d6) 8 11.21 (bs, 1 H), 8.14 (d, 2 H), 7.64-7. 88 (m, 6 H), 7.47 (d, 2 H), 6.06 (bs, 1 H), 5.18 (d, 1 H), 4.85 (bs, 2 H), 4.51-4. 66 (m, 1 H), 3.62 (bs, 1 H), 3.46 (bs, 1 H), 1.89 (bs, 2 H), 1.77 (bs, 5 H), 1.95 (s, 3 H), 1.47 (d, 6 H), 1.23-1. 36 (m, 4 H); API MS m/z = 542 [C3oH35N703+H] +.

Example 101-Preparation of Compound 118 Compound 61 (1.0 g, 2.18 mmol), 3-carboxyphenylboronic acid (1.0 g, 6.03 mmol), 2N Na2C03 (5 mL), and DME/EtOH (50 mL) were mixed together and degassed with N2 for 1 h. Pd2 (dba) 3 (366.0 mg, 0.4 mmol) and PPh3 (330.0 mg, 1.26 mmol) were added and the reaction mixture was heated to reflux for 48 h. The reaction mixture was cooled to room temperature, diluted with CH2C12 (50 mL), and extracted with aqueous 5% Na2C03 (3 x 30 mL). The combined washes were extracted with CH2C12 (3 x 30 mL) and ether (40 mL). The aqueous phase was neutralized to a pH of 5.8 using IN HCl and kept in a freezer for 1 h. The precipitate was collected, suspended in MeOH (30 mL) and the insolubles were removed by filtration. To the MeOH solution was added ether (20 mL) to precipitate the product.

The white solid was collected and dried in vacuo to offer 118 (65 mg, 6%): mp 205- 208 °C ; lH NMR (500 MHz, CD30D+NaOD) 8 8.17 (s, 1 H), 7.88 (d, 1 H), 7.80 (s, 1 H), 7.56-7. 63 (m, 3 H), 7.35-7. 41 (m, 3 H), 6.08 (bs, 1 H), 4.80 (bs, 2 H), 4.59-4. 75

(m, 1 H), 3.72-3. 82 (m, 1 H), 2.89-3. 01 (m, 1 H), 1.90-1. 99 (m, 4 H), 1.51 (d, 6 H), 1.29-1. 40 (m, 2 H), 1.12-1. 23 (m, 2 H); API MS m/z = 500 [C28H33N702+H] +.

Example 102-Preparation of Compound 119 3-Thiopheneboronic acid (4.5 g, 35.2 mmol) and 6-chloronicotinamide (5.0 g, 32.0 mmol) were dissolved in DMA (150 mL), followed by the addition of 2N Na2C03 (23 mL). N2 gas was passed through the mixture for 1 h. Pd (PPh3) 4 (0.74 g, 0.64 mmol) was added and the reaction mixture was heated to reflux for 24 h. The reaction mixture was cooled to room temperature and poured into an ice-water (1 L) and stirred for 10 min. The precipitate was collected and washed with acetone. The collected solid was suspended in EtOAc (150 mL) and heated to reflux for 5 min.

The solid was filtered and collected. After drying in vacuo, 119 (4.5 g, 69%) was obtained :'H NMR (500 MHz, DMSO-d6) 6 9.08 (s, 1 H), 8.34 (s, 1 H), 8.28 (d, 1 H), 8.20 (bs, 1 H), 7.99 (d, 1 H), 7.81 (d, 1 H), 7.71 (d, 1 H), 7.60 (bs, 1 H).

Example 103-Preparation of Compound 120 To compound 119 (4.08 g, 20.0 mmol) suspended in THF (50 mL), was added 1 M BH3-THF (164 mL). The mixture was heated to reflux for 9 h. The mixture was cooled with an ice-water bath and adjusted to a pH of 1-2, and stirred for 1 h at room temperature. The pH was adjusted to 9-10 (2N NaOH) and extracted with EtOAc (3 x 50 mL). The combined organic phases were washed with H20 (50 mL), brine (50 mL), and dried over Na2S04. After filtration and removal of the solvent, the residue was dissolved in EtOH (50 mL), followed by the addition of 1 M HCl/ether (20 mL). The mixture was concentrated to dryness to provide 120 (2.03 g, 45%) : IH NMR (500 MHz, CD30D) 8 8.93 (s, 1 H), 8.61 (d, 1 H), 8.51 (s, 1 H), 8.43 (d, 1 H), 7.81 (d, 1 H), 7.70 (d, 1 H), 3.30 (t, 2 H).

Example 104-Preparation of Compound 121 Compound 120 (2 g, 8.82 mmol), 2,6-dichloropurine (1.5 g, 8.01 mmol), EtOH (50 mL), and (i-Pr) 2NEt (3.8 mL, 22 mmol) were heated at reflux for 16 h. The reaction mixture was then cooled with an ice-water bath. The precipitate was

collected and washed with EtOH, H20, and ether. The precipitate was dried in vacuo to obtain 121 (0.84 g, 31%) :'H NMR (500 MHz, DMSO-d6) 8 11. 02 (bs, 1 H), 8.76 (bs, 1 H), 8.63 (s, 1 H), 8.07 (bs, 2 H), 7.79 (bs, 2 H), 7.71 (d, 1 H), 7.64 (d, 1 H), 4.68 (bs, 2 H).

Example 105-Preparation of Compound 122 Compound 121 (950 mg, 2.77 mmol) was dissolved in DMSO (50 mL), and then K2C03 (2.07 g, 15. 0 mmol) was added, followed by the addition of 2- iodopropane (830 sL, 8.31 mmol). The reaction mixture then was stirred at room temperature overnight. The reaction mixture was poured into an ice-water bath (400 mL), stirred for 10 min, and extracted with EtOAc (4 x 50 mL). The combined organic phases were washed with H2O (40 mL), brine (40 mL), and dried over MgS04. After filtration and removal of the solvent, the residue was dissolved in hot EtOAc (40 mL), followed by the addition of hexanes (80 mL). The precipitate was collected and dried in vacuo to obtain 122 (798 mg, 90%) :'H NMR (500 MHz, CDC13) 8 8.64 (s, 1 H), 7.83 (s, 1 H), 7.70-7. 79 (m, 2 H), 7.60 (d, 1 H), 7.55 (d, 1 H), 7.36 (d, 1 H), 6.11 (bs, 1 H), 4.77-4. 96 (m, 3 H), 1.53 (d, 6 H).

Example 106-Preparation of Compound 123 Compound 122 (780.0 mg, 2.03 mmol), trans-1, 4-diaminocyclohexane (2.3 g, 20.3 mmol), and EtOH (4 mL) were heated in a sealed tube to 150 °C for 20 h.

The reaction mixture was poured into ice-water (150 mL) and stirred for 10 min. The resulting precipitate was washed with H20 (2 x 20 mL) and dried. The solid was chromatographed on a silica gel column. After removal of the solvent and drying in vacuo, 123 (765 mg) was obtained: mp 78-81 °C ;'H NMR (500 MHz, CDC13) 8 8.63 (s, 1 H), 7.87 (s, 1 H), 7.72 (d, 1 H), 7.64 (d, 1 H), 7.55 (d, 1 H), 7.04-7. 09 (m, 1 H), 6.92 (s, 1 H), 5.95 (bs, 1 H), 4.64 (bs, 2 H), 4.33-4. 45 (m, 2 H), 3.74-3. 77 (m, 1 H), 2.67-2. 76 (m, 1 H), 2.13 (d, 2 H), 1.90 (d, 2 H), 1.63 (bs, 2 H), 1.54 (d, 6 H), 1.19- 1.30 (m, 4 H) ; 3C NMR (CDC13) 8 159.1, 155. 0,152. 7,151. 3,149. 3,143. 3,142. 3, 136.2, 134.8, 133.4, 126.4, 126.4, 123.5, 120.2, 114. 8,50. 4,50. 3,46. 5,42. 0,35. 7, 32.3, 22.8 ; API MS mlz = 463 [C24H30N8S+H]-

Example 107-Preparation of Compound 124 To an ice-cold solution of compound 123 (420 mg, 0.91 mmol) in CH2Cl2 (20 mL) was added pyridine (110 pL, 1.4 mmol), DMAP (11.0 mg, 0.09 mmol) and Ac20 (94.2 pL, 1 mmol). The reaction mixture was stirred for 30 min at 0 °C, followed by 2 h at room temperature. After removal of the solvent, the residue was chromatographed on a silica gel column. The resulting solid was recrystallized with EtOAc/MeOH and dried in vacuo to give 124 (350 mg, 79%): mp 249-252 °C ; 'H NMR (500 MHz, CDC13) 8 8.61 (s, 1 H), 7.85 (s, 1 H), 7.70 (d, 1 H), 7.62 (d, 1 H), 7.53 (d, 1 H), 7.48 (s, 1 H), 7.38 (d, 1 H), 6.00 (bs, 1 H), 5.25 (d, 1 H), 4.77 (bs, 2 H), 4.53-4. 72 (m, 2 H), 3.68-3. 77 (m, 2 H), 2.10 (d, 2 H), 2.00 (d, 2 H), 1.94 (s, 3 H), 1.52 (d, 6 H), 1.17-1. 28 (m, 4 H) ;'3C NMR (CDC13) 8 169.4, 159. 0, 155. 0, 152. 8,149. 2, 142.8, 142.3, 136. 1,134. 9,133. 4,126. 5,126. 4,123. 5,120. 2,114. 9,50. 1,48. 3,46. 5, 42.2, 32.2, 32.1, 22.8 ; API MS m/z= 505 [C26H32N8OS+H] +.

Example 108-Alternative Preparation of Compound 71 To a solution of 4-phenylbenzoic acid (5.46 g, 27.6 mmol) in methylene chloride (66 mL) was added 2 drops of DMF and oxalyl chloride (2.80 mL, 30.3 mmol). The reaction mixture was stirred overnight and added to a stirred solution of ice and ammonium hydroxide. The resulting precipitate was filtered, washed with methylene chloride, and triturated with water. The product was collected by filtration and dried in vacuo to yield 4-phenylbenzamide (3.88 g, 71%).

Under a nitrogen atmosphere, 4-phenylbenzamide (2.01 g, 10.2 mmol) was dissolved in THF (50 mL) and heated to reflux. To the mixture was added dropwise 1 M borane in THF (80.0 mL, 80.0 mmol). After refluxing for 18 h, the reaction mixture was cooled to room temperature and treated with 1 N HC1 (40 mL).

The solution was made basic via addition of 3 N NaOH (60 mL) and extracted with ethyl acetate (3 x 370 mL). The extract was washed with brine and dried over sodium sulfate. Concentration yielded 4-phenylbenzyl amine as a white solid (1.73 g, 93%).

4-Phenylbenzyl amine (1.73 g, 10.1 mmol) and 2,6-dichloropurine (1.94 g, 10.1 mmol) was dissolved in water (110 mL). To the solution was added N, N-

diisopropylethylamine (3.54 mL, 20.2 mmol). The reaction mixture was heated to reflux for 5 h and cooled to room temperature. A precipitate was collected by filtration. The solid was washed with water and ethanol and dried to yield 71 (2.35 g, 69%) :'H NMR (300 MHz, (CD3) 2SO) 8 8.69 (brs, 1 H), 8.15 (s, 1 H), 7.57-7. 68 (m, 4 H), 7.30-7. 50 (m, 5 H), 4.71 (d, 2 H).

Example 109-Preparation of Compound 126 To a stirred solution of 3-iodobenzylamine (5.00 g, 21.4 mmol) in water (100 mL) was added 2,6-dichloropurine (4.04 g, 21.4 mmol) and N, N diisopropylethylamine (7.47 mL, 42.5 mmol). The mixture was refluxed for 5 h and stored at room temperature overnight. The resulting suspension was filtered. The filter cake was triturated with water (3 x 25 mL) and ethanol (2 x 15 mL) and dried under high vacuum to yield 126 (7.49 g, 91%) : 1H NMR (300 MHz, (CD3) 2SO) 8 8.50-8. 80 (brs, 1 H), 8.17 (s, 1 H), 7.75 (s, 1 H), 7.61 (d, 1 H), 7.37 (d, 1 H), 7.14 (t, 1 H), 5.14 (brs, 1 H), 4.61 (d, 2 H).

Example 110-Preparation of Compound 127 The purine derivative 126 (7.00 g, 18.2 mmol) was dissolved in dimethylsulfoxide (120 mL). To this stirred solution was added potassium carbonate (20.0 g, 145 mmol) and 2-iodopropane (7.28 mL, 72.8 mmol). The reaction mixture was stirred under a nitrogen atmosphere for 20 h before being poured into stirred water (600 mL). After 10 min, the resulting mixture was extracted with ethyl acetate (4 x 95 mL). The combined organic layers were washed with water (25 mL) and brine (3 x 25 mL), dried over sodium sulfate, and concentrated in vacuo. The resulting material was purified by recrystallization from ethyl acetate in hexanes to yield 127 (7.51 g, 97%): mp 147-152 °C ;'H NMR (300 MHz, (CD3) 2SO) 8 8.84 (m, 1 H), 8.31 (s, 1 H), 7.74 (s, 1 H), 7.60 (d, 1 H), 7.36 (d, 1 H), 7.13 (t, 1 H), 4.51-4. 75 (m, 3 H), 1.50 (d, 6 H); API MS m/z = 429 [C, 5Hl5ClIN5+H] +.

Example 111-Preparation of Compound 128 In a sealed tube, 127 (2.57 g, 6.00 mmol), trans-1, 4- diaminocyclohexane (6.85 g, 60.0 mmol), and ethanol (10 mL) were combined. The reaction mixture was heated to 160 °C for 24 h. After cooling to room temperature, the mixture was filtered. The filtrate was concentrated and diluted with ethyl acetate (250 mL). The organic solution was washed with water (250 mL) and saturated sodium bicarbonate solution (2 x 250 mL). The organic layer was dried over sodium sulfate, filtered, and concentrated. A portion of the resulting crude product was purified via silica gel chromatography to yield 128 (181 mg) :'H NMR (300 MHz, (CD3) 2SO) 8 7.87 (brs, 1 H), 7.78 (s, 1 H), 7.70 (s, 1 H), 7.55 (d, 1 H), 7.35 (d, 1 H), 7.09 (t, 1 H), 6.06 (d, 1 H), 4.43-4. 70 (m, 3 H), 3.58 (brs, 1 H), 1.66-1. 94 (m, 4 H), 1.46 (d, 6 H), 1.00-1. 30 (m, 4 H); ESI MS m/z = 506 [C2, H2gIN7+H] +.

Example 112-Preparation of Compound 129 The amine 127 (6.06 g, 6.00 mmol) was dissolved in a mixture of tetrahydofuran (45 mL) and water (15 mL). To this stirred mixture were added sodium bicarbonate (2.02 g, 24.0 mmol) and di-tert-butyldicarbonate (2.90 g, 13.3 mmol). After 3.5 h, the solution was extracted with methylene chloride (3 x 75 mL).

The organic extracts were combined, washed with brine (225 mL), and dried over sodium sulfate. The organic liquid was concentrated and the resulting material was purified via silica gel chromatography (33: 67 to 50: 50 to 60: 40 ethyl acetate/hexanes) to yield 129 (4.94 g, 68%) :'H NMR (300 MHz, CDC13) 8 7.72 (s, 1 H), 7.58 (d, 1 H), 7.46 (s, 1 H), 7.30 (d, 1 H), 7.02 (t, 1 H), 4.50-4. 76 (m, 4 H), 4.40 (m, 1 H), 3.70 (brs, 1 H), 3.43 (brs, 1 H), 1.90-2. 20 (m, 4 H), 1.51 (d, 6 H), 1.45 (s, 9 H), 1.13-1. 35 (m, 4 H); API MS m/z = 607 [C26H36IN702+H] +.

Example 113-Preparation of Compound 130 To a stirred solution of 129 (1.00 g, 1.65 mmol) in ethylene glycol dimethyl ether (40 mL) was added 3-thiopheneboronic acid, triphenylphosphine (250 mg, 0.950 mmol), and 2 M sodium carbonate solution (3.8 mL). The mixture was purged with nitrogen for 10 min and tris (dibenzylideneacetone) dipalladium (0) (64.0

mg, 0.060 mmol) was added. After refluxing overnight under nitrogen, the reaction mixture was cooled to room temperature and diluted with water (100 mL). The resulting solution was extracted with methylene chloride (3 x 50 mL). The organic extracts were combined, washed with brine (30 mL), and dried over sodium sulfate.

The organic liquid was filtered and concentrated in vacuo. Purification via silica gel chromatography (50: 50 ethyl acetate/hexanes) yielded 130 (0.87 g, 94%) : 1H NMR (300 MHz, CDC13) 8 7.23-7. 30 (m, 8 H), 6.02 (brs, 1 H), 4.78 (d, 2 H), 4.53-4. 68 (m, 1 H), 4.32 (m, 1 H), 3.70 (m, 1 H), 3.28-3. 55 (m, 1 H), 1.80-2. 19 (m, 4 H), 1.53 (d, 6 H), 1.45 (s, 9 H), 1.05-1. 32 (m, 4 H); ESI MS m/z = 562 [C3oH39N702S+H] +.

Example 114-Preparation of Compound 131 To a solution of 130 in EtOAc was added 1 N HC1. After concentration of the solution, isolate 131 (658 mg, 91%): mp 211-216 °C ;'H NMR (300 MHz, CD30D) 8 8.40 (brs, 1 H), 7.25-7. 85 (m, 7 H), 4.60-5. 10 (m, 5 H), 3.91 (m, 1 H), 3.16 (m, 1 H), 1.94-2. 33 (m, 4 H), 1.32-1. 79 (m, 10 H); ESI MS m/z = 462 [C25H3, N7S+H] +.

Example 115-Preparation of Compound 132 Following procedures outlined above for acetylation, prepared 132 from 131 (352 mg, 80%): mp 209-211 °C ;'H NMR (300 MHz, CDCI3) 8 7.27-7. 67 (m, 8 H), 6.25 (brs, 1 H), 5.19 (d, 1 H), 4.79 (d, 2 H), 4.52-4. 67 (m, 2 H), 3.69 (m, 2 H), 2.09 (m, 2 H), 1.88-1. 98 (m, 5 H), 1.50 (d, 6 H), 1.06-1. 33 (m, 4 H); ESI MS m/z = 504 [C27H33N7OS+H] +.

Example 116-Preparation of Compound 133 Following general procedures outilined above for Suzuki Coupling reaction, prepared 133 from 129 (0.61 g, 67%) :'H NMR (300 MHz, CDC13) 6 7.30- 7.62 (m, 10 H) 6.00 (brs, 1 H), 4.83 (d, 2 H), 4.54-4. 70 (m, 2 H), 4.37 (m, 1 H), 3.71 (m, 1 H), 3.39 (m, 1 H), 2.12 (m, 2 H), 2.47 (m, 2 H), 1.53 (d, 6 H), 1.45 (s, 9 H), 1.19 (m, 4 H); ESI MS m/z = 556 [C32H41N702+H] +.

Example 117-Preparation of Compound 134 To a stirred solution of 133 (530 mg, 0.950 mmol) in methanol (3 mL) was added 1 N HC1 in diethyl ether (9.50 mL, 9.50 mmol). After stirring for 3.5 h, hydrogen chloride gas was gently bubbled through the solution. After 20 min, the solution was concentrated in vacuo. The resulting material was recrystallized from methanol in ether to afford 134 in quantitative yield :'H NMR (300 MHz, CD30D) 8 8.33 (brs, 1 H), 7.67 (s, 1 H), 7.52-7. 64 (m, 3 H), 7.29-7. 50 (m, 5 H), 4.80-5. 00 (m, 4 H), 4.72 (m, 1 H), 3.87 (m, 1 H), 3.14 (m, 1 H), 2.02-2. 25 (m, 4 H), 1.59 (d, 6 H), 1.47 (m, 4 H); ESI MS m/z = 456 [C27H33N7+H] +.

