AU2015203139A1 - Modulators of toll-like receptors - Google Patents

Modulators of toll-like receptors Download PDF

Info

Publication number
AU2015203139A1
AU2015203139A1 AU2015203139A AU2015203139A AU2015203139A1 AU 2015203139 A1 AU2015203139 A1 AU 2015203139A1 AU 2015203139 A AU2015203139 A AU 2015203139A AU 2015203139 A AU2015203139 A AU 2015203139A AU 2015203139 A1 AU2015203139 A1 AU 2015203139A1
Authority
AU
Australia
Prior art keywords
substituted
compound
formula
pct
heterocyclyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2015203139A
Other versions
AU2015203139B2 (en
Inventor
Manoj C. Desai
Randall L. Halcomb
Paul Hrvatin
Hon Chung Hui
Ryan Mcfadden
Paul A. Roethle
Hong Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2009/067002 external-priority patent/WO2010077613A1/en
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Priority to AU2015203139A priority Critical patent/AU2015203139B2/en
Publication of AU2015203139A1 publication Critical patent/AU2015203139A1/en
Application granted granted Critical
Publication of AU2015203139B2 publication Critical patent/AU2015203139B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

iXIts'a5eNRP4,INccAkO %)4l&V l ]K32& Provided are modulators of TLRs of Formula 11: NH2 1 a Formula I pharmaceutically acceptable salts thereof, compositions containing such compounds, and therapeutic methods that include the administration of such compounds-