Example 118-Preparation of Compound 135 Following procedures outlined above for acetylation, prepared 135 from 134 (195 mg, 82%): mp 183-185 °C ;'H NMR (300 MHz, CDC13) 8 7.30-7. 64 (m, 10 H), 6.19 (brs, 1 H), 5.12 (d, 1 H), 4.81 (d, 2 H), 4.60 (m, 2 H), 3.68 (m, 2 H), 2.09 (m, 2 H), 1.86-1. 99 (m, 5 H), 1.51 (d, 5 H), 1.04-1. 32 (m, 4 H); ESI MS m/z = 498 [C29H35N70+H] +.

Example 119-Preparation of Compound 136 To a mixture of 71 (2.00 g, 5.96 mmol) in dimethylsulfoxide (44 mL) was added potassium carbonate (6.56 g, 47.7 mmol) and iodoethane (2.00 mL, 24.4 mmol). After stirring overnight, the reaction mixture was poured into a stirred solution of water (300 mL). After 2 d, it was filtered. The filtrate was extracted with ethyl acetate (2 x 180 mL). The organic extracts were combined and washed with brine (150 mL). The organic layer was dried over magnesium sulfate. Concentration afforded 136 (1.90 g, 88%).

Example 120-Preparation of Compound 137 In a sealed tube, 136 (0.60 g, 1.66 mmol), trans-1, 4- diaminocyclohexane (1.93 g, 16.8 mmol), and potassium iodide (10 mg), were dissolved in ethanol (18 mL). The mixture was heated to 160 °C. After 4 d, the mixture was cooled to room temperature and filtered. The filtrate was dissolved in

ethyl acetate and washed with water (2 x 100 mL) and brine (100 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated. The material was purified by silica gel chromatography and recrystallization from ethanol in hexanes (1: 10) to yield 137 (886 mg, 41%) : mp 175-182 °C ;'H NMR (500 MHz, CD30D) 8 7.30-7. 75 (m, 10 H), 4.55-4. 95 (m, 4 H), 4.10 (q, 2 H), 3.78 (m, 1 H), 2.99 (m, 1 H), 2.11 (d, 2 H), 1.99 (d, 2 H), 1.44 (m, 3 H), 1.31 (m 4 H); ESI MS m/z = 442 [C26H3lN7+H]- Example 121-Preparation of Compound 138 Compound 71 (2.02 g, 6.02 mmol), iodomethane (1.50 mL, 24.4 mmol), and potassium carbonate were dissolved in dimethylsulfoxide (44 mL) and stirred overnight. The reaction mixture was poured into 150 mL of stirring water. The organic and aqueous layers were separated. The organic layer was washed with brine (3 x 100 mL) and dried over magnesium sulfate. The solids were removed by filtration and the solution was concentrated in vacuo to afford 138 (1.93 g, 93%).

Example 122-Preparation of Compound 139 In a sealed tube, 138 (1.80 g, 5.15 mmol) and trans-1, 4- diaminocyclohexane (5.90 g, 51.7 mmol) were dissolved in ethanol (85 mL). The mixture was heated to 140 °C. After heating overnight, the reaction mixture was cooled to room temperature and concentrated. The resulting solid was dissolved in ethyl acetate (160 mL) and washed with water (160 mL) and brine (2 x 100 mL). The organic layer was dried over magnesium sulfate and filtered. Concentration afforded a solid, which was purified by silica gel chromatography and recrystallization from ethanol in hexanes (1: 20) and ethyl acetate in hexanes to yield 139 (850 mg, 38%): mp 182-184 °C ;'H NMR (500 MHz, CD30D) 8 7.25-7. 70 (m, 10 H), 4.64-4. 90 (m, 4 H), 3.75 (m, 1 H), 3.65 (s, 3 H), 2.68 (m, 1 H), 2.05 (m, 2 H), 1.88 (m, 2 H), 1.25 (m, 4 H); ESI MS m/z = 428 [C25H29N7+H] +.

Example 123-Preparation of Compound 140 The HCI salt of 139 (86.7 mg, 0.162 mmol) was suspended in methylene chloride (20 mL). The suspension was immersed in an ice bath while triethylamine (0.16 mL, 1.12 mmol) and a catalytic amount of DMAP were added.

Acetyl chloride (0.04 mL, 0.560 mmol) was added to the mixture. The reaction was quenched by the addition of 5% aqueous NaHC03 solution (50 mL). The aqueous layer was extracted with methylene chloride (2 x 50 mL). The extracts were washed with brine (100 mL), dried over magnesium sulfate, filtered, and concentrated. The resulting solid material was dried in vacuo. Purification by silica gel chromatography and recrystallization from ethyl acetate in hexanes (5: 60) afforded 140 (6.4 mg, 8%): 'H NMR (300 MHz, CD30D) 8 7.50-7. 68 (m, 5 H), 7.25-7. 47 (m, 5 H), 5.22 (m, 3 H), 4.20-5. 05 (m, 2 H), 3.75 (m, 1 H), 3.65 (s, 3 H), 3.35-3. 47 (m, 1 H), 2.78 (m, 1 H), 1.95-2. 16 (m, 2 H), 1.80-1. 95 (m, 2 H), 1.17-1. 40 (m, 4 H).

Example 124-Preparation of Compound 144 Compound 71 (2.03 g, 5.96 mmol), 1-iodopropane (2.25 mL, 24.4 mmol), and potassium carbonate (6.61 g, 47.7 mmol) were dissolved in dimethylsulfoxide (44 mL) and allowed to stir overnight. The reaction mixture was added to 300 mL of stirring water and stirred for 2 d. The resulting precipitate was collected by filtration and dried in vacuo to afford 141 (2.07 g, 92%).

Example 125-Preparation of Compound 142 In a sealed tube, 141 (1.82 g, 4.81 mmol) and trans-1, 4- diaminocyclohexane (5.67 g, 49.7 mmol) were dissolved in ethanol (53 mL). The mixture was heated to 140 °C for 3 d. After cooling to room temperature, the reaction mixture was concentrated and dissolved in ethyl acetate (100 mL). This solution was washed with water (100 mL) and brine (2 x 100 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated. The product was purified by silica gel chromatography and recrystallizations from ethanol in hexanes (1: 20) to yield 142 (523 mg, 24%): mp 133-138 °C ;'H NMR (500 MHz, CD30D) 8 7.25-7. 70 (m, 10 H), 4.65-4. 85 (m, 4 H), 4.02 (t, 2 H), 3.76 (m, 1 H), 2.85 (m, 1 H), 2.08 (d, 2 H), 1.94

(d, 2 H), 1.86 (q, 2 H), 1.20-1. 42 (m, 4 H), 0.93 (t, 3 H); ESI MS m/z = 456 [C27H33N7+H] +.

Example 126-Preparation of Compound 143 Compound 71 (2.01 g, 5.98 mmol), iodocyclopentane (2.80 mL, 24.2 mmol), and potassium carbonate (6.75 g, 48.9 mmol) were dissolved in dimethylsulfoxide (44 mL) and allowed to stir under nitrogen overnight. The reaction mixture was added to 150 mL of stirring water and diluted with 150 mL ethyl acetate.

The organic and aqueous phases were separated. The organic phase was washed with brine (3 x 100 mL) and dried over magnesium sulfate. After filtering, the organic liquid was concentrated and the resulting solid was dried in vacuo to afford 143 (1.29 g, 55%).

Example 127-Preparation of Compound 144 In a sealed tube, 143 (304 mg, 0.749 mmol) and trans-1, 4- diaminocyclohexane (891 mg, 7.80 mmol) were dissolved in ethanol (10 mL). The mixture was heated to 140 °C for 4 d. After cooling to room temperature, the reaction mixture was diluted with ethyl acetate (160 mL). This solution was washed with water (160 mL) and brine (2 x 100 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated. The material was purified by silica gel chromatography and recrystallizations from ethanol in hexanes (1: 20) to yield 144 (299 mg, 19%): mp 144-146 °C ; tH NMR (500 MHz, CD30D) 8 7.28-7. 75 (m, 10 H), 4.68-4. 85 (m, 4 H), 3.75 (m, 1 H), 2.82 (m, 1 H), 1.70-2. 25 (m, 13 H), 1.20-1. 43 (m, 4 H); ESI MS m/z = 482 [C29H35N7+H] +.

Example 128-Preparation of Compound 145 Compound 71 (2.01 g, 5.98 mmol), allylbromide (2.10 mL, 24.4 mmol), and potassium carbonate (6.61 g, 47.8 mmol) were dissolved in dimethylsulfoxide (44 mL) and stirred overnight. The reaction mixture was added to 150 mL of stirring water and diluted with 150 mL ethyl acetate. The organic and aqueous phases were separated. The organic phase was washed with brine (3 x 100

mL), dried over magnesium sulfate, filtered, and concentrated. The resulting solid was dried in vacuo to afford 145 (1.98 g, 88%).

Example 129-Preparation of Compound 146 In a sealed tube, 145 (1.99 mg, 5.29 mmol), trans-1, 4- diaminocyclohexane (6.21 g, 54.3 mmol), and 2,6-di-tert-butylphenol (1.13 g, 5.48 mmol) were dissolved in ethanol (60 mL). The mixture was heated to 140 °C for 4 d.

After cooling to room temperature, the reaction was concentrated and diluted with ethyl acetate (175 mL). This organic solution was washed with water (175 mL) and brine (2 x 100 mL) and concentrated. The product was purified by silica gel chromatography and recrystallizations from ethanol in hexanes (1: 20) to yield 146 (432 mg, 18%): mp 111-114 °C ; IH NMR (300 MHz, CD30D) 8 7.25-7. 72 (m, 10 H), 5.95-6. 10 (m, 1 H), 5.21 (d, 1 H), 5.10 (d, 1 H), 4.82 (m, 2 H), 4.77 (s, 2 H), 4.67 (d, 2 H), 3.75 (m, 1 H), 2.87 (m, 1 H), 2.07 (d, 2 H), 1.92 (d, 2 H), 1.15-1. 47 (m, 4 H); ESI MS m/z= 454 [C27H31N7+H] +.

Example 130-Preparation of Compound 147 Compound 71 (2.07 g, 6.17 mmol), 2-iodobutane (3.10 mL, 26.9 mmol), and potassium carbonate (6.78 g, 49.1 mmol) were dissolved in dimethylsulfoxide (44 mL) and allowed to stir under nitrogen overnight. The reaction mixture was diluted with ethyl acetate (300 mL). The organic material was washed with water (200 mL) and brine (300 mL) and dried over magnesium sulfate. After filtration, the material was concentrated and the resulting solid was dried in vacuo to afford 147 (1.29 g, 55%).

Example 131-Preparation of Compound 148 In a sealed tube, 147 (1.29 g, 3.30 mmol) and trans-1, 4- diaminocyclohexane (3.80 g, 33.2 mmol) were dissolved in ethanol (70 mL). The mixture was heated to 140 °C. After 4 d, the reaction was cooled to room temperature, concentrated, and dissolved in ethyl acetate (160 mL). This solution was washed with water (160 mL) and brine (2 x 100 mL) and dried over magnesium sulfate. The

organic liquid was filtered and concentrated. The resulting solid was dried in vacuo and purified by silica gel chromatography and recrystallizations from ethanol in hexanes (1: 40) to yield 148 (229 mg, 15%): mp 146-150 °C ;'H NMR (500 MHz, CD30D) 5 7.70 (s, 1 H), 7.25-7. 60 (m, 9 H), 4.65-4. 90 (m, 4 H), 4.37 (m, 1 H), 3.74 (m, 1 H), 2.86 (m, 1 H), 1.82-2. 15 (m, 6 H), 1.50 (d, 3 H), 1.20-1. 43 (m, 4 H), 0.87 (t, 3 H).

Example 132-Preparation of Compound 149 To a solution of 61 (0.90 g, 1.96 mmol) in ethylene glycol dimethyl ether (54 mL), were added 3,5-dimethylphenylboronic acid (0.59 g, 3.93 mmol), triphenylphosphine (0.26 g, 0.99 mmol), and 2 M sodium carbonate solution (10 mL).

The solution was refluxed for 20 min and cooled to room temperature.

Tris (dibenzylideneacetone) dipalladium (0) (0.66 g, 0.072 mmol) was added and the reaction returned to reflux. After refluxing overnight, the reaction mixture was cooled to room temperature and another 100 mg 3,5-dimethylphenylboronic acid were added.

After refluxing for another 5 h, the reaction was quenched with 50 mL water. The aqueous solution was extracted with methylene chloride (3 x 50 mL). The extracts were combined and washed with water (50 mL) and brine (50 mL). The organic solution was dried over sodium sulfate, filtered, and concentrated. The product was purified by silica gel chromatography to yield 149: mp 86-90 °C ;'H NMR (300 MHz, CDC13) 8 7.34-7. 58 (m, 5 H), 7.20 (s, 2 H), 6.97 (s, 1 H), 5.93 (brs, 1 H), 4.54- 4.90 (m, 4 H), 3.66-3. 85 (m, 1 H), 2.70 (m, 1 H), 2.37 (s, 6 H), 2.05-2. 20 (m, 4 H), 1.80-1. 95 (m, 2 H), 1.54 (d, 6 H), 1.10-1. 35 (m, 4 H); API MS m/z = 484 [C29H37N7+H] +.

Example 133-Preparation of Compound 150 To a stirred, 0 °C solution of 149 (500 mg, 0.97 mmol) in methylene chloride (20 mL), were added pyridine (120 uL), DMAP (11.8 mg, 0.097 mmol), and acetic anhydride (91 L, 0.097 mmol). After 40 min, the reaction mixture was warmed to room temperature and stirred for 3 h. Another 100 uL acetic anhydride was added. After 1 h, the solution was concentrated and dried in vacuo. The resulting

material was purified by silica gel chromatography (95: 5: 1 CH2Cl2/methanol/NH40H) to yield 150 (400 mg, 80%): mp 207-210 °C ;'H NMR (300 MHz, CDC13) 8 7.46- 7.56 (m, 3 H), 7.36-7. 45 (m, 2 H), 7.18 (s, 2 H), 6.98 (s, 1 H), 5.98 (brs, 1 H), 5.27 (d, 1 H), 4.80 (d, 2 H), 4.56-4. 70 (m, 1 H), 3.68-3. 84 (m, 1 H), 2.37 (s, 6 H), 1.90-2. 23 (m, 7 H), 1.54 (d, 6 H), 1.15-1. 48 (m, 4 H); API MS m/z = 526 [C3lH39N7O+H] +.

Example 134-Preparation of Compound 152 To a solution of 149 (500 mg, 1.03 mmol) in 1,2-dichloroethane (4 mL) was added propionaldehyde (90 L, 1.24 mmol). After stirring for 10 min, sodium triacetoxyborohydride (306 mg, 1.44 mmol) was added. The reaction mixture stirred under nitrogen for 1.5 h before being quenched with saturated sodium bicarbonate solution (5 mL). The resulting solution was extracted with ethyl acetate (3 x 7 mL). The organic extracts were combined and dried over sodium sulfate. The organic liquid was filtered and concentrated. Purification by silica gel chromatography (90: 10: 1 CH2Cl2/methanol/NH40H) yielded 152.

Example 135-Preparation of Compound 152-HCI To a stirred solution of 152 (120 mg, 0.211 mmol) in ethyl acetate (10 mL) was added 2 M HC1 in diethyl ether (127 uL). After 20 min, the solution was concentrated and dried in vacuo to yield the HC1 salt of 152 :'H NMR (300 MHz, CD30D) 8 8.48 (s, 1 H), 7.40-7. 68 (m, 4 H), 7.20 (s, 2 H), 6.98 (s, 1 H), 4.66-5. 07 (m, 3 H), 3.80-4. 00 (m, 1 H), 2.90-3. 50 (m, 5 H), 2.01-2. 45 (m 10 H), 1.36-1. 90 (m, 14 H), 1.02 (t, 6 H); API MS m/z= 568 [C35H49N7+H] +.

Example 136-Preparation of Compound 151 To a solution of 149 (302 mg, 0.624 mmol) in 1,2-dichloroethane (2.5 mL) was added propionaldehyde (36. 0 uL, 0.500 mmol). After stirring for 15 min under nitrogen, sodium triacetoxyborohydride (93.0 mg, 0.874 mmol) was added.

After 10 min, another 93.0 mg (0.874 mmol) sodium triacetoxyborohydride were added. The reaction mixture stirred under nitrogen for 1.5 h before being quenched with saturated sodium bicarbonate solution (5 mL). The resulting solution was

extracted with ethyl acetate (3 x 7 mL). The organic extracts were combined and dried over sodium sulfate. The organic liquid was filtered and concentrated. Purification by silica gel chromatography (90: 10: 1 CH2C12/methanol/NH40H) yielded 151.

Example 137-Preparation of Compound 151-HCI To a stirred solution of 151 (90.0 mg, 0.171 mmol) in ethyl acetate (8 mL) was added 2 M HC1 in diethyl ether (103 L). After 20 min, the solution was concentrated and dried in vacuo to yield the HCl salt of 151: mp 280-290 °C ; lH NMR (300 MHz, CD30D) 8 8.45 (brs, 1 H), 7.38-7. 67 (m, 4 H), 7.29 (s, 2 H), 6.97 (s, 1 H), 4.63-5. 40 (m, 4 H), 3.78-3. 97 (m, 1 H), 2.85-3. 23 (m, 3 H), 2.07-2. 44 (m, 8 H), 1.32-1. 87 (m, 14 H), 1.02 (t, 3 H); API MS m/z= 526 [C32H43N7+H] +.

Example 138-Preparation of Compound 153 To a solution of 61 (2.03 g, 4.44 mmol) in ethylene glycol dimethyl ether (100 mL), was added 2,5-dimethoxyphenylboronic acid (2.42 g, 13.3 mmol), tris (dibenzylideneacetone) dipalladium (0) (0. 135 g, 0.148 mmol), triphenylphosphine (0.581 g, 2.22 mmol), and 2 M sodium carbonate solution (10 mL). The dispersion was refluxed overnight under nitrogen. After cooling to room temperature, the reaction mixture was diluted with 100 mL water. The aqueous solution was extracted with methylene chloride (3 x 100 mL). The extracts were combined and washed with water (300 mL) and brine (300 mL). The organic solution was dried over sodium sulfate, filtered, and concentrated. The product was purified by silica gel chromatography (90: 10: 1 CH2Cl2/methanol/NH4OH) to yield 153 (900 mg, 39%).

Example 139-Preparation of 153 #HCl To a stirred solution of 153 (100 mg, 0.194 mmol) in ethyl acetate (10 mL), was added 2 M HCl in diethyl ether (116 1L). After 20 min, the solution was concentrated to afford the HCl salt of 153 in quantitative yield: mp 278-288 °C ;'H NMR (300 MHz, CD30D) 8 8.34 (brs, 1 H), 7.36-7. 53 (m, 4 H), 6.77-7. 01 (m, 3 H), 5.30 (brs, 1 H), 4.65-5. 20 (m, 4 H), 3.80-3. 95 (m, 1 H), 3.76 (s, 1 H), 3.69 (s, 1 H),

3.05-3. 22 (m, 1 H), 2.03-2. 30 (m, 4 H), 1.35-1. 71 (m, 10 H); API MS m/z= 516 [C29H37N702+H] +.

Example 140-Preparation of Compound 154 To a 0 °C stirred solution of 153 (300 mg, 0.582 mmol) in methylene chloride (12 mL) was added pyridine (70 uL), acetic anhydride (54.0 L, 0.582 mmol), and DMAP (7.10 mg, 0. 582 mmol). The solution was stirred for 40 min before warming to room temperature. After 3 h, another 100 uL acetic anhydride was added. After stirring for 1 h, the reaction mixture was concentrated and dried in vacuo. The material was purified by silica gel chromatography (90: 10: 1 CH2Cl2/methanol/NH40H) to yield 154: mp 185-192 °C ;'H NMR (300 MHz, CD30D) 8 7.78 (s, 1 H), 7.29-7. 45 (m, 4 H), 6.87-6. 98 (m, 1 H), 6.75-6. 86 (m, 2 H), 4.75 (s, 2 H), 4.55-4. 68 (m, 1 H), 3.74 (s, 3 H), 3.67 (s, 3 H), 1.97-2. 13 (m, 2 H), 1.81-1. 96 (m, 5 H), 1.53 (d, 6 H), 1.21-1. 40 (m, 4 H); API MS m/z = 558 [C3lH39N703+H] +.

Example 141-Preparation of Compound 155 To a solution of 153 (242 mg, 0.469 mmol) in 1,2-dichloroethane (1.5 mL) was added propionaldehyde (27.0 pL, 0.375 mmol). After stirring for 15 min under nitrogen, sodium triacetoxyborohydride (140 mg, 0.657 mmol) was added. The reaction mixture was stirred under nitrogen overnight before being concentrated.

Purification by silica gel chromatography (90: 10: 1 CH2C12/methanol/NH40H) yielded 155.

Example 142-Preparation of Compound 155'HC1 To a stirred solution of 155 (30.0 mg, 0.054 mmol) in ethyl acetate (6 mL) was added 2 M HCl in diethyl ether (40. 0 uL). After 20 min, the solution was concentrated and dried in vacuo to afford the HC1 salt of 155 in quantitative yield: mp 264-268 °C ; lH NMR (300 MHz, CD30D) 8 8.41 (brs, 1 H), 7.46 (m, 4 H), 6.98 (d, 1 H), 6.76-6. 93 (m, 2 H), 4.63-5. 07 (m, 4 H), 3.89 (m, 1 H), 3.77 (s, 3 H), 3.71 (s,

3 H), 3.13 (m, 1 H), 2.97 (m, 2 H), 2.10-2. 35 (m, 4 H), 1.35-1. 88 (m, 12 H), 1.02 (t, 3 H); API MS m/z = 558 [C32H43N702+H] +.

Example 143-Preparation of Compound 156 To a solution of 153 (242 mg, 0.469 mmol) in 1,2-dichloroethane (1. 5 mL) was added propionaldehyde (27.0 1L, 0.375 mmol). After stirring for 15 min under nitrogen, sodium triacetoxyborohydride (140 mg, 0.657 mmol) was added. The reaction mixture stirred under nitrogen overnight before being concentrated.

Purification by silica gel chromatography (90: 10: 1 CHC13/methanol/NH40H) yielded 156.

Example 144-Preparation of Compound 156 HCI To a stirred solution of 156 (160 mg, 0.267 mmol) in ethyl acetate (8 mL) was added 2 M HCl in diethyl ether (170 pL). After 20 min, the solution was concentrated and dried in vacuo to afford the HC1 salt of 156 in quantitative yield: mp 235-243 °C ;'H NMR (300 MHz, CD30D) 8 8.43 (brs, 1 H), 7.47 (m, 4 H), 6.94- 7.04 (d, 1 H), 6.80-6. 93 (m, 2 H), 4.67-5. 10 (m, 4 H), 3.81-3. 98 (m, 1 H), 3.76 (s, 3 H), 3.71 (s, 3 H), 2.93-3. 48 (m, 5 H), 2.00-2. 35 (m, 4 H), 1.36-1. 90 (m, 14 H), 1.01 (t, 6 H); API MS milz = 601 [C35H49N702+H] +.

Example 145-Preparation of Compound 157 To a solution of 61 (1.50 g, 3.27 mmol) in ethylene glycol dimethyl ether (75 mL), was added 5-methyl-2-thiopheneboronic acid (1.40 g, 9.82 mmol), tris (dibenzylideneacetone) dipalladium (0) (100 mg, 0.109 mmol), triphenylphosphine (430 mg, 1.64 mmol), and 2 M sodium carbonate solution (10 mL). The solution was refluxed under nitrogen for 2 d. After cooling to room temperature, the reaction mixture was diluted with 100 mL water. The aqueous solution was extracted with methylene chloride (3 x 100 mL). The extracts were combined and washed with water (300 mL) and brine (300 mL). The organic solution was dried over sodium sulfate, filtered, and concentrated. The product was purified by silica gel chromatography (90: 10: 1 CHCl3/methanol/NH40H) to yield 157 (1.04 g, 67%).

Example 146-Preparation of Compound 157#HCl To a stirred solution of 157 (150 mg, 0.269 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (161 uL). After 20 min, the solution was concentrated to afford the HCI salt of 157 in quantitative yield: mp 300 °C ;'H NMR (300 MHz, CD30D) 8 8.35 (brs, 1 H), 7.55 (d, 2 H), 7.39 (d 2 H), 7.17 (d, 1 H), 6.74 (d, 1 H), 4.62-5. 40 (m, 5 H), 3.78-3. 94 (m, 1 H), 3.03-3. 20 (m, 1 H), 2.47 (s, 3 H), 1.98-2. 29 (m, 4 H), 1.44-1. 74 (m, 10 H); API MS m/z= 476 [C26H33N7S+H] +.

Example 147-Preparation of Compound 159 To a solution of 157 (300 mg, 0.631 mmol) in 1, 2-dichloroethane (2.0 mL) was added propionaldehyde (36.0 pL, 0.505 mmol). After stirring for 30 min under nitrogen, sodium triacetoxyborohydride (190 mg, 0.883 mmol) was added. The reaction mixture stirred under nitrogen for 3 h before being concentrated. Purification by silica gel chromatography (90: 10: 1 CHCl3/methanol/NH40H) yielded 159: mp 150-155 °C ;'H NMR (300 MHz, CD30D) 8 7.81 (s, 1 H), 7.51 (d, 2 H), 7.33 (d, 2 H), 7.11 (d, 1 H), 6.73 (d, 1 H), 4.80-5. 05 (m, 1 H), 4.55-4. 78 (m, 3 H), 3.65-3. 80 (m, 1 H), 3.01 (m, 1 H), 2.91 (t, 2 H), 2.47 (s, 3 H), 2.02-2. 19 (m, 4 H), 1.60-1. 75 (m, 8 H), 1.54 (d, 6 H), 1.19-1. 50 (m, 4 H), 1.01 (t, 3 H); API MS m/z = 518 [C29H39N7S+H] +.

Example 148-Preparation of Compound 158 To a 0 °C stirred solution of 157 (300 mg, 0.631 mmol) in methylene chloride (12 mL) was added pyridine (76 pL), acetic anhydride (58 ; je, 0.631 mmol), and DMAP (7.7 mg, 0.063 mmol). After 20 min, the reaction mixture warmed to room temperature. After mixing overnight, the reaction mixture was concentrated and dried in vacuo. The material was purified by silica gel chromatography (90: 10: 1 CHCl3/methanol/NH40H) to yield 158: mp 225-230 °C ;'H NMR (300 MHz, CDC13) 8 7.39-7. 51 (m, 3 H), 7.32 (d, 2 H), 7.07 (d, 1 H), 6.71 (m, 1 H), 6.14 (brs, 1 H), 5.32 (d, 1 H), 4.53-4. 82 (m, 4 H), 3.74 (m, 2 H), 2.50 (s, 3 H), 1.90-2. 23 (m, 7 H), 1.51 (d, 6 H), 1.12-1. 38 (m, 4 H); API MS m/z = 518 [C28H3sN7OS+H] +.

Example 149-Preparation of Compound 160 To a suspension of 61 (1.00 g, 2.18 mmol) in ethylene glycol dimethyl ether (50 mL), was added 4-methylthiophene-2-boronic acid (0.93 g, 6.54 mmol), tris (dibenzylideneacetone) dipalladium (0) (67.0 mg, 0.073 mmol), triphenylphosphine (287 mg, 1.09 mmol), and 2 M sodium carbonate solution (10 mL). The solution was refluxed under nitrogen for 2 d. After cooling to room temperature, the reaction mixture was diluted with 100 mL water. The aqueous solution was extracted with methylene chloride (3 x 100 mL). The extracts were combined and washed with water (300 mL) and brine (300 mL). The organic solution was dried over sodium sulfate, filtered, and concentrated. The product was purified by silica gel chromatography (90: 10: 1 CHC13/methanol/NH40H) to yield 160 (450 mg, 44%).

Example 150-Preparation of Compound 160 *HCI To a stirred solution of 160 (50.0 mg, 0.105 mmol) in ethyl acetate (4 mL) was added 2 M HCl in diethyl ether (100 I1L). After 20 min, the solution was concentrated to afford the HCl salt of 160 in quantitative yield: mp 308-315 °C ; IH NMR (300 MHz, CD30D) 8 8.31 (brs, 1 H), 7.60 (d, 2 H), 7. 39 (d, 2 H), 7.20 (s, 1 H), 6.94 (s, 1 H), 4.65-5. 10 (m, 3 H), 3.86 (m, 1 H), 3.13 (m, 1 H), 2.26 (s, 3 H), 2.00- 2.12 (m, 4 H), 1.32-1. 72 (m, 10 H); API MS m/z = 476 [C26H33N7S+H] +.

Example 151-Preparation of Compounds 162 and 163 To a stirred solution of 160 (300 mg, 0.631 mmol) in 1,2- dichloromethane (3 mL) was added propionaldehyde (36.0 pL). After stirring under nitrogen for 15 min, sodium triacetoxyborohydride (161 mg, 0.757 mmol) was added.

The mixture was stirred overnight. The reaction was concentrated and purified by silica gel chromatography (95: 4.5 : 1 CHCl3/methanol/NH40H) to yield 162 (80 mg, 31%), and 163 (110 mg). For 162 : lH NMR (300 MHz, CD30D) 8 7. 79 (s, 1 H), 7.54 (d, 2 H), 7.33 (d, 2 H), 7.16 (s, 1 H), 6.89 (s, 1 H), 4.71 (s, 2 H), 4.60 (m, 1 H), 3.70 (m, 1 H), 3.31 (t, 2 H), 2.42-2. 62 (m, 3 H), 2.23 (s, 3 H), 1.87-2. 10 (m, 4 H), 1.51 (d, 6 H), 1.10-1. 33 (m, 4 H), 0.94 (t, 3 H).

Example 152-Preparation of Compound 162-Cl To a stirred solution of 162 (80.0 mg, 0. 155 mmol) in ethyl acetate (6 mL) was added 2 M HCl in diethyl ether (100 nul). After 20 min, the solution was concentrated to afford the HCl salt of 162 in quantitative yield: mp 225-240 °C ; lH NMR (300 MHz, CD30D) 8 8.36 (brs, 1 H), 7.62 (d, 2 H), 7.42 (d, 2 H), 7.22 (s, 1 H), 6.95 (s, 1 H), 4.65-5. 05 (m, 4 H), 3.87 (m, 1 H), 3.11 (m, 1 H), 2.98 (t, 2 H), 2.07-2. 36 (m, 5 H), 1.32-1. 85 (m, 14 H), 1.04 (t, 3 H); API MS m/z= 518 [C29H39N7S+H] +.

Example 153-Preparation of Compound 163#HCl To a stirred solution of 163 (110 mg, 0.196 mmol) in ethyl acetate (8 mL) was added 2 M HCl in diethyl ether (120 µL). After 20 min, the solution was concentrated to afford the HCl salt of 163 in quantitative yield: mp 227-229 °C; 1H NMR (300 MHz, CD30D) 8 8.39 (brs, 1 H), 7.60 (d, 2 H), 7.41 (d, 2 H), 7.22 (s, 1 H), 6.94 (s, 1 H), 4.63-5. 10 (m, 3 H), 3.85 (m, 1 H), 2.93-3. 47 (m, 5 H), 1.92-2. 40 (m, 7 H), 1.32-1. 90 (m, 14 H), 1.04 (t, 6 H) ; API MS m/z= 560 [C32H45N7S+H] +.

Example 154-Preparation of Compound 161 To a 0 °C stirred solution of 160 (100 mg, 0.210 mmol) in methylene chloride (5 mL) were added pyridine (26.0 1L), acetic anhydride (20.0 PL, 0.210 mmol), and DMAP (3.0 mg, 0.021 mmol). After 40 min, the reaction mixture warmed to room temperature. The reaction mixture was concentrated and dried in vacuo. The material was purified by silica gel chromatography (90: 10: 1 CHCl3/methanol/NH4OH) to yield 161: mp 222-223 °C ; lH NMR (300 MHz, CD30D) 8 7.78 (s, 1 H), 7.52 (d, 2 H), 7.35 (d, 2 H), 7.16 (s, 1 H), 6.89 (s, 1 H), 4.83- 5.05 (m, 1 H), 4.72 (s, 2 H), 4.64 (m, 1 H), 3.73 (m, 1 H), 3.61 (m, 1 H), 2.25 (s, 3 H), 1.97-2. 13 (m, 2 H), 1.81-1. 96 (m, 5 H), 1.54 (d, 6 H), 1.19-1. 40 (m, 4 H); API MS m/z = 518 [C28H3sN70S+H] +.

Example 155-Preparation of Compound 164 To a solution of 61 (1.20 g, 2.62 mmol) in ethylene glycol dimethyl ether (75 mL), was added furan-3-boronic acid (0.88 g, 7.85 mmol), tris (dibenzylideneacetone) dipalladium (0) (80.0 mg, 0.087 mmol), triphenylphosphine (343 mg, 1.31 mmol), and 2 M sodium carbonate solution (10 mL). The solution was refluxed under nitrogen overnight. After cooling to room temperature, the reaction mixture was diluted with 100 mL water. The aqueous solution was extracted with methylene chloride (3 x 150 mL). The extracts were combined and washed with water (450 mL) and brine (450 mL). The organic solution was dried over sodium sulfate, filtered, and concentrated. The product was purified by silica gel chromatography (90: 10: 1 CHCl3/methanol/NH40H) to yield 164 (700 mg, 60%).

Example 156-Preparation of Compound 164 EICI To a stirred solution of 164 (100 mg, 0.224 mmol) in ethyl acetate (6 mL) was added 2 M HCl in diethyl ether (135 uL). After 20 min, the solution was concentrated to afford the HCl salt of 164 in quantitative yield: mp 320-330 °C ;'H NMR (300 MHz, CD30D) 8 8.35 (brs, 1 H), 7.90 (s, 1 H), 7.57 (m, 3 H), 7.42 (d, 2 H), 6.79 (s, 1 H), 4.65-5. 07 (m, 5 H), 3.87 (m, 1 H), 3.13 (m, 1 H), 2.00-2. 30 (m, 4 H), 1.35-1. 75 (m, 10 H); API MS m/z = 446 [C25H3lN70+H] +.

Example 157-Preparation of Compound 166 To a stirred solution of 164 (200 mg, 0.449 mmol) in 1,2- dichloromethane (2 mL) was added propionaldehyde (26. 0 uL). After stirring under nitrogen for 20 min, sodium triacetoxyborohydride (114 mg, 0.539 mmol) was added.

The mixture was stirred overnight. The reaction was concentrated and purified by silica gel chromatography (90: 10: 1 CHC13/methanol/NH40H) to yield 166.

Example 158-Preparation of Compound 166-HCl To a stirred solution of 166 (120 mg, 0.227 mmol) in ethyl acetate (7 mL) was added 2 M HCl in diethyl ether (150 pL). After 20 min, the solution was concentrated to afford the HCl salt of 166 in quantitative yield: mp 285-286 °C ; IH

NMR (300 MHz, CD30D) 8 8.42 (brs, 1 H), 7.90 (s, 1 H), 7.49-7. 62 (m, 3 H), 7.43 (d, 2 H), 6.80 (s, 1 H), 4.76-5. 10 (m, 3 H), 3.91 (m, 1 H), 2.93-3. 49 (m, 5 H), 2.00- 2.40 (m, 4 H), 1.35-1. 95 (m, 14 H), 1.03 (t, 6 H); API MS m/z = 530 [C3lH43N70+H] +.

Example 159-Preparation of Compound 165 To a 0 °C stirred solution of 164 (200 mg, 0.449 mmol) in methylene chloride (8 mL) was added pyridine (55. 0 L), acetic anhydride (46. 0 uL, 0.449 mmol), and DMAP (6.0 mg, 0.045 mmol). After 40 min, the reaction mixture warmed to room temperature. After stirring overnight, the reaction mixture was concentrated and dried in vacuo. The material was purified by silica gel chromatography (90: 10: 1 CHCl3/methanol/NH40H) to yield 165 (190 mg, 87%): mp 241-243 °C ;'H NMR (300 MHz, CD30D) 8 7.86 (s, 1 H), 7.80 (s, 1 H), 7.45-7. 54 (m, 3 H), 7.32-7. 40 (m, 2 H), 6.76 (s, 1 H), 4.71 (s, 2 H), 4.62 (m, 1 H), 3.74 (m, 1 H), 3.61 (m, 1 H), 1.73-2. 11 (m, 7 H), 1.54 (d, 6 H), 1.20-1. 40 (m, 4 H); API MS m/z = 488 [C27H33N702+H] +.

Example 160-Preparation of Compounds 167 and 168 Compound 75 (0.500 g, 1.10 mmol) was dissolved in 1,2- dichloroethane (10 mL). To this stirred solution was added acetaldehyde (0.054 g, 1.22 mmol) and sodium triacetoxyborohydride (0.360 g, 1.71 mmol). After 1.5 h, the reaction was quenched with saturated sodium bicarbonate solution (10 mL). The mixture was extracted with ethyl acetate (10 mL). The organic layers were combined, dried over sodium sulfate, and concentrated. The resulting material was purified via silica gel chromatography (60: 1: 1 CH2Cl2/methanol/triethylamine) to yield 167 (213 mg, 40%), and 168 (109 mg, 19%).

Example 161-Preparation of Compound 167-Cl To a stirred solution of 167 (213 mg, 0.440 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.264 mL). The organic liquid was concentrated to affordthe HCl salt of 167 in quantitative yield :'H NMR (300 MHz, CD30D) 8 8.30 (brs, 1 H), 7.30-7. 63 (m, 10 H), 4.71-4. 80 (m, 3 H), 3.65-3. 72 (m, 1

H), 3.48 (q, 2 H), 3.10 (brs, 2 H), 2.10-2. 20 (m, 4 H), 1.60 (d, 6 H), 1.30-1. 59 (m, 7 H); API MS m/z = 484 [C29H37N7+H] +.

Example 162-Preparation of Compound 168-Cl To a stirred solution of 168 (109 mg, 0.213 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.128 mL). The organic liquid was concentrated to afford the HCl salt of 168 in quantitative yield :'H NMR (300 MHz, CD30D) 8 7.25-7. 67 (m, 10 H), 4.60-4. 80 (m, 3 H), 3.68-3. 80 (m, 1 H), 3.48 (q, 4 H), 2.78-2. 95 (m, 4 H), 2.02-2. 17 (m, 2 H), 1.10-1. 59 (m, 10 H), 1.35 (t, 6 H); API MS m/z= 512 [C3lH4lN7+H] +.

Example 163-Preparation of Compounds 169 and 170 Compound 75 (0.500 g, 1.10 mmol) was dissolved in 1,2- dichloroethane (10 mL). To this stirred solution was added butyraldehyde (0.072 g, 1.00 mmol) and sodium triacetoxyborohydride (0.297 g, 1.40 mmol). After 2.5 h, the reaction was quenched with saturated sodium bicarbonate solution (10 mL). The mixture was extracted with ethyl acetate (10 mL). The organic layers were combined, dried over sodium sulfate, and concentrated. The resulting material was purified via silica gel chromatography (200: 10: 1 CH2Cl2/methanol/NH40H) to yield 169 (180 mg, 32%), and 170 (160 mg, 26%).

Example 164-Preparation of Compound 169-HCI To a stirred solution of 169 (170 mg, 0.332 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.199 mL). The organic liquid was concentrated to afford the HCl salt of 169 in quantitative yield :'H NMR (300 MHz, CD30D) 8 8.30-8. 34 (m, 1 H), 7.30-7. 68 (m, 10 H), 5.30 (brs, 1 H), 4.65-4. 90 (m, 4 H), 3.80-3. 92 (m, 2 H), 2.94-3. 35 (m, 4 H), 2.15-2. 32 (m, 4 H), 1.41-1. 75 (m, 12 H), 1.00 (t, 3 H).

Example 165-Preparation of Compound 171 To a stirred, 0 °C solution of 169 (125 mg, 0.228 mmol) in methylene chloride (10 mL), was added pyridine (46 pL), DMAP (6.0 mg, 0.046 mmol), and acetic anhydride (24.0 L, 0.251 mmol). After 1 h under a nitrogen atmosphere, the reaction mixture was warmed to room temperature. After stirring overnight, another 2.2 equivalents of acetic anhydride and 0.2 equivalents of DMAP were added and the mixture was heated to reflux. Following concentration, the material was diluted with ethyl acetate (20 mL) and saturated sodium bicarbonate solution (20 mL). The organic layer was concentrated and dried in vacuo. The resulting material was purified via silica gel chromatography (90: 10: 1 CH2Cl2/methanol/NH40H) and trituration with hexanes to yield 171 (26 mg): API MS m/z = 554 [C33H43N70+H] +.

Example 166-Preparation of Compound 170-HCI To a stirred solution of 170 (150 mg, 0.264 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.158 mL). The organic liquid was concentrated to afford the HCl salt of 170 in quantitative yield :'H NMR (300 MHz, CD30D) 6 8.15-8. 25 (m, 1 H), 7.31-7. 68 (m, 10 H), 4.65-4. 90 (m, 3 H), 3.70-3. 95 (m, 1 H), 2.95-3. 41 (m, 6 H), 2.05-2. 32 (m, 4 H), 1.31-1. 79 (m, 18 H), 1.00 (t, 6 H).

Example 16-Preparation of Compounds 172 and 173 Compound 75 (0.500 g, 1.10 mmol) was dissolved in 1,2- dichloroethane (10 mL). To this stirred solution was added cyclopropanecarboxaldehyde (0.070 g, 1.00 mmol) and sodium triacetoxyborohydride (0.297 g, 1.40 mmol). After 3 h, the reaction was quenched with saturated sodium bicarbonate solution (10 mL). The mixture was extracted with ethyl acetate (10 mL).

The organic layers were combined, dried over sodium sulfate, and concentrated. The resulting material was purified via silica gel chromatography (200: 10: 1 CH2CI2/methanol/NH40H) to yield 172 (103 mg, 18%), and 173 (160 mg, 26%).

Example 168-Preparation of Compound 172-HCl To a stirred solution of 172 (103 mg, 0.202 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.121 mL). The organic liquid was concentrated to afford the HCl salt of 172 in quantitative yield : 1H NMR (300 MHz, CD30D) 8 8.30 (brs, 1 H), 7.30-7. 69 (m, 10 H), 4.69-4. 92 (m, 4 H), 3.80-3. 92 (m, 1 H), 2.84-3. 19 (m, 3 H), 2.11-2. 28 (m, 4 H), 1.37-1. 72 (m, 10 H), 1.05-1. 15 (m, 1 H), 0.68-0. 74 (m, 2 H), 0.38-0. 42 (m, 2 H).

Example 169-Preparation of Compound 173-HCI To a stirred solution of 173 (160 mg, 0.284 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.170 mL). The organic liquid was concentrated to afford the HCl salt of 173 in quantitative yield : IH NMR (300 MHz, CD30D) 8 8.30-8. 41 (m, 1 H), 7.30-7. 68 (m, 10 H), 5.30 (brs, 1 H), 4.68-4. 90 (m, 4 H), 3.55-3. 95 (m, 2 H), 3.05-3. 20 (m, 4 H), 2.00-2. 32 (m, 4 H), 1.10-1. 90 (m, 10 H), 0.75-0. 80 (m, 4 H), 0.35-0. 50 (m, 4 H).

Example 170-Preparation of Compounds 174 and 175 Compound 75 (1.50 g, 3.29 mmol) was dissolved in 1,2-dichloroethane (30 mL). To this stirred solution was added propionaldehyde (0.174 g, 2.99 mmol) and sodium triacetoxyborohydride (0.888 g, 4.19 mmol). After 1.5 h, the reaction was quenched with saturated sodium bicarbonate solution (30 mL). The mixture was extracted with ethyl acetate (30 mL). The organic layers were combined, dried over sodium sulfate, and concentrated. The resulting material was purified via silica gel chromatography (200: 10: 1 CH2Cl2/methanol/NH40H) to yield 174 (317 mg, 19%): 'H NMR (300 MHz, CD30D) 8 7.26-7. 80 (m, 10 H), 4.62-4. 81 (m, 3 H), 3.74 (brs, 1 H), 2.41-2. 62 (m, 3 H), 1.90-2. 11 (m, 4 H), 1.52 (d, 6 H), 1.12-1. 52 (m, 8 H), 0.92 (t, 3 H), and 175 (320 mg, 18%) :'H NMR (300 MHz, CD30D) 8 8.18-8. 28 (m, 1 H), 7.28-7. 68 (m, 10 H), 4.65-4. 90 (m, 3 H), 3.81-3. 94 (m, 1 H), 2.94-3. 25 (m, 4 H), 2.02- 2.31 (m, 6 H), 1.40-1. 81 (m, 14 H), 1.05 (t, 6 H).

Example 171-Preparation of Compound 267 The HCl salt of 167 (10 mg, 0.037 mmol), was dispersed in ethyl acetate and neutralized with sodium bicarbonate. The organic material was dried over magnesium sulfate and concentrated. The solid was dissolved in dry CH2Cl2 (10 mL) and cooled to 0 °C. To the solution was added DMAP (9 mg), pyridine (0.074 mL) and acetic anhydride (0.037 mL). The ice bath was removed after 1 h. After being stirred overnight, additional DMAP and Ac20 was added in portions to consume starting material by TLC analysis. The mixture was heated to reflux for 2 d. Upon cooling, the mixture was concentrated in vacuo, then neutralized with aqueous sodium bicarbonate, extracted with ethyl acetate, dried and concentrated. The residue was purified by chromatography to provide 267: API MS m/z = 526 [C31H39N70+H] +.

Example 172-Preparation of Compound 177 To a solution of 61 (1.00 g, 2.21 mmol) and 3-tolylboronic acid (0.33 g, 2.43 mmol) in tetrahydrofuran (5 mL) was added tris (dibenzylideneacetone) dipalladium (0) (0.010 g, 0.011 mmol), tri-tert- butylphosphine (5.5 mg, 0.027 mmol), and potassium fluoride (0.42 g, 7.29 mmol).

After mixing overnight at room temperature, the reaction mixture was refluxed for 24 h and cooled to room temperature. The reaction mixture was diluted with ether (50 mL) and filtered through Celite. The organic liquid was concentrated and the resulting material was purified via silica gel chromatography (90: 10: 1 CH2Cl2/methanol/NH40H) to yield 177 (0.70 g, 71%).

Example 173-Preparation of Compound 177 HCI To a stirred solution of 177 (449 mg, 0. 956 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (575 pL). The organic liquid was concentrated to afford the HCl salt of 177 in quantitative yield: mp 186-195 °C ;'H NMR (300 MHz, CD30D) 8 7.82 (s, 1 H), 7.54 (d, 2 H), 7.20-7. 48 (m, 5 H), 7.12 (d, 1 H), 4.83-5. 10 (m, 2 H), 4.77 (s, 2 H), 4.64 (m, 1 H), 3.77 (m, 1 H), 3.03 (m, 1 H), 2.38 (s, 3 H), 1.93-2. 20 (m, 4 H), 1.19-1. 70 (m, 10 H); API MS m/z = 470 [C28H35N7+H] +.

Example 174-Preparation of Compound 178 To a stirred solution of 177 (219 mg, 0.466 mmol) in methylene chloride (25 mL) was added acetic anhydride (48 pL, 0.513 mmol), DMAP (5.7 mg, 0.047 mmol), and pyridine (57. 0 uL, 0.699 mmol). The mixture was placed under a nitrogen atmosphere and immersed in an ice water bath. After 30 min, the reaction mixture was warmed to room temperature and stirred for another 1.5 h. The solution was concentrated and the resulting material was purified via silica gel chromatography (95: 5: 1 CH2Cl2/methanol/NH40H) to afford 178.

Example 175-Preparation of Compound 178-HCI To a stirred solution of 178 (50.0 mg, 0.098 mmol) in ethyl acetate (5 mL) was added 2 M HCl in diethyl ether (59.0 I1L). The organic liquid was concentrated to afford the HCl salt of 178 in quantitative yield: mp 165-174 °C ; IH NMR (300 MHz, CD30D) S 8.26 (brs, 1 H), 7.61 (d, 2 H), 7.47 (d, 2 H), 7.24-7. 41 (m, 3 H), 7.15 (d, 1 H), 4.62-5. 08 (m, 4 H), 3.83 (m, 1 H), 3.67 (m, 1 H), 2.39 (s, 3 H), 1.87-2. 20 (m, 7 H), 1.60 (d, 6 H), 1.40 (m, 4 H); API MS m/z = 512 [C3oH37N70+H] +.

Example 176-Preparation of Compound 179 To a solution of 61 (2.00 g, 4.36 mmol) and 3-methoxyphenyl boronic acid (0.73 g, 4.79 mmol) in tetrahydrofuran (10 mL) was added tris (dibenzylideneacetone) dipalladium (0) (20 mg, 0.022 mmol), tri-tert- butylphosphine (10.0 mg, 0.052 mmol), and potassium fluoride (0.84 g, 14.39 mmol).

After refluxing overnight, the reaction mixture was diluted with ether (50 mL) and filtered through Celite. The organic liquid was concentrated and the resulting material was purified via silica gel chromatography (95: 5: 1 CH2Cl2/methanol/NH40H) to yield 179 (1.18 g, 56%).

Example 177-Preparation of Compound 179'HC1 To a stirred solution of 179 (980 mg, 2.02 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (1.21 mL). The organic liquid was

concentrated to afford the HCl salt of 179 in quantitative yield: mp 182-189 °C ;'H NMR (300 MHz, CD30D) 6 7.80 (s, 1 H), 7.54 (d, 2 H), 7.40 (d, 2 H), 7.30 (t, 1 H), 7.12 (m, 2 H), 6.87 (m, 1 H), 4.76 (s, 2 H), 4.62 (m, 1 H), 3.67-3. 90 (m, 4 H), 2.94 (m, 1 H), 1.90-2. 20 (m, 4 H), 1.17-1. 65 (m, 10 H); API MS m/z = 486 [C2sH3sN70+H] Example 178-Preparation of Compound 180 To a stirred solution of 179 (200 mg, 0.412 mmol) in methylene chloride (25 mL) was added acetic anhydride (43 JuL, 0.450 mmol), DMAP (5.0 mg, 0.041 mmol), and pyridine (50.0 pL, 0.618 mmol). The mixture was placed under a nitrogen atmosphere and immersed in an ice water bath. After 30 min, the reaction mixture was warmed to room temperature and stirred for another 1.5 h. The solution was concentrated and the resulting material was purified via silica gel chromatography (95: 5: 1 CH2Cl2/methanol/NH4OH) to afford 180.

Example 179-Preparation of Compound 180-HCI To a stirred solution of 180 (60.0 mg, 0.114 mmol) in ethyl acetate (5 mL) was added 2 M HCl in diethyl ether (68. 0 L). The organic liquid was concentrated to afford the HCl salt of 180 in quantitative yield :'H NMR (300 MHz, CD30D) 6 8.20 (brs, 1 H), 7.62 (d, 2 H), 7.46 (d, 2 H), 7.34 (t, 1 H), 7.10-7. 20 (m, 2 H), 6.90 (1 H), 4.60-5. 10 (m, 4 H), 3.83 (s, 3 H), 3.64 (m, 1 H), 3.20-3. 42 (m, 1 H), 1.87-2. 18 (m, 7 H), 1.60 (d, 6 H), 1.22-1. 50 (m, 4 H); API MS m/z = 528 [C3oH37N702+H] +.

Example 180-Preparation of Compound 181 To a solution of 61 (2.00 g, 4.36 mmol) and furan-2-boronic acid (1.50 g, 13.1 mmol) in ethylene glycol dimethyl ether (150 mL) was added tris (dibenzylideneacetone) dipalladium (0) (120 mg, 0.130 mmol), tri-tert- butylphosphine (570 mg, 2.18 mmol), and 2 M sodium carbonate solution (12.5 mL, 25.3 mmol). After refluxing overnight, 2 more equivalents of furan-2-boronic acid were added. The reaction was refluxed for 24 h, cooled to room temperature, and

diluted with water (50 mL). The aqueous mixture was extracted with methylene chloride (3 x 80 mL). The extracts were combined and washed with water (250 mL) and brine (250 mL). The organic phase was dried over sodium sulfate and filtered.

The organic liquid was concentrated and the resulting material was purified via silica gel chromatography (95: 5: 1 CH2Cl2/methanol/NH40H) to yield 181.

Example 181-Preparation of Compound 181-Cl To a stirred solution of 181 (600 mg, 1.35 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.810 mL). The organic liquid was concentrated to afford the HCI salt of 181 (406 mg, 68%) :'H NMR (300 MHz, CD30D) 5 8.06 (s, 1 H), 7.66 (d, 2 H), 7.53 (s, 1 H), 7. 41 (d, 2 H), 6.74 (m, 1 H), 6.49 (m, 1 H), 4.60-5. 00 (m, 5 H), 3.82 (m, 1 H), 3.10 (m, 1 H), 1.95-2. 20 (m, 4 H), 1.20- 1.61 (m, 10 H); API MS m/z= 446 [C25H3lN7O+H] +.

Example 182-Preparation of Compound 182 To a stirred solution of 181 (750 mg, 1.68 mmol) in methylene chloride (30 mL) was added acetic anhydride (0.18 mL, 1.85 mmol), DMAP (20.8 mg, 0.17 mmol), and pyridine (0.20 mL, 2.52 mmol). The mixture was placed under a nitrogen atmosphere and immersed in an ice water bath. After 30 min, the reaction mixture was warmed to room temperature and stirred for another 1.5 h. The solution was concentrated and the resulting material was purified via silica gel chromatography (95: 5: 1 CH2Cl2/methanol/NH40H) to afford 182 (530 mg, 65%).

Example 183-Preparation of Compound 182 HCI To a stirred solution of 182 (300 mg, 0.620 mmol) in ethyl acetate (10 mL) was added 2 M HCl in diethyl ether (0.370 mL). The organic liquid was concentrated to afford the HC1 salt of 182 in quantitative yield :'H NMR (300 MHz, CD30D) 8 8.25 (brs, 1 H), 7.56-7. 73 (m, 2 H), 7.54 (s, 1 H), 7.30-7. 47 (m, 2 H), 6.75 (m, 1 H), 6.49 (m, 1 H), 4.60-5. 05 (m, 4 H), 3.73-3. 90 (m, 1 H), 3.55-3. 73 (m, 1 H), 1.82-2. 23 (m, 7 H), 1.15-1. 70 (m, 10 H); API MS m/z = 488 [C27H33N702+H] +.

Example 184-Preparation of Compound 183 To a stirred mixture of 6-chloronicotinamide (5.00 g, 31.9 mmol) in ethanol (13 mL) and toluene (80 mL) was added 3-fluorobenzeneboronic acid (4.92 g, 35.1 mmol) and 2 M sodium carbonate solution (32 mL). The suspension was heated to 80 °C and degassed with argon for 1 h. After cooling to room temperature, tetrakis (triphenylphophine) palladium (0) (1.11 g, 0.958 mmol) was added. The reaction mixture was refluxed under argon for 3 h. After cooling to room temperature, the mixture was diluted with water (100 mL) and filtered. The filter cake was washed with water and dried in vacuo to afford 183 (6.38 g, 92%).

Example 185-Preparation of Compound 184 To a stirred suspension of 183 (3. 00 g, 13.9 mmol) in tetrahydrofuran (25 mL) was added dropwise 1 M borane in THF (97.0 mL, 97.0 mmol). After refluxing for 2 h, the reaction mixture was cooled in an ice bath. The mixture was acidified to pH 1 with 2 N HC1 and stirred for 1 h. The pH was raised to a value of 10 by adding 6 N NaOH and the resulting solution was extracted with ethyl acetate (3 x 50 mL). The extractions were combined, washed with brine (150 mL), and dried over sodium sulfate. The suspension was filtered and concentrated. The resulting material was purified by precipitation as the HCl salt from an ethanol solution. The product was recovered by filtration and dried in vacuo to. yield 184 (1.69 g, 51%) : tH NMR (300 MHz, CD30D) 8 8.92 (s, 1 H), 8.41 (d, 1 H), 8.27 (d, 1 H), 7.77-7. 90 (m, 2 H), 7.35-7. 70 (m, 2 H), 4.36 (s, 2 H).

Example 186-Preparation of Compound 185 The amine 184 (1.84 g, 7.69 mmol), 2,6-dichloropurine (1.31 g, 6.99 mmol), and N, N-diisopropylethylamine (2.68 mL, 15.4 mmol) were dissolved in ethanol (65 mL). After refluxing overnight, the solution was immersed in an ice water bath for 20 min. The mixture was filtered and cake was washed with water. The cake was triturated with ethanol and diethyl ether and dried in vacuo to afford 185 (1.12 g, 47%).

Example 187-Preparation of Compound 186 To a stirred solution of 185 (1.00 g, 2.94 mmol) in dimethylsulfoxide (100 mL) was added potassium carbonate (2.19 g, 15.9 mmol) and 2-iodopropane (0.88 mL, 8.81 mmol). The mixture was placed under an argon atmosphere and stirred overnight. The reaction mixture was poured into stirred water (300 mL) and the resulting solution was extracted with ethyl acetate (3 x 300 mL). The extractions were combined, washed with water (900 mL) and brine (900 mL), and dried over magnesium sulfate. Following filtration, the organic liquid was concentrated. The material was purified by recrystallization from ethyl acetate in hexanes to yield 186 (0.92 g, 82%).

Example 188-Preparation of Compound 187 In a sealed tube were combined 186 (640 mg, 1.67 mmol), trans-1, 4- diaminocyclohexane (0.96 g, 8.36 mmol), and ethanol (3.5 mL). The reaction mixture was heated to 150 °C for 4 d and cooled to room temperature. The solution was poured into stirred ice water (5 mL) and the resulting mixture was extracted with methylene chloride (3 x 5 mL). The extractions were combined, washed with water (15 mL) and brine (15 mL), and dried over sodium sulfate. The organic liquid was concentrated. Purification by column chromatography (97: 3 CH2C12/methanol) and trituration with hexanes yielded the free base, 187 :'H NMR (300 MHz, CDC13) 8 8.68 (s, 1 H), 7.60-7. 82 (m, 4 H), 7.50 (s, 1 H), 7.42 (q, 1 H), 7.11 (m, 1 H), 6.07 (m, 1 H), 4.85 (d, 2 H), 4.60 (m, 2 H), 3.74 (m, 1 H), 2.69 (m, 1 H), 2.02-2. 19 (m, 2 H), 1.78-1. 96 (m, 2 H), 1.52 (d, 6 H), 1.10-1. 36 (m, 4 H); API MS m/z = 475 [C26H3lFN8+H].

Example 189-Preparation of Compound 188 The free amine 187 (80. 0 mg, 0.170 mmol) was dissolved in methylene chloride (4 mL). The solution was immersed in an ice water bath and acetic anhydride (17.5 pL, 0.185 mmol), DMAP (2.0 mg, 0. 017 mmol), and pyridine (20.0 pL, 0.252 mmol) were added. After stirring for 30 min, the solution was warmed to room temperature and concentrated. Purification via silica gel chromatography and

trituration with hexanes yielded 188 (67 mg, 78%): mp 199-230 °C ;'H NMR (300 MHz, CDCl3) 8 8.71 (s, 1 H), 7.62-7. 85 (m, 4 H), 7.52 (s, 1 H), 7.42 (m, 1 H), 7.10 (m, 1 H), 6.01 (m, 1 H), 5.28 (d, 1 H), 4.81 (d, 2 H), 4.62 (m, 2 H), 3.72 (m, 2 H), 1.90-2. 27 (m, 7 H), 1.53 (d, 6 H), 1.24 (m, 4 H); API MS milz = 517 [C28H33FN80+H] +.

Example 190-Preparation of Compound 189 To a stirred mixture of 6-chloronicotinamide (3.00 g, 19.2 mmol) in ethanol (7.6 mL) and toluene (48 mL) were added 3-methoxyphenylboronic acid (3.20 g, 21.1 mmol) and 2 M sodium carbonate solution (19 mL). The suspension was heated to 80 °C and degassed with argon for 1 h. After cooling to room temperature, tetrakis (triphenylphophine) palladium (0) (664 mg, 0.575 mmol) was added. The reaction mixture was refluxed under argon for 3 h. After cooling to room temperature, the mixture was diluted with water (100 mL) and filtered. The filter cake was washed with water and dried in vacuo to afford 189 (3.62 g, 83%).

Example 191-Preparation of Compound 190 To a stirred solution of 189 (3.00 g, 13.1 mmol) in tetrahydrofuran (25 mL) was added dropwise 1 M borane in THF (92.0 mL, 92.0 mmol). After refluxing for 4 h, the reaction mixture was cooled in an ice bath. The mixture was acidified to pH 1 with 2 N HCl and stirred for 1 h. The pH was raised to a value of 10 by adding 6 N NaOH and the resulting solution was extracted with ethyl acetate (3 x 50 mL).

The extractions were combined, washed with brine (150 mL), and dried over sodium sulfate. The suspension was filtered and concentrated. The resulting material was purified by precipitation as the HCl salt from an ethanol solution. The product was recovered by filtration and dried in vacuo to yield 190 (1.81 g, 55%).

Example 192-Preparation of Compound 191 The amine 190 (1.80 g, 7.18 mmol), 2,6-dichloropurine (1.22 g, 6.53 mmol), and N, N diisopropylethylamine (1.86 g, 6.53 mmol) were dissolved in ethanol (82 mL). After refluxing overnight, the dispersion was immersed in an ice water bath

for 60 min. The mixture was filtered and cake was washed with water. The cake was triturated with ethanol and diethyl ether and dried in vacuo to afford 191 (1.04 g, 44%).

Example 193-Preparation of Compound 192 To a stirred solution of 191 (1.04 g, 2.84 mmol) in dimethylsulfoxide (60 mL) was added potassium carbonate (2.12 g, 15.3 mmol) and 2-iodopropane (0.85 mL, 8.52 mmol). The mixture was placed under an argon atmosphere and stirred overnight. The reaction mixture was poured into stirred water (60 mL) and the resulting solution was extracted with ethyl acetate (3 x 60 mL). The extractions were combined, washed with water (180 mL) and brine (180 mL), and dried over magnesium sulfate. Following filtration, the organic liquid was concentrated. The material was purified by recrystallization from ethyl acetate in hexanes (1: 40) to yield 192.

Example 194-Preparation of Compound 193 In a sealed tube were combined 192 (400 mg, 1.09 mmol), trans-1, 4- diaminocyclohexane (1.25 g, 10.9 mmol), and ethanol (4.0 mL). The reaction mixture was heated to 150 °C for 24 h and cooled to room temperature. The solution was filtered and the filtrate was concentrated. Purification by column chromatography (97: 3 CH2Cl2/methanol) yielded the free base, 193 (240 mg, 45%).

Example 195-Preparation of Compound 194 The free amine 193 (130 mg, 0.27 mmol) was dissolved in methylene chloride (6 mL). The solution was immersed in an ice water bath and acetic anhydride (28. 0 uL, 0.294 mmol), DMAP (3.2 mg, 0.026 mmol), and pyridine (33. 0 pL, 0.401 mmol) were added. After stirring for 30 min, the solution was warmed to room temperature and concentrated. Purification via prep-TLC (9: 1 CH2Cl2/methanol) and trituration with ethyl acetate yielded 194: mp 161-163'C ;'H NMR (300 MHz, CDC13) 8 8.76 (s, 1 H), 7.77 (d, 1 H), 7.64 (d, 1 H), 7.50 (m, 3 H), 7.35 (t, 1 H), 6.73-

7.00 (m, 2 H), 5.33 (d, 1 H), 4.50-4. 92 (m, 4 H), 3.56-4. 00 (m, 5 H), 1.84-2. 22 (m, 7 H), 1.55 (d, 6 H), 1.25 (m, 4 H); API MS m/z = 529 [C29H36N802+H] +.

Example 196-Preparation of Compound 195 To a stirred mixture of 6-chloronicotinamide (2.00 g, 12.8 mmol) in ethanol (5.0 mL) and toluene (32 mL) was added thiophene-2-boronic acid (1.80 g, 14.1 mmol) and 2 M sodium carbonate solution (13 mL). The suspension was heated to 80 °C and degassed with argon for 1 h. After cooling to room temperature, tetrakis (triphenylphophine) palladium (0) (443 mg, 0.383 mmol) was added. The reaction mixture was refluxed under argon for 3 h. After cooling to room temperature, another 0.950 g thiophene-2-boronic acid and 280 mg tetrakis (triphenylphophine) palladium (0) were added to the reaction mixture. It was refluxed for 4 h and cooled to room temperature. The mixture was diluted with water (50 mL) and filtered. The filter cake was washed with water and dried in vacuo to afford 195 (1.65 g, 63%).

Example 197-Preparation of Compound 196 To a stirred solution of 195 (1.40 g, 6.86 mmol) in tetrahydrofuran (23 mL) was added dropwise 1 M borane in THF (48.0 mL, 48.0 mmol). After refluxing for 1 h, the reaction mixture was cooled in an ice bath. The mixture was acidified to pH 1 with 2 N HCl and stirred for 1 h. The pH was raised to a value of 10 by adding 6 N NaOH and the resulting solution was extracted with ethyl acetate (3 x 50 mL). The extractions were combined, washed with brine (150 mL), and dried over sodium sulfate. The suspension was filtered and concentrated. The resulting material was purified by precipitation as the HCl salt from an ethanol solution. The product was recovered by filtration and dried in vacuo to yield 196 (0.87 g, 56%).

Example 198-Preparation of Compound 197 The amine 196 (210 mg, 1.10 mmol), 2,6-dichloropurine (188 mg, 1.00 mmol), and N, N diisopropylethylamine (286 g, 2.21 mmol) were dissolved in ethanol (12 mL). After refluxing overnight, the suspension was immersed in an ice water bath

for 60 min. The mixture was filtered and cake was washed with water. The cake was triturated with ethanol and diethyl ether and dried in vacuo to afford 197 (206 mg, 60%).

Example 199-Preparation of Compound 198 To a stirred solution of 197 (200 mg, 0.583 mmol) in dimethylsulfoxide (12 mL) was added potassium carbonate (435 mg, 3.15 mmol) and 2-iodopropane (0.18 mL, 1.75 mmol). The mixture was placed under an argon atmosphere and stirred overnight. The reaction mixture was poured into stirred water (15 mL) and the resulting solution was extracted with ethyl acetate (3 x 30 mL). The extractions were combined, washed with water (90 mL) and brine (90 mL), and dried over magnesium sulfate. Following filtration, the organic liquid was concentrated to yield 198 (200 mg, 89%).

Example 200-Preparation of Compound 199 In a sealed tube were combined 198 (100 mg, 0.260 mmol), trans-1, 4- diaminocyclohexane (297 mg, 2.60 mmol), and ethanol (2.0 mL). The reaction mixture was heated to 150 °C for 2 d and cooled to room temperature. The solution was filtered and diluted with ethanol. The filtrate was concentrated, and converted to its HCl salt to afford 199 :'H NMR (300 MHz, CD30D) 8 9.00 (s, 1 H), 8.72 (d, 2 H), 8.46 (d, 1 H), 8.27 (m, 1 H), 8.15 (d, 1 H), 7.54 (m, 1 H), 4.89-5. 35 (3 H), 4.04 (m, 1 H), 3.35 (m, 1 H), 2.20-2. 50 (m, 4 H), 1.55-1. 90 (m, 10 H); ESI MS m/z = 463 [C24H3oNgS+H]''.

Example 201-Preparation of Compound 200 The free amine 199 (100 mg, 0.216 mmol) was dissolved in methylene chloride (5 mL). The solution was immersed in an ice water bath and acetic anhydride (20.0 pL, 0.216 mmol), DMAP (2.6 mg, 0.021 mmol), and pyridine (33. 0 L, 0.324 mmol) were added. After stirring for 30 min, the solution was warmed to room temperature and concentrated. Purification via prep-TLC (10: 1 CH2CIJmethanol) yielded 200 (20 mg): mp 206-208 °C ; lH NMR (300 MHz, CDC13) 8 8.56 (s, 1 H),

7.70 (d, 1 H), 7. 58 (d, 1 H), 7. 53 (m, 2 H), 7. 37 (m, 1 H), 7.10 (m, 1 H), 6.15 (brs, 1 H), 5.33 (d, 1 H), 4.76 (d, 2 H), 4.61 (m, 2 H), 3.70 (m, 2 H), 1.75-2. 15 (m, 7 H), 1.52 (d, 6 H), 1.23 (m, 4 H); API MS mlz = 505 [C26H32N8OS+H] +.

Example 202-Preparation of Compound 201 To a stirred solution of 6-chloronicotinamide (2.80 g, 17.9 mmol) in ethanol (7.5 mL) and toluene (48 mL) was added furan-2-boronic acid (3.00 g, 26.8 mmol) and 2 M sodium carbonate solution (18 mL). The suspension was heated to 80 °C and degassed with argon for 1 h. After cooling to room temperature, tetrakis (triphenylphophine) palladium (0) (619 mg, 0.536 mmol) was added. The reaction mixture was refluxed under argon for 2 d then cooled to room temperature.

The mixture was diluted with water (75 mL) and filtered. The filter cake was washed with water and dried in vacuo to afford 201 (1.95 g, 58%).

Example 203-Preparation of Compound 202 To a stirred solution of 201 (1.69 g, 8.98 mmol) in tetrahydrofuran (34 mL) was added dropwise 1 M borane in THF (50.0 mL, 50.0 mmol). After refluxing for 2 h, the reaction mixture was cooled in an ice bath. The mixture was acidified to pH 1 with 2 N HCl and stirred for 1 h. The pH was raised to a value of 10 by adding 6 N NaOH and the resulting solution was extracted with ethyl acetate (3 x 50 mL).

The extractions were combined, washed with brine (150 mL), and dried over sodium sulfate. The suspension was filtered and concentrated. The resulting material was purified by precipitation as the HCl salt from an ethanol solution. The product was recovered by filtration and dried in vacuo to yield 202 (1.12 g, 50%).

Example 204-Preparation of Compound 203 The amine 202 (177 mg, 1.02 mmol), 2,6-dichloropurine (173 mg, 0.920 mmol), and N, N diisopropylethylamine (267 g, 2.07 mmol) were dissolved in ethanol (11 mL). After refluxing overnight, the suspension was immersed in an ice water bath for 60 min. The mixture was filtered and cake was washed with water.

The cake was triturated with ethanol and diethyl ether and dried in vacuo to afford 203 (166 mg, 54%).

Example 205-Preparation of Compound 204 To a stirred solution of 203 (166 mg, 0.508 mmol) in dimethylsulfoxide (11 mL) was added potassium carbonate (379 mg, 2.74 mmol) and 2-iodopropane (0.150 mL, 1.52 mmol). The mixture was placed under an argon atmosphere and stirred overnight. The reaction mixture was poured into stirred water (15 mL) and the resulting solution was extracted with ethyl acetate (3 x 30 mL). The extractions were combined, washed with water (90 mL) and brine (90 mL), and dried over magnesium sulfate. Following filtration, the organic liquid was concentrated to yield 204 (178 mg, 95%).

Example 206-Preparation of Compound 205 In a sealed tube were combined 204 (170 mg, 0.461 mmol), trans-1, 4- diaminocyclohexane (526 mg, 4.61 mmol), and ethanol (2.5 mL). The reaction mixture was heated to 150 °C for 4 d and cooled to room temperature. The solution was filtered and concentrated to afford 205.

Example 207-Preparation of Compound 206 The free amine 205 (100 mg, 0.224 mmol) was dissolved in methylene chloride (5 mL). The solution was immersed in an ice water bath and acetic anhydride (21. 0 uL, 0.224 mmol), DMAP (2.7 mg, 0.022 mmol), and pyridine (34.0 PL, 0.336 mmol) were added. After stirring for 30 min, the solution was stored at 0 °C overnight. Purification via prep-TLC (9: 1 CH2Cl2/methanol) yielded 206 (43 mg): mp 216-218 °C ;'H NMR (300 MHz, CDC13) 8 8.63 (s, 1 H), 7.72 (d, 1 H), 7.61 (d, 1 H), 7.50 (m, 2 H), 6.99 (m, 1 H), 6.50 (m, 1 H), 6.15 (brs, 1 H), 5.36 (d, 1 H), 4.79 (d, 2 H), 4.62 (m, 2 H), 3.68 (m, 2 H), 1.78-2. 20 (m, 7 H), 1.54 (d, 6 H), 1.22 (m, 4 H) ; API MS mlz = 489 [C26H32N802+H] +.

Example 208-Preparation of Compound 207 Prepared by reaction of 72 with ethylene diamine by general methods described above (91%) :'H NMR (300 MHz, (CD3) 2SO) 8 8.06 (brs, 3 H), 7.65 (d, 4 H), 7.31-7. 52 (m, 6 H), 4.68-4. 90 (m, 3 H), 3.61 (m, 2 H), 3.01 (m, 2 H), 1.54 (d, 6 H); ESI MS m/z=402 [C23H27N7+H]+.

Example 209-Preparation of Compound 208 Prepared by reaction of 207 under standard acetylation conditions (35%) :'H NMR (300 MHz, CDC13) 5 7.49-7. 62 (m, 5 H), 7.30-7. 48 (m, 5 H), 6.44 (brs, 1 H), 6.13 (brs, 1 H), 5. 05 (t, 1 H), 4.82 (d, 2 H), 4.65 (m, 1 H), 3.58 (m, 2 H), 3.45 (m, 2 H), 1.87 (s, 3 H), 1.54 (d, 6 H); ESI MS m/z = 444 [C25H29N70+H] +.

Example 210-Preparation of Compound 211 Prepared by reaction of 72 and 1,3-propanediamine (28%) :'H NMR (300 MHz, (CD3) 2SO) 8 8.00-8. 22 (m, 3 H), 7.63-7. 70 (d, 4 H), 7.32-7. 59 (m, 6 H), 4.84 (m, 2 H), 4.70 (m, 1 H), 4.43 (m, 2 H), 2.88 (m, 2 H), 1.88 (m, 2 H), 1.52 (d, 6 H); ESI MS m/z = 416 [C24H29N7+H] +.

Example 211-Preparation of Compound 212 Prepared by reaction of 211 with acetic anhydride under standard conditions (44%): mp 106-107 °C ; IH NMR (300 MHz, CDC13) 6 7.50-7. 62 (m, 5 H), 7.28-7. 50 (m, 5 H), 5.87 (brs, 2 H), 4.93 (m, 1 H), 4.84 (m, 2 H), 4.66 (m, 1 H), 3.49 (m, 2 H), 3.33 (m, 2 H), 1.91 (s, 3 H), 1.74 (m, 2 H), 1.55 (d, 6 H); ESI MS m/z = 458 [C26H3lN70+H] +.

Example 212-Preparation of Compound 213 Prepared by the general methods described above (48%) :'H NMR (300 MHz, CDC13) 8 7.29-7. 66 (m, 10 H), 6.40 (brs, 1 H), 4.71-5. 03 (m, 3 H), 4.61 (m, 1 H), 3.30-3. 55 (m, 2 H), 2.73 (m, 2 H), 2.32 (m, 2 H), 1.38-1. 85 (m, 10 H); ESI MS m/z = 430 [C25H31N7+H]+.

Example 213-Preparation of Compound 214 Prepared by the general methods described above (45%) :'H NMR (300 MHz, CDC13) 8 7.27-7. 65 (m, 10 H), 5.92 (m, 1 H), 5.43 (m, 1 H), 4.83 (m, 3 H), 4.66 (m, 1 H), 3.46 (m, 2 H), 3.25 (m, 2 H), 1.93 (s, 3 H), 1.76 (m, 3 H), 1.40-1. 70 (m, 8 H); ESI MS m/z = 472 [C27H33N70+H] +.

Example 214-Preparation of Compounds 209 and 210 Prepared by the general methods described above. For 210 (17%) : 1H NMR (300 MHz, (CD3) 2SO) 8 7.31-7. 72 (m, 10 H), 4.64-4. 92 (m, 3 H), 3.73 (m, 4 H), 2.84-3. 33 (m, 6 H), 1.43-1. 79 (m, 10 H), 0.85 (m, 6 H); ESI MS m/z = 486 [C29H39N7+H] +. For 209 :'H NMR (300 MHz, CDC13) 8 7.26-7. 63 (m, 10 H), 6.46 (brs, 1 H), 5.83 (brs, 1 H), 4.85 (m, 2 H), 4.66 (m, 1 H), 3.79 (d, 2 H), 3.11 (m, 2 H), 2.76 (m, 2 H), 1.76 (m, 2 H), 1.50 (d, 6 H), 0.84 (m, 3 H); ESI MS m/z = 444 [C26H33N7+H] +.

Example 215-Preparation of Compound 215 Prepared by the general methods described above:'H NMR (300 MHz, CDCl3) 8 7.22-7. 60 (m, 10 H), 6.18 (brs, 1 H), 5.11 (brs, 1 H), 4.81 (m, 2 H), 4.64 (m, 1 H), 3.40 (m, 2 H), 2.87 (m, 2 H), 2.77 (m, 2 H), 1.70-2. 00 (m, 4 H), 1.55- 1.70 (m, 2 H), 1.51 (m, 6 H), 0.92 (t, 3 H); ESI MS m/z = 472 [C28H37N7+H] +.

Example 216-Preparation of Compound 216 Prepared by the general methods described above (71%) :'H NMR (300 MHz, CDCl3) 5 7.28-7. 61 (m, 10 H), 6.01 (brs, 1 H), 4.86 (d, 2 H), 4.66 (m, 1 H), 3.79-3. 89 (m, 1 H), 3.70-3. 79 (m, 1 H), 3.57-3. 70 (m, 2 H), 3.27-3. 37 (m, 1 H), 2.10-2. 23 (m, 1 H), 1.68-1. 82 (m, 1 H), 1.55 (d, 6 H); ESI MS m/z = 428 [C2sH29N7+H] +.

Example 217-Preparation of Compound 217 Prepared by the general methods described above: TLC silica gel Rf = 0.52 (20: 1: 0. 01-CH2Cl2/MeOH/NH40H).

Example 218-Preparation of Compound 218 Prepared by the general methods described above : IH NMR (300 MHz, (CD3) 2SO) 8 8.02 (brs, 1 H), 7.88 (s, 1 H), 7.54-7. 68 (m, 4 H), 7.40-7. 50 (m, 3 H), 7.30-7. 40 (m, 1 H), 7.26 (s, 1 H), 6.77 (s, 1 H), 4.50-4. 72 (m, 4 H), 2.78 (t, 2 H), 2.31 (m, 1 H), 1.68 (d, 2 H), 1.49 (d, 6 H); ESI MS m/z = 470 [C27H3lN70+H] +.

Example 219-Preparation of Compound 219 Prepared by the general methods described above (66%) :'H NMR (300 MHz, CDC13) 8 7.28-7. 62 (m, 10 H), 5.89 (brs, 1 H), 4.86 (d, 4 H), 4.67 (m, 1 H), 2.81 (t, 2 H), 2.58 (d, 2 H), 1.77 (d, 2 H), 1.40-1. 69 (m, 10 H), 1.06-1. 31 (m, 2 H); ESI MS m/z = 456 [C27H33N7+H] +.

Example 220-Preparation of Compound 221 The compound 220 (100 mg, 0.170 mmol) was dissolved in methanol (25 mL). To the stirred solution was added ammonium formate (100 mg), and Pd/C (10.0 mg). After refluxing for 2 h, more ammonium formate (100 mg) and Pd/C (10.0 mg) were added. The reaction was cooled to room temperature and filtered through Celite. The filtrate was concentrated in vacuo. The resulting material was purified via silica gel chromatography (3: 1: 0.01 CH2Cl2/MeOH/NH40H) to yield 221 (26.9 mg, 34%): 1H NMR (300 MHz, CDC13) 6 7.29-7. 63 (m, 10 H), 5.95 (brs, 1 H), 4.73-4. 93 (m, 3 H), 4.64 (m, 1 H), 3.32 (t, 2 H), 3.10 (d, 2 H), 2.59 (t, 2 H), 1.76 (m, 2 H), 1.54 (d, 6 H), 1.28 (m, 3 H), 0.90 (m, 1 H); ESI MS m/z = 456 [C27H33N7+H] +.

Example 221-Preparation of Compound 222 Prepared by the general methods described above :'H NMR (300 MHz, CDC13) 8 7.28-7. 61 (m, 10 H), 6.20 (brs, 1 H), 4.70-4. 91 (m, 3 H), 4.54-4. 70 (m, 1 H), 4.28 (brs, 1 H), 3.75-3. 90 (m, 1 H), 3.08-3. 11 (m, 1 H), 2.80-2. 93 (m, 1 H), 2.28-2. 41 (d, 1 H), 1.95-2. 10 (m, 1 H), 1.84-1. 95 (m, 1 H), 1.70-1. 84 (m, 1 H), 1.60- 1.70 (m, 1 H), 1.52 (d, 6 H), 1.22-1. 41 (m, 2 H), 0.94-1. 22 (m, 2 H), 0.89 (t, 1 H); ESI MS m/z = 456 [C27H33N7+H] +.

Example 222-Preparation of Compound 223 Prepared by the general Suzuki coupling conditions of 62 with boronic ester as shown in Scheme LXXV (58%): mp 200-206 °C ;'H NMR (300 MHz, (CD3) 2SO) 8 7.22-8. 00 (m, 9 H), 6.10 (m, 2 H), 4.40-4. 76 (m, 4 H), 3.63 (m, 1 H), 1.62-2. 01 (m, 7 H), 1.35-1. 60 (d, 6 H), 1.08-1. 35 (m, 4 H); ESI MS mlz = 527 [CsoHssNsO+H] .

Example 223-Preparation of Compound 224 Prepared by the general Suzuki coupling conditions of 61 and 3,4- dimethylbenzeneboronic acid :'H NMR (300 MHz, CDCl3) 8 7.53 (m, 3 H), 7.41 (d, 2 H), 7.26-7. 38 (m, 2 H), 7.19 (d, 1 H), 4.79 (s, 2 H), 4.64 (m, 1 H), 3.80 (m, 1 H), 3.12 (m, 1 H), 2.10-2. 36 (m, 10 H), 1.43-1. 72 (m, 8 H), 1.27 (m, 4 H); ESI MS m/z = 484 [C29H37N7+H] +.

Example 224-Preparation of 5-Bromo-2-cyanopyridine 2,5-Dibromopyridine (20.0 g, 84.4 mmol) was dissolved in dimethylformamide (422 mL). To the stirred solution was added copper (I) cyanide.

After refluxing for 5 h, the mixture was cooled to room temperature and stored overnight. The reaction mixture was diluted with ethyl acetate (1200 mL) and filtered through a Buchner funnel containing sand, Celite, and silica gel layers. The filtrate was concentrated to a volume of 400 mL. This organic liquid was diluted with water (300 mL) and the resulting liquid was extracted with ethyl acetate (2 x 200 mL). The organic extracts were combined, washed with water (2 x 300 mL) and brine (1 x 250 mL), and dried over magnesium sulfate. After concentration, the product was purified via silica gel chromatography (50: 50 ethyl acetate/CH2Cl2) to afford the title compound (9.79 g).

Example 225-Preparation of Compound 225 Prepared by reation of 5-bromo-2-cyanopyridine with benzeneboronic acid under standard Suzuki conditions (68%).

Example 226-Preparation of Compound 226 In a Parr shaker vessel were combined 225 (300 mg, 1.67 mmol), glacial acetic acid (25 mL), and 10% palladium on carbon catalyst (177 mg, 0.167 mmol). The solution was agitated under 45 psig hydrogen gas for 2 h. The resulting dispersion was filtered through a Buchner funnel. The filtrate was concentrated.

Purification by acid/base extraction yielded 226 (240 mg, 78%).

Example 227-Preparation of Compound 229 Following the general schemes outlined above, compound 226 was transformed into 227 (57% yield). Compound 227 was then transformed into 228 in 83% yield. Compound 228 was then converted into compound 229 and then its HCl salt (75%) :'H NMR (300 MHz, CD30D) 8 9.11 (s, 1 H), 8.90 (d, 2 H), 8.23 (d, 1 H), 7.83 (m, 2 H), 7.58 (m, 3 H), 5.23 (m, 2 H), 4.70-5. 01 (m, 1 H), 3.72 (m, 1 H), 3.09 (m, 1 H), 1.80-2. 15 (m, 4 H), 1.20-1. 80 (m, 10 H); ESI MS m/z = 457 [C26H32N8+H] +.

Example 228-Preparation of Compound 230 In a flask immersed in an ice water bath were combined 187 (30.0 mg, 0.055 mmol), BOC-L-alanine (10.4 mg, 0.055 mmol), HATU (25.0 mg, 0.066 mmol), N, N-diisopropylethylamine (0.050 mL, 0.274 mmol), and dimethylformamide (0.500 mL) for 10 min then warmed to room temperature. After stirring overnight, the reaction mixture was diluted with methylene chloride (50 mL). The organic material was washed with 1 M citric acid (2 x 50 mL), saturated sodium bicarbonate solution (50 mL), and brine (50 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo. To remove remaining dimethylformamide, the resulting material was dissolved in ethyl acetate (50 mL) and rinsed with 5% lithium chloride solution (3 x 50 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo to yield 230 (28.0 mg, 79%) : 1H NMR (300 MHz, CDCl3) 8 8.71 (s, 1 H), 7.62-7. 83 (m, 4 H), 7.37-7. 60 (m, 2 H), 7.11 (m, 1 H), 6.11 (brs, 1 H), 6.00 (d, 1 H), 5.01 (brs, 1 H), 4.81 (d, 2 H), 4.64 (m, 2 H), 4.08 (m, 1 H), 3.72 (m, 2 H), 2.12 (m, 2 H), 2.00 (m, 2 H), 1.54 (d, 6 H), 1.43 (s, 9 H), 1.15-1. 38 (m, 7 H); ESI MS milz = 646 [C34H44FN903+H] +.

Example 229-Preparation of Compound 231 To a stirred solution of 230 in methylene chloride (2 mL) was added HCl in ethanol (2 mL). After stirring for 10 min, the solution was concentrated in vacuo to yield 231 (15.4 mg) :'H NMR (300 MHz, CD30D) 8 8.92 (s, 1 H), 8.35- 8.67 (m, 2 H), 8.32 (d, 1 H), 7. 83 (t, 2 H), 7.69 (m, 1 H), 7.40 (m, 1 H), 4.65-5. 20 (m, 3 H), 3.60-4. 00 (m, 2'H), 1.80-2. 30 (m, 4 H), 2.65 (d, 3 H), 1.08-1. 58 (m, 12 H); ESI MS =546 [C29H36FN90+H] .

Example 230-Preparation of Compound 232 In a flask immersed in an ice water bath were combined 187 (30.0 mg, 0.055 mmol), BOC-glycine (9.6 mg, 0.055 mmol), HATU (25.0 mg, 0.066 mmol), N, N-diisopropylethylamine (0.05 mL, 0.274 mmol), and dimethylformamide (0.50 mL) for 10 min then warmed to room temperature. After stirring overnight, the reaction mixture was diluted with methylene chloride (50 mL). The organic material was washed with 1 M citric acid (2 x 50 mL), saturated sodium bicarbonate solution (50 mL), and brine (50 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo. To remove remaining dimethylformamide, the resulting material was dissolved in ethyl acetate (50 mL) and rinsed with 5% lithium chloride solution (3 x 50 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo to yield 232 (33.0 mg) :'H NMR (300 MHz, CDC13) 6 8.65 (s, 1 H), 7.52-7. 75 (m, 4 H), 7.44 (s, 1 H), 7.35 (q, 1 H), 7.01 (t, 1 H), 6.05 (brs, 1 H), 4.75 (d, 2 H), 4.57 (m, 2 H), 3.70 (m, 2 H), 2.05 (m, 2 H), 1.92 (m, 2 H), 1.46 (d, 6 H), 1.40 (s, 9 H), 1.20 (m, 6 H); ESI MS m/z= 632 [C33H42FN903+H] +.

Example 231-Preparation of Compound 233 To a stirred solution of 232 in methylene chloride (2 mL) was added HCl in ethanol (2 mL). After stirring for 10 min, the solution was concentrated in vacuo to yield 233 (10.6 mg) :'H NMR (300 MHz, CD30D) 8 8.95 (s, 1 H), 8.28- 8.75 (m, 3 H), 7.79 (t, 2 H), 7.69 (m, 1 H), 7.46 (t, 1 H), 4.63-5. 20 (m, 3 H), 3.59-3. 92 (m, 2 H), 1.96-2. 24 (m, 4 H), 1.62 (d, 6 H), 1.05-1. 52 (m, 8 H); ESI MS m/z = 532 [C28H34FN90+H] +.

Example 232-Preparation of Compound 239 Reaction of 234 with 1 under standard conditions provides 236 (90%).

Reaction of 236 with trans-1, 4-cyclohexanediamine provides 237 (95%). Boc protection of 237 followed by Suzuki coupling provides 238 in 50% yield. Compound 238 was added to a 1: 1 mixture of methylene chloride and trifluroacetic acid. After stirring for 2 h, the solution was concentrated in vacuo. The resulting material was purified via silica gel chromatography (94: 5: 1 CH2Cl2 : MeOH: NH40H) to afford 239: 'H NMR (300 MHz, CDCl3) 8 7.51-7. 61 (m, 4 H), 7.48 (s, 1 H), 7.41 (t, 2 H), 7.31 (m, 3 H), 5.61 (m, 1 H), 4.63 (m, 2 H), 3.87 (m, 2 H), 3.18 (m, 1 H), 3.00 (t, 2 H), 2.20-2. 35 (m, 3 H), 1.72 (m, 4 H), 1.52 (d, 6 H), 1.25 (m, 4 H).

Example 233-Preparation of Compound 240 Compound 239 was acetylated under the general conditions described above to provide 240 (73%). Salt formation occurred in 71% yield :'H NMR (300 MHz, CDCl3) 8 7.28-7. 62 (m, 10 H), 5.67 (m, 1 H), 4.64 (m, 2 H), 3.68-3. 97 (m, 3 H), 3.00 (t, 2 H), 2.23 (m, 2 H), 1.84-2. 11 (m, 7 H), 1.53 (d, 6 H), 1.30 (m, 4 H).

Example 234-Preparation of Compound 241 Reductive amination of 239 with propionaldehyde followed by salt formation provided 241: ESI MS m/z = 512 [C3lH41N7+H] +.

Example 235-Preparation of Compound 242 Compound 237 was Boc-protected and then treated with 3- thiopheneboronic acid under standard Suzuki condition to prepare 242 :'H NMR (300 MHz, CDCl3) 8 7.33-7. 76 (m, 6 H), 7.28 (m, 2 H), 5.72 (brs, 1 H), 4.64 (m, 1 H), 4.43 (m, 1 H), 3.83 (m, 2 H), 3.47 (m, 1 H), 2.97 (t, 2 H), 2.21 (m, 2 H), 2.08 (m, 2 H), 1.53 (d, 6 H), 1.46 (s, 9 H), 1.29 (m, 4 H); ESI MS m/z = 576 [C31H41N702S+H] +.

Example 236-Preparation of Compound 243 Compound 242 was deprotected with HCl in methanol to provide 243: 'H NMR (500 MHz, CDCl3) 6 7.68 (m, 1 H), 7.54 (d, 2 H), 7.47 (s, 1 H), 7.42 (s, 1

H), 7.37 (s, 1 H), 7.28 (m, 2 H), 5.68 (brs, 1 H), 4.63 (m, 2 H), 3.85 (m, 3 H), 2.99 (t, 2 H), 2.71 (m, 1 H), 2.18 (d, 2 H), 1.90 (d, 2 H), 1.52 (d, 6 H), 1.25 (m, 4 H).

Example 237-Preparation of Compound 245 To a stirred solution of sodium hydride (423 mg, 17.6 mmol) in tetrahydrofuran (12 mL), was added 4-phenylphenol (2.00 g, 11.8 mmol). After 1 h, BOC-2-aminoethylbromide (3.90 g, 17.6 mmol) was added to the solution. After stirring overnight, the reaction mixture was quenched with 2 N potassium hydroxide solution (10 mL). The resulting mixture was extracted with methylene chloride (12 mL). The organic layer was concentrated and the crude material was purified via silica gel chromatography to yield 245.

Example 238-Preparation of Compound 246 The protected amine 245 was added to 10 mL of an 1: 1 mixture of methylene chloride and trifluoroacetic acid. After concentration, the material was diluted with 2 N potassium hydroxide solution (10 mL). The aqueous layer was extracted with methylene chloride (2 x 10 mL). The organic extracts were combined, dried over magnesium sulfate, and concentrated in vacuo to afford the product (400 mg). Reaction with 1 under standard conditions provided 246 (91%).

Example 239-Preparation of Compound 248 Compound 246 was transformed into 247 under standard conditions (80%). Reaction of 247 with trans-1, 4-cyclohexanediamine provided 248. Salt formation provided the target compound (68%) :'H NMR (500 MHz, CDC13) 8 7.53 (m, 4 H), 7.40 (m, 2 H), 7.29 (m, 2 H), 7.00 (d, 2 H), 5.93 (brs, 1 H), 4.61 (m, 2 H), 4.22 (t, 2 H), 4.02 (m, 2 H), 3.78 (m, 1 H), 2.70 (m, 1 H), 2.18 (d, 2 H), 1.90 (d, 2 H), 1.53 (d, 6 H), 1.25 (m, 4 H).

Example 240-Preparation of Compound 250 Reductive amination of 248 with propionaldehyde and salt formation under standard conditions described above provided 250: ESI MS m/z = 528 [C31H4lN70+H] +.

Example 241-Preparation of Compound 249 N-Acetylation of 248 and salt formation under standard conditions provided 249 :'H NMR (300 MHz, CDC13) 8 6.95-7. 60 (m, 10 H), 5.97 (brs, 1 H), 5.24 (d, 1 H), 4.63 (m, 2 H), 4.23 (t, 2 H), 4.02 (m, 2 H), 3.78 (m, 2 H), 2.21 (m, 2 H), 2.04 (m, 2 H), 1.94 (s, 3 H), 1. 55 (d, 6 H), 1. 30 (m, 4 H).

Example 242-Preparation of Compound 255 Utilizing reaction conditions described in general above, 251 was converted to 252 (100%). Compound 252 was converted to 253 then 254 and then Boc-protected to make 255 (21%).

Example 243-Preparation of Compound 256 Compound 255 was treated with phenylboronic acid under standard Suzuki condions. The product was dissolved in methanol and immersed in an ice water bath. Hydrogen chloride gas was bubbled through the solution. The solution was concentrated in vacuo and the resulting material was purified via preparatory HPLC (acetonitrile/water/trifluoroacetic acid) to yield 256 (8 mg).

Example 244-Preparation of Compound 257 Compound 255 was treated with 3-thiopheneboronic acid under standard Suzuki conditions. The product was dissolved in methanol and immersed in an ice water bath. Hydrogen chloride gas was bubbled through the solution. The solution was concentrated in vacuo and the resulting material was purified via preparatory HPLC (acetonitrile/water/trifluoroacetic acid) to yield 257 :'H NMR (300 MHz, CDC13) 8 7.31-7. 58 (m, 8 H), 5.99 (brs, 1 H), 5.10-5. 50 (m, 1 H), 4.49-

4.69 (m, 2 H), 3.67 (m 1 H), 2.67 (m, 1 H), 2.13 (m, 2 H), 1.90 (m, 2 H), 1.62 (d, 3 H), 1.53 (d, 6 H), 1.21 (m, 4 H).

Example 245-Preparation of Compound 258 Reaction of 75 with propionoyl chloride under standard conditions provides 258 (89%): mp 182-183 °C Example 246-Preparation of Compound 259 Reaction of 75 with methyl chloroformate under standard conditions provides 259 (68%): mp 148-150 °C.

Example 247-Preparation of Compound 260 Reaction of 75 with methanesulfonyl chloride under standard conditions provides 260 (56%): mp 143-145 °C.

Example 248-Preparation of Compound 261 Reaction of 75 with cyclopropanecarbonyl chloride under standard conditions provides 261 (87%): mp 196-204 °C.

Example 249-Preparation of Compound 262 Compound 75 (250 mg, 0.549 mmol) and succinic anhydride (60.0 mg, 0.600 mmol) were dissolved in xylene (30 mL). A few drops of dimethylformamide were added to the solution. After refluxing for 48 h, the mixture was concentrated in vacuo. The resulting material was purified via silica gel chromatography (99.5 : 0.5 CH2Cl2/MeOH) and recrystallized from CHzCh in hexanes (1: 10) to yield 262 (30. 0 mg, 10%): mp 141-147 °C.

Example 250-Preparation of Compound 263 The amine 75 (200 mg, 0.439 mmol) was dissolved in methylene chloride (15 mL). The stirred solution was cooled to-78 °C and N, N- diisopropylethylamine (113 mg, 0.878 mmol) and trifluromethylsulfonylchloride

(81.4 mg, 0.483 mmol) were added. After 30 min, the solution was warmed to room temperature. The mixture was cooled to-78 °C and another 1.10 equivalents of trifluoromethylsulfonylchloride and 1.50 equivalents of N, N diisopropylethylamine were added. After warming to room temperature, the solution was concentrated. The resulting material was purified via silica gel chromatography (99: 1 CH2Cl2/MeOH) and recrystallization from ether in hexanes to afford 263 (60 mg, 23%): mp 131-136 °C.

Example 251-Preparation of Compound 264 Prepared by standard Suzuki coupling of 61 to provide 264 (65%): mp 186-190 °C.

Example 252-Preparation of Compound 265 N-Acetylation of 264 under standard conditions provides 265 (37%): mp 241-246 °C.

Example 253-Preparation of Compound 266 Suzuki coupling of 61 with 2-chlorobenzeneboronic acid provides 266 (13%): API MS m/z = 490 [C27H32C1N7+H] +.

Example 254-Description of Biological Assays A. Immunopurification of CyclinA/cdk2 and CyclinE/cdk2 Complexes.

CyclinA/cdk2 and cyclinE/cdk2 assays were carried out with cyclin/cdk complexes isolated from HeLa S-3 suspension cultures. HeLa cells were grown in spinner flasks at 37 °C in Joklik's modified minimum essential media (MEM) supplemented with 7% horse serum. After growing in medium supplemented with 2 mM thymidine for 16-18 h, cultures were arrested at the Gl/S border and cyclinA/cdk2 and cyclinE/cdk2 were isolated from cell lysates by immunoprecipitation with antibodies specifically directed against each cyclin subunit.

Rabbit anti-cyclinA (H-432) and the mouse monoclonal antibody against cyclinE

(HE111) were purchased from Santa Cruz Biotechnology. Cells blocked at the appropriate stage of the cell cycle were disrupted in lysis buffer (50 mM Tris, pH 8.0, 250 mM NaCl, 0.5% NP-40 plus protease and phosphatase inhibitors) and centrifuged at 10,000 x g to remove insoluble material. To isolate cyclin/cdk complexes, 1 pg of anti-cyclin antibody was incubated with lysate from 1 x 107 cells for 1 h at 4 °C.

Protein A-coated agarose beads were then added for 1 h to collect antibody-bound immune complexes. The immobilized cyclin/cdk complexes were then washed 4 x with lysis buffer to reduce nonspecific protein binding. The complexes were then washed 1 x in kinase assay buffer (50 mM Tris-HCl, pH 7.4, 10 mM MgCl2, 1 mM DTT) and aliquoted into individual assay tubes.

B. Immunopurification of CyclinB/cdkl Complex.

HeLa cells are blocked at the G1/S border by culturing in the presence of 2 mM thymidine for 20 h. The cells are then rinsed 3x in phosphate buffered saline and resuspended in regular medium. After 4 h of culture, the mitotic blocker, nocodazole is added to a final concentration of 75 ng/ml. Sixteen hours later, the cells are harvested by centrifugation, washed in PBS, and lysed in cold Lysis Buffer (50 mM Tris pH 8.0, 250 mM NaCl, 0.5% NP-40, 1 mM DTT, 25 ug/ml leupeptin, 25 p g/ml aprotinin, 15 pg/ml benzamidine, 1 mM PMSF, 50 mM sodium fluoride, 1 mM sodium orthovanadate) for 15 min at 1 x 107 cells/ml. The lysate is then clarified by centrifugation at 10,000 x g for 10 min. The supernatant is collected and diluted 1: 5 with Lysis Buffer. Monoclonal antibody against cyclinB (GNS 1) is added to the supernatant to a final concentration of 5 ug/ml and shaken at 4 °C for 2 h. The immune complexes are then collected by the addition of 200 PI of protein agarose beads for 1 h. The beads are washed 4x in lysis buffer and 1 x in kinase assay buffer.

C. Protein Kinase Assays and Determination of IC50 Values.

CyclinA/cdk2 assays were carried out with complexes isolated from 0.5 x 106 cells. CyclinE/cdk2 assays were carried out with complexes isolated from 4 x 106 cells. CyclinB/cdkl assays were carried out with complexes isolated from 4 x 104 cells. After centrifugation, the wash buffer was removed and the complexes resuspended in 15 pl of kinase assay buffer (kinase wash buffer + 167 pg/ml histone H1). Compounds being tested for inhibition were added prior to the addition of [y 32p] ATP to a final concentration of 15 uM. The tubes were incubated at 30 °C for 5

min and the reactions were stopped by the addition of an equal volume of 2 x SDS- PAGE sample buffer. The samples were then subjected to electrophoresis on 10% SDS-PAGE to resolve the histone H1 from other reaction components. The amount of radioactive phosphate transferred to histone H1 was quantified on a Storm Phosphorimager (Molecular Dynamics).

Prior to the protein kinase assay, test compounds were dissolved in DMSO at a concentration of 25 mM and were diluted to produce final concentrations of 0.1, 1.0, and 10. 0 uM in the kinase assays. To eliminate possible effects of differences in DMSO concentration, the DMSO was kept constant at 0.04%, including the control reaction. Duplicate assays were performed at each concentration. The activity was plotted as the percent of activity in the absence of added test compound versus test compound concentration. ICso values were calculated using GraphPad Prism data analysis software.

D. Measuring the Inhibition of Cell Growth.

Growth inhibition (iso) values were measured with HeLa S-3 cells selected for growth on plastic. The procedure was based on the protocol of Skehan et al. (Skehan, P. , et al. , J. Natl. Cancer Inst., 82: 1107-1112 (1990), which is hereby incorporated by reference) HeLa cells were plated at 2 x 104 cells/well in 96 well plates. One day later, a control plate was fixed by addition of TCA to 5%. After five rinses with tap water the plate was air dried and stored at 4 °C. Test compounds were added to the remaining plates at 10-fold dilutions between 0. 01 and 100 uM. Two days later all plates were fixed as described above. Cells were then stained by the addition of 100 ul per well of 0.4% sulforhodamine B (SRB) in 1% acetic acid for 30 min at 4 °C. Wells were then quickly rinsed 5 x with acetic acid (1%) and allowed to air dry. The SRB was then solubilized by the addition of 100 1 per well of unbuffered 10 mM Tris base. Dye was quantified by measuring absorbance at 490 nm on a Molecular Devices kinetic microplate reader. Growth at each inhibitor concentration relative to the untreated control was calculated according to the following equation: percent growth = 100 x (T-To)/ (C-To), where T was the average optical density (OD) of the test wells after 2 days of treatment, To was the average OD of the wells in the control plate on day 0 and C was the average OD of untreated

wells. Plots of percent growth versus inhibitor concentration were used to determine the GIso.

The data below shown in Table 2 summarizes the in vitro cyclin/cdk inhibition constants (IC5o) and growth inhibition constants (GIso) of HeLa Cells for the compounds of the current invention. Replicate experimental results are summarized below.

Table 2: In Vitro Cyclin/cdk Inhibition (IC50) and Growth Inhibition (GIso) of HeLa Cells For Compounds of the Current Invention. Compound ICso CyclinA/cdk2 ICso CyclinE/cdk2 ICso CyclinB/cdkl GIso HeLa Cells (µM) (µM) (µM) (µM) 5 >10 12 7 5 0. 4 0. 6 >10 12 2 1 3 0.06 0. 7 3 0.003 0. 9 0.5 0.001 0.2 0. 1 0.02 0.0001 13 4 2 4 3 1 0. 3 2 0.8 0.9 14 3 0. 4 7 0. 4 3 2 0. 03 0.03 17 1 1 10 0. 4 2 0.9 3 0. 6 1 0. 2 11 0.25 >10 9 0.4 10 2 0.3 0.4 25 1 4 >10 2 6 1 >10 0.4 > 10 9 > 1 32 2 3--5 5 0. 9 0. 7 33 >10 4 >10 1 13 6 2 8 0. 9 34 12 5 >10 7 13 2 6 7 36 >10 >10 >10 20 >10 >10 20 >10 >10 38 >10 >10 >10 0.6 >10 >10 1 0.6 40 >10 >10 >10 9 >10 >10 25 >10 43 >10 >10 >10 4 >10 >10 4 8 46 >10 6 >10 25 8 3 >10 48 22 1 >10 0.3 6 5 0. 6 0.5 50 >10 >10 >10 3 7 9 > 10 53 >10 15 >10 0. 2 >10 4 0.3 0.5 58 11 2 12 2 4 4 0. 5 0.7 60 >10 12 >10 7 0.4 >10 6 73 >50 4 >10 0.3 14 12 0.5 > 10 > 10 0.3 > 10 > 10 0.5 74 5 2 6 0. 2 2 3 0.01 1 2 0.05 0.03 0. 05 75 3 3 6 0.09 0.02 0. 005 76 12 3 6 0. 07 11 5 0.01 3 2 0.06 0.2 0. 04 77 >10 4 >10 0.15 >10 14 0.5 0.3 78 0.9 0.6 0.8 0.05 0.9 0.3 0.8 0.025 0.7 0.2 0. 08 0.002 79 10 2 3 0.07 0.5 0.1 0.007 1 0. 08 0. 004 0.4 80 >10 >10 >10 >100 >10 4 >10 2 86 0.9 0.4 2 0. 2 0.7 0.2 0.03 0.4 0.4 0.01 0.6 0.03 0. 01 0.2 87 4 1 5 0.07 2 0.3 0.01 0.5 0.1 0. 004 0.006 0.03 0.006 0. 001 0. 0001 88 3 4 >10 0.1 >10 >10 0.05 2 5 0.04 0.005 93 0.2 0.09 0.9 0.3 0.3 0.1 0.08 0.3 94 0.6 0.3 0.4 0.1 0.2 0.3 0.07 0.4 95 1 1 4 0.08 2 0.7 0.003 0.0005 96 8 4 6 0.04 0.01 97 > 10 3 10 3 98 6 2 >10 >10 2 2 11 99 >10 9 >10 5 100 >10 4 >10 0. 6 101 3 1 4 1 0. 9 0. 7 1 102 > 10 4--4 103 0.6 0.2 1 0. 03 0.7 0. 2 0.008 0.02 0.01 104 7 1 2 0.4 8 1 0. 2 106 11 3 -- 0.3 4 1 0. 1 107 1 2--0. 4 4 0. 3 108 10 >10 -- 3 >10 >10 5 109 0.6 0. 1 -- 0. 04 < 0. 0001 110 0.6 2 -- 0.02 0.03 0.02 0.01 111 0.2 0. 07 -- 0. 02 0. 0006 112 2 2 -- <0.001 0.002 0.02 0.006 0. 0006 113 0.4 0. 3 -- <0. 001 0. 00001 0.03 0. 001 0.02 114 3 0.7 -- >10 115 30.4 116 > >10 -- >10 >10 117 > 10 3 3 118 6 1 -- >10 >10 123 0.2 0. 04 -- <0.001 < 0.001 0.0001 124 2 0. 8 -- 0.003 < 0.001 < 0. 0001 130---->10 >10 131 -- -- -- 3 2 132 -- -- -- 4 3 133----->10 >10 134 -- -- -- 2 3 135 4 3 137 -- -- -- 0.05 0. 06 0. 05 139 -- -- -- 0.2 0. 07 140-----1 2 142 -- -- -- 0.4 0. 5 144 -- -- -- 0. 4 0.4 146 -- -- -- 0. 7 0.3 146 -- -- -- 0.7 0.3 148 -- -- -- 1 1 149 -- -- -- 0.3 0.2 150 -- -- -- 0. 3 0.2 151 -- -- -- 0.8 0.6 152 -- -- -- 0.7 0.3 153 -- -- -- 3 2 154 -- -- -- 0. 6 0.9 155 -- -- -- 0. 5 0.8 156 -- -- -- 3 2 157 -- -- -- 0. 4 0.5 158 -- -- -- 0. 6 0.4 159 4 3 160 -- -- -- 0. 2 0.3 161---0. 2 0.4 162 -- -- -- 0. 2 0.3 163 -- -- -- 2 3 164----0. 2 0.1 165 -- -- -- 0.2 0.1 166----4 2 167 -- -- -- 2 0.9 168 -- -- -- 4 3 158 -- -- -- 0.6 0.4 159 -- -- -- 4 3 160 -- -- -- 0.2 0.3 161 -- -- -- 0.2 0.4 162 -- -- -- 0.2 0.3 163 -- -- -- 2 3 164 -- -- -- 0.2 0.1 165 -- -- -- 0.2 0.1 166 -- -- -- 4 2 167 -- -- -- 2 0.9 168 -- -- -- 4 3 169 -- -- -- 0.5 170----4 2 171 3 3 172 0. 3 0. 3 173 3 3 174 -- -- -- 0.04 0.03 0. 1 0.06 0.4 0.4 175 -- -- -- 0. 6 0.3 177-----02 0.06 0. 06 178 -- -- -- 0. 4 0.2 179-----0. 1 0.05 0. 05 180 -- -- -- 0.4 0.3 181 -- -- -- 0. 04 182-----0. 3 0.3 187 -- -- -- 0.05 0.03 188 -- -- -- 0.2 0.07 194 -- -- -- 0.06 0.04 199 -- -- -- 0. 2 0. 09 200 -- -- -- 0. 3 0.2 206 -- -- -- 0. 2 0.2 207 -- -- -- 0.4 0.2 208 -- -- -- 4 3 209 -- -- -- 2 2 210 -- -- -- 3 4 211 -- -- -- 0.6 0.3 212 -- -- -- 5 3 213 -- -- -- 3 2 2 5 215 -- -- -- 2 3 216 -- -- -- 0. 5 0.5 217 -- -- -- 4 4 218 -- -- -- 3 5 219 -- -- -- 0. 4 0.6 221 -- -- -- 2 2 222 -- -- -- 1 2 223---0. 04 0.1 224 -- -- -- 2 2 229 0. 4 230 0. 3 231 -- -- -- 0. 04 232 -- -- -- 0.3 233 -- -- -- 0. 5 239 -- -- -- 4 6 240 -- -- -- 8 8 zozo 4 242 -- -- -- 7 >10 243 -- -- -- 3 3 248 -- -- -- 3 4 249 -- -- -- >10 >10 250 -- -- -- 3 6 256 -- -- -- 4 3 257 -- -- -- 3 3 258 -- -- -- 0. 2 0.3 0.4 259 -- -- -- 0. 3 0.4 0.7 260 -- -- -- 0.2 0.1 0.2 261 -- -- -- 0.3 0.3 0. 3 262-----0. 3 0. 2 0. 5 263------2 3 4 264------0. 3 0. 3 0. 5 265------0. 3 0. 3 0. 4 266------0. 3 0. 3 0. 5 267------0. 8 0. 6

The data below shown in Table 3 summarizes the in vitro cyclin/cdk inhibition (IC50) and growth inhibition (GIso) of HeLa Cells for several reference compounds in comparison to several compounds of the current invention. The chemical structures are provided.

Table 3: In Vitro cyclin/cdk Inhibition (IC50) and Growth Inhibition (GIso) of HeLa Cells For Reference Compounds in Comparison to Several Compounds of the Current Invention. Compound Structure ICSa tCso ICso GIso HeLa CyclinA/cdk2 CycUnE/cdk2 CyclinB/cdkl Cells (tiM) (tiM) (tiM) (M) Olomoucine 0. 5-24 1-14 7-23 75 (n > 10) (n > 10) (n > 10) NH N ho I HO CH, CH Roscovitine < 2. 1 0. 04-30 4 0. 7 25 3 30 NH > 10 H3C N--, N 25 Ho N HIC H. C Flavopiridol OH O 0. 06 0. 6 0. 06 0. 18 0. 2 0. 04 (n = 2) HO 0 OH NI OH N Hz NU NEZ N N Zu H2N hic) H3c' 126 EN 0. 6 0. 06 2 2 0. 8 0. 06 0. 2 4 NH 6 NH N i N N H N NHZ CH3 Hic 74 5 2 6 0. 2 0. 01 0. 05 NH N N NU I N N El NHZ CH3 H3C 127 0. 3-2 0. 04-0. 07 0. 5-2 7-15 (n > 15) (n > 15) (n > 15) (n > 5) nu HaC N i N ho H. cl rich3 Hic 88 I w 3 4 > 10 0. 1 i 0. 05 0. 04 N NH Hic nez Ho JHI"NN H H, C HC

The following data in Tables 4,5, 6, and 7 summarize the growth inhibition properties of several compounds of the current invention and olomoucine against 60-human transformed cell lines. These data were cooperatively obtained at the National Cancer Institute in their 60-cell line growth inhibition assay according to published procedures (Boyd, M. R., "Anticancer Drug Development Guide, "Preclinical Screening, Clinical Trials, and Approval ;

Teicher, B. Ed. ; Humana Press ; Totowa, NJ, 23-42 (1997), which is hereby incorporated by reference).

Table 4: In Vitro Growth Inhibition (GIso) of NCI Human Transformed Cell Lines of Several Compounds of the Current Invention. Cancer Type Cell Line 73 GI50 (µM) 17 GI50 (µM) 33 GI50 (µM) 38 GI50 (µM) Breast BT-549 0. 25 0. 40 51. 3 0. 32 Breast HS 578T 0.10 6.31 -- -- Breast MCF7 0. 16 0. 16 5. 2 0. 20 Breast MDA-MB-231/ATCC 0. 50----0. 06 Breast MDA-MB-435 0. 25 0. 20 4. 9 0. 05 Breast MDA-N 0. 13 0. 11---- Breast NCI/ADR-RES 0. 40 0. 28 6. 3 0. 32 Breast T-47D 0. 25 0. 13 3. 9 0. 25 CNS SF-268 0. 16 0. 04 6. 3 0. 20 CNS SF-295 0.25 0. 19 7. 8 0. 50 CNS SF-539 0. 76 0. 40 89. 1 1. 26 CNS SNB-19 0. 43 0. 14 38. 0 0. 50 CNS SNB-75 0.02 0. 02---- CNS U251 0. 32 0. 40 3. 7 0. 20 Colon COLO 205 0. 28 0.05 7. 8 0. 16 Colon HCC-2998 0. 20 0. 03 > 1000 7.94 Colon HCT-116 0.20 0.16 6. 2 0. 32 Colon HCT-15 0. 18 0. 04 8. 9 0. 25 Colon HT29--0. 10 8. 9 0. 25 Colon KM12 0. 13 0. 03 4.1 0.16 Colon SW-620--0. 01 2.9 0.03 Leukemia CCRF-CEM 0. 25 0. 16 4.6 0. 20 Leukemia HL-60 (TB) 3. 2 0. 04 Leukemia K-562 0. 16 0.16 3. 1 0. 25 Leukemia MOLT-4 0. 32 0. 25 3. 8 0. 25 Leukemia RPMI-8226 0. 03 0. 03 1. 5-- Leukemia SR--0. 50 4.5 3.98 Melanoma LOX IMVI--0. 32 16.6 0. 40 Melanoma M14 0. 03 0.03 7.8 0.05 Melanoma MALME-3M 0. 27 19.95 11.7 0.25 Melanoma SK-MEL-2 0. 63 1.00 > 1000 2.00 Melanoma SK-MEL-28 0. 45 0. 12 5. 9 0. 03 Melanoma SK-MEL-5 0. 25 0. 32 16. 2 0. 32 Melanoma UACC-257 0. 16 0. 20 75. 9 0. 50 Melanoma UACC-62 0. 30 0. 27 8. 3 1. 00 Non-Small Cell A549/ATCC 0. 03 0. 03 4. 6 0. 13 Lung Non-Small Cell EKVX 0.25 2.51 6.9 0.20 Lung Non-Small Cell HOP-62 0.06 0. 20 > 1000 0.32 Lung Non-Small Cell HOP-92 1.00 1. 58 0. 32 Lung Non-Small Cell NCI-H226 0.22 0. 11 Lung Non-Small Cell NCI-H23 0. 32 0.16 26.3 0.32 Lung Non-Small Cell NCI-H322M 0.16 > 1000 38.9 0.40 Lung Non-Small Cell NCI-H460 0.40 0.41 25.7 3.16 Lung Non-Small Cell NCI-H522----4. 2-- Lung Ovarian IGROV1 0. 32 0.20 10.0 0.16 Ovarian OVCAR-3 0. 30 0. 65 > 1000 1. 00 Ovarian OVCAR-4 0. 32 0. 32 31. 6 1. 26 Ovarian OVCAR-5 0. 25 0. 26 > 1000 0. 40 Ovarian OVCAR-8--0. 13 6. 6 0. 25 Ovarian SK-OV-3 0. 95 0. 40 > 1000 3. 98 Prostate DU-145 7. 08 0.63 17.8 1.26 Prostate PC-3 0. 35 0.20 > 1000 0. 40 Renal 786-0 0. 20 0. 25 18. 6 0. 32 Renal A498 2. 88 1. 58--1. 26 Renal ACHN 0. 32 0. 40 5. 2 2. 00 Renal CAKI-1 1. 66 0.13 4. 4 0. 20 Renal RXF 393 0. 09 0. 02 13. 2 0. 13 Renal SN12C--0. 56-- Renal TK-10----83 0. 40 Renal UO-31 0. 06 0. 10 8. 1 0. 13

Table 5: In Vitro Growth Inhibition (GIso) of NCI Human Transformed Cell Lines of Several Compounds of the Current Invention. Cancer Type Cell Line 43 GI50 (µM) 48 GI50 (µM) 75 GI50 (µM) 76 GI50 (µM) Breast BT-549 4. 0 0.01 <0.01 <0. 01 Breast HS 578T -- 0.03 <0.01 <0. 01 Breast MCF7 2. 7 0. 25 < 0. 01 < 0. 01 Breast MDA-MB-231/ATCC 3.2 0.09 < 0. 01 < 0. 01 Breast MDA-MB-435 2. 1------ Breast MDA-N--0. 02 < 0. 01 < 0. 01 Breast NCI/ADR-RES 5. 2 0.12 0.48 0. 015 Breast T-47D 2. 2 0.15 < 0. 01 < 0. 01 CNS SF-268 3. 0 < 0. 01 < 0. 01 < 0. 01 CNS SF-295 4. 0 0. 24 < 0. 01 < 0. 01 CNS SF-539 3. 4 0. 38 0. 02 0.054 CNS SNB-19 5. 0 0. 02 < 0. 01 < 0. 01 CNS SNB-75--< 0. 01 < 0. 01 < 0. 01 CNS U251 2.3 0.17 < 0. 01 0.020 Colon COLO 205 1. 6 0. 03 < 0. 01 < 0. 01 Colon HCC-2998 3. 4------ Colon HCT-116 2. 1 0. 19 < 0. 01 0. 014 Colon HCT-15 3. 9 0.02 0.03 < 0. 01 Colon HT29 3. 6 < 0. 01 < 0. 01 < 0. 01 Colon KM12 2. 3 0.02 < 0. 01 < 0. 01 Colon SW-620 1. 6 < 0. 01 < 0. 01 < 0. 01 Leukemia CCRF-CEM 2. 8 0.03 < 0. 01 < 0. 01 Leukemia HL-60 (TB) 2. 1 Leukemia K-562 3.1 0. 16 < 0. 01 < 0. 01 Leukemia MOLT-4 2. 0 0.05 < 0. 01 < 0. 01 Leukemia RPMI-8226--< 0. 01 < 0. 01 < 0. 01 Leukemia SR 2. 2 0. 16 < 0. 01 < 0. 01 Melanoma LOX IMVI 3. 4 0. 19 < 0. 01 < 0. 01 Melanoma M14 2. 2 < 0. 01 < 0. 01 < 0. 01 Melanoma MALME-3M 3. 0 0.13 < 0. 01 < 0. 01 Melanoma SK-MEL-2 61. 7 0. 48 0. 02 0.112 Melanoma SK-MEL-28 2. 3 < 0. 01 < 0. 01 < 0. 01 Melanoma SK-MEL-5 2. 1 0. 17 0. 01 0. 013 Melanoma UACC-257 4.8 0.04 < 0. 01 < 0. 01 Melanoma UACC-62 3.3 0.10 0.01 0. 018 Non-Small Cell Lung A549/ATCC 4.1 < 0. 01 < 0. 01 < 0. 01 Non-Small Cell Lung EKVX 2. 8------ Non-Small Cell Lung HOP-62 3. 3 0.03 < 0.01 <0.01 Non-Small Cell Lung HOP-92 2. 6 0. 46 < 0. 01 0.017 Non-Small Cell Lung NCI-H226-------- Non-Small Cell Lung NCI-H23 4. 3 0.07 < 0. 01 < 0. 01 Non-Small Cell Lung NCI-H322M 3. 5 0. 03 < 0. 01 < 0. 01 Non-Small Cell Lung NCI-H460 3.2 0.25 < 0. 01 0.047 Non-Small Cell Lung NCI-H522--< 0. 01 < 0. 01 < 0. 01 Ovarian IGROV1 3.4 0.23 < 0. 01 < 0. 01 Ovarian OVCAR-3 9. 3 0. 17 < 0. 01 < 0. 01 Ovarian OVCAR-4 8.9 0. 20 < 0. 01 < 0. 01 Ovarian OVCAR-5 3. 6 0.16 < 0. 01 < 0. 01 Ovarian OVCAR-8 3. 9 0. 10 < 0. 01 < 0. 01 Ovarian SK-OV-3 72. 4 1. 38 0. 03 0. 051 Prostate DU-145 2. 6 0. 55 < 0. 01 0.043 Prostate PC-3 38. 9 0. 23 < 0. 01 < 0. 01 Renal 786-0 3.1 0. 25 < 0. 01 < 0. 01 Renal A498 3.0 0. 39 0. 01 < 0. 01 Renal ACHN 3. 1 0. 25 0. 02 0.025 Renal CAKI-1 3. 0------ Renal RXF 393 1. 9 < 0.01 <0.01 <0. 01 Renal SN12C--0. 03 < 0. 01 < 0. 01 Renal TK-10 3.2 0. 37 < 0. 01 0. 013 Renal UO-31 2.8 < 0. 01 0. 03 < 0. 01 Table 6: In Vitro Growth Inhibition (GIso) of NCI Human Transformed Cell Lines of Several Compounds of the Current Invention. Cancer Type Cell Line 79 GIso (llM) 87 GIso (llMj 12 GI50 ( Breast BT-549 < 0. 01 0.02 0.041 Breast HS 578T < 0. 01 < 0. 01 < 0.005 Breast MCF7 < 0. 01 0. 04 < 0. 005 Breast MDA-MB-231/ATCC < 0. 01 < 0. 01 < 0.005 Breast MDA-MB-435 < 0. 01 < 0. 01 < 0. 005 Breast MDA-N < 0. 01 0. 014 < 0. 005 Breast NCI/ADR-RES 0. 86 0.28 1.26 Breast T-47D < 0. 01 0.048 0.0088 CNS SF-268 < 0. 01 < 0. 01 < 0.005 CNS SF-295 < 0. 01 0. 047 0. 018 CNS SF-539 < 0. 01 0. 081 0. 022 CNS SNB-19 < 0. 01 0. 038 0. 016 CNS SNB-75 < 0. 01 0. 012 < 0. 005 CNS U251 < 0. 01 0.028 0.0078 Colon COLO 205 < 0. 01 < 0. 01 < 0.005 Colon HCC-2998 < 0. 01 < 0. 01 < 0.005 Colon HCT-116 < 0. 01 0. 037 0. 0089 Colon HCT-15 < 0. 01 0. 066 0.17 Colon HT29 < 0. 01 < 0. 01 < 0. 005 Colon KM12 < 0. 01 < 0. 01 < 0.005 Colon SW-620 < 0. 01 < 0. 01 < 0.005 Leukemia CCRF-CEM < 0. 01 < 0. 01 < 0.005 Leukemia HL-60 (TB) < 0. 01 < 0. 01 < 0.005 Leukemia K-562 < 0. 01 0. 024 < 0.005 Leukemia MOLT-4 < 0. 01 0. 02 < 0.005 Leukemia RPMI-8226 < 0. 01 < 0. 01 < 0.005 Leukemia SR < 0. 01 0.032 < 0.005 Melanoma LOX IMVI < 0. 01 0. 027 < 0.005 Melanoma M14 < 0. 01 < 0. 01 < 0.005 Melanoma MALME-3M < 0. 01 0. 024 0. 010 Melanoma SK-MEL-2 < 0. 01 0. 056 0. 0096 Melanoma SK-MEL-28 < 0. 01 < 0. 01 0. 01 Melanoma SK-MEL-5 < 0. 01 0.028 0.014 Melanoma UACC-257 < 0. 01 0.017 0.008 Melanoma UACC-62 < 0. 01 0.045 0.027 Non-Small Cell Lung A549/ATCC < 0. 01 < 0. 01 < 0.005 Non-Small Cell Lung EKVX < 0. 01 0. 081 0. 023 Non-Small Cell Lung HOP-62 < 0. 01 0.01 < 0.005 Non-Small Cell Lung HOP-92 < 0. 01 0.088 0.011 Non-Small Cell Lung NCI-H226 < 0. 01 0.0. 052 0. 021 Non-Small Cell Lung NCI-H23 < 0. 01 0.022 < 0.005 Non-Small Cell Lung NCI-H322M < 0. 01 0. 021 < 0.005 Non-Small Cell Lung NCI-H460 < 0. 01 0. 22 0. 015 Non-Small Cell Lung NCI-H522 < 0. 01 < 0. 01 < 0.005 Ovarian IGROVI < 0. 01 0.052 0.013 Ovarian OVCAR-3 < 0. 01 0. 05 0.012 Ovarian OVCAR-4 < 0. 01 0. 048 < 0. 005 Ovarian OVCAR-5 < 0. 01 0. 051 0.017 Ovarian OVCAR-8 < 0. 01 0. 033 0.0076 OvarianSK-OV-3<0. 01035 0.018 Prostate DU-145 < 0. 01 0. 22 0.017 Prostate PC-3 < 0. 01 0. 018 < 0. 005 Renal 786-0 < 0. 01 0. 047 0. 0065 Renal A498 < 0. 01 0. 10 0. 016 Renal ACHN < 0. 01 0. 19 0.039 Renal CAKI-1 < 0. 01 0. 064 0.038 Renal RXF 393 < 0. 01 0. 011 < 0. 005 Renal SN12C < 0.01 < 0.01 < 0. 005 Renal TK-10 < 0. 01 0. 029 0. 01 Renal UO-31 < 0. 01 0. 016 0. 063 Table 7: In Vitro Growth Inhibition (GIso) of NCI Human Transformed Cell Lines of Several Compounds of the Current Invention and Olomoucine. Cancer Type Cell Line 74 GIso (pM) 78 GIso (pM) 77 GIso (pM) Olomoucine GIso (µM) Breast BT-549 0.16 0.04 < 0. 01 79 Breast HS 578T < 0. 01 < 0. 01 63 Breast MCF7 < 0. 01 < 0. 01 0.03 50 Breast MDA-MB-231/ATCC < 0. 01 < 0. 01 0. 04 100 Breast MDA-MB-435------63 Breast MDA-N < 0. 01 < 0. 01 0.01 79 Breast NCI/ADR-RES 0. 24 14. 45 0. 03 100 Breast T-47D < 0. 01 0. 03 0. 01 63 CNS SF-268 <0.01 -- <0. 01 50 CNS SF-295 < 0. 01 0.21 0.04 79 CNS SF-539 0. 07--0. 22 32 CNS SNB-19 < 0. 01 < 0. 01 0. 03 63 CNS SNB-75 < 0. 01 < 0. 01 < 0. 01 25 CNS U251 < 0. 01 0. 02 0.09 50 Colon COLO 205 < 0. 01 < 0. 01 0.02 32 Colon HCC-2998--< 0. 01--63 Colon HCT-116 < 0. 01 0.03 0. 05 40 Colon HCT-15 < 0. 01 1.48 < 0. 01 40 Colon HT29 < 0. 01 < 0. 01 < 0. 01 63 Colon KM12 < 0. 01 < 0. 01 < 0. 01 40 Colon SW-620 < 0. 01 < 0. 01 < 0. 01 40 Leukemia CCRF-CEM < 0. 01--< 0. 01 40 Leukemia HL-60 (TB)--< 0. 01--40 Leukemia K-562 < 0. 01 0. 02 0.02 100 Leukemia MOLT-4 < 0. 01 < 0. 01 0.01 63 Leukemia RPMI-8226 < 0. 01 < 0. 01 < 0. 01 50 Leukemia SR < 0. 01--0. 02 25 Melanoma LOX IMVI < 0. 01 0. 04 32 Melanoma M14 < 0. 01 < 0. 01 < 0. 01 100 Melanoma MALME-3M 0. 01 0.01 0. 05 100 Melanoma SK-MEL-2 0. 06 0.02 0. 51 100 Melanoma SK-MEL-28 < 0. 01 0.01 < 0. 01 50 Melanoma SK-MEL-5 0. 06 0.10 0. 08 40 Melanoma UACC-257 < 0. 01 0.02 0. 02 79 Melanoma UACC-62 0. 04 0.03 0. 12 32 Non-Small Cell A549/ATCC < 0. 01 < 0. 01 < 0. 01 50 Lung Non-Small Cell EKVX -- 0. 05 -- 100 Lung Non-Small Cell HOP-62 < 0. 01 0.02 < 0. 01 32 Lung Non-Small Cell HOP-92 0. 03--0. 13 50 Lung Non-Small Cell NCI-H226--0. 02--50 Lung Non-Small Cell NCI-H23 < 0. 01 0.01 0.01 79 Lung Non-Small Cell NCI-H322M < 0. 01 < 0. 01 < 0. 01 63 Lung Non-Small Cell NCI-H460 < 0. 01 0.05 0.22 63 Lung Non-Small Cell NCI-H522 < 0. 01 < 0. 01 < 0. 01 40 Lung Ovarian IGROV1 < 0. 01 < 0. 01 0. 09 40 Ovarian OVCAR-3 < 0. 01 0.03 0. 02 79 Ovarian OVCAR-4 < 0. 01 0. 02 < 0 01 100 Ovarian OVCAR-5 0. 03 < 0. 01 0. 04 40 Ovarian OVCAR-8 < 0. 01 0. 02 0.02 63 Ovarian SK-OV-3 0.22 0.06 0.19 100 Prostate DU-145 0. 02 0.06 0. 13 40 Prostate PC-3 < 0. 01 < 0. 01 0.02 100 Renal 786-0 < 0. 01 0. 04 0. 03 63 Renal A498 0.03 0.03 0. 03 32 Renal ACHN 0.03 0. 32 0. 11 25 Renal CAKI-1--0. 79--32 Renal RXF 393 < 0. 01 < 0. 01 < 0. 01 20 Renal SN12C < 0. 01 < 0. 01 < 0. 01 100 Renal TK-10 < 0. 01 0. 07 0. 05 63 Renal UO-31 0. 01 0. 17 < 0. 01 32

The following data in Table 8 summarize the in vivo properties of several compounds of the current invention. These data were cooperatively obtained at the National Cancer Institute in their Hollow Fiber Assay according to published procedures (Hollingshead, M. G. , et al"In Vivo Cultivation of Tumor Cells in Hollow Fibers,"Life Sciences, 1995, 57 (2), 131-141 which is hereby incorporated by reference).

Table 8: In Vivo Evaluation of Several Compounds of the Current Invention. Compound MTD (mg/kg) IP Score SC score Cell Kill Cell Types Killed 73 100 2 0 N 17 100 8 0 Y H522 38 100 0 4 N 78 6.3 34 0 Y H23, H522, OVCAR3, SF29 79 6.3 26 6 N 86 6. 3 38 0 Y OVCAR 3, OVCAR 5, H522 87 25 30 2 Y H522 12 3.1 26 4 Y H522, MDA-MB-435 93 25 22 8 OVCAR-3 94 50 22 2 Y COL0205, OVCAR-3, H522 103 6. 3 38 6 Y OVCAR 3, OVCAR 5, H522, MDA-MB-435, SF295 109 400 18 10 Y522 112 50 18 2 Y OVCAR-3 113 200 18 4 Y H23, H522, OVCAR-3 110 50 14 0 Y OVCAR-3 124 25 28 0 Y H23, H522, OVCAR-3, MDA-MB-435

The following data in Table 9 summarize the in vivo properties of several compounds of the current invention. The protocol for the experiment is as follows. The dose-range finding study consists of four groups of three athymic mice each (four dose levels). The compound is administered on the basis of individual animal body weight. The route is intraperitoneal (IP) and the treatment schedule is daily for 14 days (qd x 14) or once every 4 days for 12 days (q4d x 3). The mice were observed for survival, and body weights recorded weekly.

The efficacy study consists of three compound-treated groups (six mice/group), a positive control-treated group (six mice), and a vehicle-treated control group of 12 mice. Test compounds were administered IP under the treatment schedules listed above (qd x 14 or q4d x 3), whereas the positive control agent (Taxol) was administered intravenously (IV) at a dosage level of 15 mg/kg/dose for five consecutive days (qd x 5). All agents were administered on the basis of individual animal body weight. Treatment began when the implanted tumors were approximately 100 mg in size (range of 65 to 200 mg). The mice were observed daily for survival. Each tumor was measured by caliper in two dimensions and converted to tumor mass using the formula for a prolate ellipsoid (a x b2/2) and assuming unit density. Tumor measurements and animal body weights were recorded twice weekly. Antitumor activity was assessed by the delay in tumor growth of the treated groups in comparison to the vehicle-treated control group, partial and complete regressions, and tumor-free survivors.

Table 9: In Vivo Evaluation of Several Compounds of the Current Invention. Compound Dose (mg/kg) Route Schedule Tumor T-C (days) free/total 78 0. 5 IP qd x 14 0/6 0.8 78 0.33 IP qd x 14 0/6 1.8 78 0.22 IP qd x 14 0/6-0.8 78 1.5 IP q4d x 3 0/6 2.5 78 1.0 IP q4d x 3 0/6 1. 1 78 0.67 IP q4d x 3 0/6 2.0 12 0. 6 IP q4d x 3 0/6 0.3 12 0.4 IP q4d x 3 0/6 0.2 12 0.27 IP q4d x 3 0/6 1.2 87 15 IP q4d x 3 0/6 2.5 87 10 IP q4d x 3 0/6 2.9 87 6.7 IP q4d x 3 0/6 1.4

Although the invention has been described in detail for the purpose of illustration, it is understood that such detail is solely for that purpose, and variations can be made therein by those skilled in the art without departing from the spirit and scope of the invention which is defined by the following claims.