Description

H:\dar\Intenvoven\NRPorbi\DCC\DAR\7909048_ Ldox-1 1/06/2015 MODULATORS OF TOLL-LIKE RECEPTORS This application is a divisional of Australian Patent Application No. 2009333559, the entire content of which is incorporated herein by reference. 5 FIELD OF THE INVENTION This application relates generally to pteridinone and pyrimidinodiazepinone derivatives and pharmaceutical compositions which selectively modulate toll-like receptors (such as TLR7), and methods of making and using such compounds. 10 BACKGROUND OF THE INVENTION The innate immune system provides the body with a first line defense against invading pathogens. In an innate immune response, an invading pathogen is recognized by a germline encoded receptor, the activation of which initiates a signaling cascade that leads to the induction of 15 cytokine expression. Innate immune system receptors have broad specificity, recognizing molecular structures that are highly conserved among different pathogens. One family of these receptors is known as Toll-like receptors (TLRs), due to their homology with receptors that were first identified and named in Drosophila, and are present in cells such as macrophages, dendritic cells, and epithelial cells. 20 There are at least ten different TLRs in mammals. Ligands and corresponding signaling cascades have been identified for some of these receptors. For example, TLR2 is activated by the lipoprotein of bacteria (e.g., E. coli.), TLR3 is activated by double-stranded RNA, TLR4 is activated by lipopolysaccharide (i.e., LPS or endotoxin) of Gram-negative bacteria (e.g., Salmonella and E. coli 0157:H7), TLR5 is activated by flagellin of motile bacteria (e.g., Listeria), TLR7 recognizes and 25 responds to imiquimod and TLR9 is activated by unmethylated CpG sequences of pathogen DNA. The stimulation of each of these receptors leads to activation of the transcription factor NF-KB, and other signaling molecules that are involved in regulating the expression of cytokine genes, including those encoding tumor necrosis factor-alpha (TNF-a), interleukin-1 (IL-1), and certain chemokines. Agonists of TLR-7 are immunostimulants and induce the production of endogenous 30 interferon-a in vivo. There are a number of diseases, disorders, and conditions linked to TLRs such that therapies using a TLR agonist are believed promising, including but not limited to melanoma, non small cell lung carcinoma, hepatocellular carcinoma, basal cell - 1 - WO 2010/077613 PCT/US2009/067002 carcinoma, renal cell carcinoma, myeloma, allergic rhinitis, asthma, COPD, ulcerative colitis, hepatic fibrosis, and viral infections such as HBV, Flaviviridae viruses, HCV, HPV, RSV, SARS, HIV, or influenza. The treatment of Flaviviridae virus infections with TLR agonists is particularly 5 promising. Viruses of the Flaviviridae family comprise at least three distinguishable genera including pestiviruses, flaviviruses, and hepaciviruses (Calisher, et al., J. Gen. Virol., 1993, 70, 37-43). While pestiviruses cause many economically important animal diseases such as bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV, hog cholera) and border disease of sheep (BDV), their importance in human disease is 10 less well characterized (Moennig, V., et a/., Adv. Vir. Res. 1992, 48, 53-98). Flaviviruses are responsible for important human diseases such as dengue fever and yellow fever while hepaciviruses cause hepatitis C virus infections in humans. Other important viral infections caused by the Flaviviridae family include West Nile virus (WNV) Janpanese encephalitis virus (JEV), tick-borne encephalitis virus, Junjin virus, Murray Valley 15 encephalitis, St Louis enchaplitis, Omsk hemorrhagic fever virus and Zika virus. Combined, infections from the Flaviviridae virus family cause significant mortality, morbidity and economic losses throughout the world. Therefore, there is a need to develop effective treatments for Flaviviridae virus infections. The hepatitis C virus (HCV) is the leading cause of chronic liver disease 20 worldwide (Boyer, N. et al. J Hepatol. 32:98-112, 2000) so a significant focus of current antiviral research is directed toward the development of improved methods of treatment of chronic HCV infections in humans (Di Besceglie, A.M. and Bacon, B. R., Scientific American, Oct.: 80-85, (1999); Gordon, C. P., et al., J. Med. Chem. 2005, 48, 1-20; Maradpour, D.; et al., Nat. Rev. Micro. 2007, 5(6), 453-463). A number of HCV 25 treatments are reviewed by Bymock et at. in Antiviral Chemistry & Chemotherapy, 11:2; 79-95 (2000). Currently, there are primarily two antiviral compounds, ribavirin, a nucleoside analog, and interferon-alpha (a) (IFN), that are used for the treatment of chronic HCV infections in humans. Ribavirin alone is not effective in reducing viral RNA levels, has significant toxicity, and is known to induce anemia. The combination of IFN 30 and ribavirin has been reported to be effective in the management of chronic hepatitis C (Scott, L. J., et al. Drugs 2002, 62, 507-556) but less than half the patients given this treatment show a persistent benefit. HCV is recognized by innate virus-sensing mechanisms that induce a rapid !FN response (Dustin, et aL, Annu. Rev. /mmunol. 2007, 25, 71-99). It is likely that the 2 WO 2010/077613 PCT/US2009/067002 sources of the IFN are, at least, the infected hepatocytes and particularly the plasmacytoid dendritic cells (pDC) that highly express TLR 7 receptors and secrete high amounts of IFN. Horsmans, et at (Hepatology, 2005, 42, 724-731), demonstrated that a once daily 7-day treatment with the TLR 7 agonist isatoribine reduces plasma virus 5 concentrations in HCV infected patients. Lee, et aL(Proc. Nati, Acad. Sct USA, 2006, 103, 1828-1833), demonstrated that TLR 7 stimulation can induce HCV immunity by both an IFN and IFN-independent mechanisms. These workers also revealed that TLR 7 is expressed in normal as well as HCV infected hepatocytes. These combined results support the conclusion that stimulation of TLR 7 receptors, such as through the 10 administration of a TLR 7 agonist, is a viable mechanism for effectively treating natural HCV infections. Given the need for more effective treatments for HCV infections, there is a need to develop safe and therapeutically effective TLR 7 agonists. 15 SUMMARY OF THE INVENTION Provided is a compound of Formula I:
NH
2 H 0 N N- Ll N N'Z 20 Formula 11 or a pharmaceutically acceptable salt or ester thereof, wherein: Y-Z is -CR 4 R-, -CR 4
R
5
-CR
4
R
5 -, -C(O)CR 4
R
5 -, -CR 4
R
5 C(O)-, -NR 8 C(O)-, -C(O)NR"-, -CR 4
R
5 S(O)2-, or -CR 5
=CR
5 -; 25 L' is -NR8-, -0-, -S-, -N(R 8 )C(O) -, -S(O) 2 -, -S(O) -, -C(O)N(R)-, -N(R 8
)S(O)
2 -, -S(O)2N(R')- or a covalent bond; R' is alkyl, substituted alkyl, haloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, heteroalkyl, substituted heteroalkyl, carbocyclyl, substituted 3 WO 2010/077613 PCT/US2009/067002 carbocyclyl, carbocyclylalkyl, substituted carbocyclylalkyl, heterocyclyl, substituted heterocyclyl, heterocyclylalkyl, or substituted heterocyclylalkyl, arylalkyl, substituted arylalkyl, heteroarylalkyl, substituted heteroarylalkyl, carbocyclylheteroalkyl, substituted carbocyclylheteroalkyl, heterocyclylheteroalkyl, substituted heterocyclylheteroalkyl, 5 arylheteroalkyl, substituted arylheteroalkyl, heteroarylheteroalkyl, or substituted heteroaryiheteroalkyl; X1 is alkylene, substituted alkylene, heteroalkylene, substituted heteroalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, carbocyclylene, substituted carbocyclylene, heterocyclylene, substituted heterocyclylene, -NR 8 -, -0-, 10 C(O)-, -S(Q)-, S(0)2-, or a bond; D is carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6
R
7 ; or D is a heterocyclyl, substituted heterocyclyl, heteroaryl or substituted heteroaryl 15 wherein said heterocyclyl, substituted heterocyclyl, heteroaryl or substituted heteroaryl comprises one to four nitrogen atoms; each L 2 is independently alkylene, substituted alkylene, heteroalkylene, substituted heteroalkylene, or a covalent bond; each R 3 is independently halogen, cyano, azido, nitro, alkyl, substituted alkyl, 20 hydroxyl, amino, heteroalkyl, substituted heteroalkyl, alkoxy, haloalkyl, haloalkoxy, -CHO, -C(O)OR, -S(O)R, -S(O) 2
R
8 ; -C(O)NRR 1 ", -N(R 9
)C(O)R
8 , carbocyclyl, substituted carbocyclyl, carbocyclylalkyl, substituted carbocyclylalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, -S(O) 2
NR
9 R' , -N(R 9
)S(O)
2
R
8 , -N(R 9 )S(0) 2 0R' 0 , OS(0) 2
NR
9
R
1 ; 25 n is 0, 1, 2, 3, 4 or 5;
R
4 and R 5 are each independently H, alkyl, substituted alkyl, haloalkyl, heteroalkyl, substituted heteroalkyl, carbocyclyl, substituted carbocyclyl, carbocyclylalkyl, substituted carbocyclylalkyl, heterocyclyl, substituted heterocyclyl, heterocyclyalkyl, substituted heterocyclylalkyl, arylalkyl, substituted arylalkyl, heteroarylalkyl, substituted 30 heteroarylalkyl, carbocyclylheteroalkyl, substituted carbocyclylheteroalkyl, heterocyclylheteroalkyl, substituted heterocyclylheteroalkyl, arylheteroalkyl, substituted arylheteroalkyl, heteroaryheteroalkyl, or substituted heteroarylheteroalkyi,, cyano, azido, OR', -C(O)H, -C(O)R 8 , -S(O)R, -S(0) 2
R
8 , -C(O)OR, or -C(O)NR 9
R
1 0 ; or
R
4 and R 5 , taken together with the carbon to which they are both attached, form a 35 carbocycle, substituted carbocycle, heterocycle or substituted heterocycle; or 4 WO 2010/077613 PCT/US2009/067002
R
4 and R5, when on the same carbon atom, taken together with the carbon to which they are attached are -C(0)- or -C(NR6)-; or two R 4 or two R 5 on adjacent carbon atoms when taken together with the carbons to which they are attached form a 3 to 6 membered carbocycle, substituted carbocycle, 5 heterocycle or substituted heterocycle; R* and R T are each independently H, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, haloalkyl, heteroalkyl, substituted heteroalkyl, carbocyclyl, substituted carbocyclyl, carbocyclylalkyl, substituted carbocyclylalkyl, heterocyclyl, substituted heterocyclyl, heterocyclylaikyl, substituted heterocyclylalkyl, 10 arylalkyl, substituted arylalkyi, heteroarylalkyl, substituted heteroarylalkyl, carbocyclylheteroalkyl, substituted carbocyclylheteroalkyl, heterocyclylheteroalkyl, substituted heterocyclylheteroalkyl, arylheteroalkyl, substituted arylheteroalkyl, heteroarylheteroalkyl, or substituted heteroaryiheteroalkyl, -C(O)H, -C(O)R 8 , -S(O)R,
-S(O)
2 R, -C(O)OR, or -C(O)NR 9
R'
0 , S(0) 2
NR
9
R'
0 ; or 15 R 6 and R, taken together with the nitrogen to which they are both attached, form a substituted or unsubstituted heterocycle, which may contain one or more additional heteroatoms selected from N, 0, P, or S; or
R
7 taken together with L2, and the N to which they are both attached, forms a substituted or unsubstituted 3 to 8 membered heterocycle which may contain one or more 20 additional heteroatoms selected from N, 0, S, or P;
R
8 is H, alkyl, substituted alkyl, haloalkyl, alkenyi, substituted alkenyl, alkynyl, substituted alkynyl, heteroalkyl, substituted heteroalkyl, carbocyclyl, substituted carbocyclyl, carbocyclylalkyl, substituted carbocyclylalkyl, heterocyclyl, substituted heterocyclyl, heterocyclylalkyl, substituted heterocyclylalkyl, arylalkyl, substituted 25 arylalkyl, heteroarylalkyl, substituted heteroarylalkyl, carbocyclyheteroalkyl, substituted carbocyclylheteroalkyl, heterocyclylheteroalkyl, substituted heterocyclylheteroalkyl, arylheteroalkyl, substituted arylheteroalkyl, heteroarylheteroalkyl, or substituted heteroarylheteroalky; and
R
9 and R' are each independently H, alkyl, substituted alkyl, alkenyl, substituted 30 alkenyl, alkynyl, substituted alkynyl, haloalkyi, heteroalkyl, substituted heteroalkyl, carbocyclyl, substituted carbocyclyl, carbocyclylalkyl, substituted carbocyclylalkyl, heterocycly, substituted heterocyclyi, heterocyclylalkyl, substituted heterocyclylalkyl, arylalkyl, substituted arylalkyl, heteroarylalkyl, substituted heteroarylalkyl, carbocyclylheteroalkyl, substituted carbocyclylheteroalkyl, heterocyclylheteroalkyl, 5 WO 2010/077613 PCT/US2009/067002 substituted heterocyclylheteroalkyl, arylheteroalkyl, substituted arylheteroalkyl, heteroarylheteroalkyl, or substituted heteroarylheteroalky; or
R
9 and R1 0 , taken together with the nitrogen to which they are both bonded, form a substituted or unsubstituted heterocycle; 5 wherein each substituted alkyl, substituted alkenyl, substituted alkynyl, substituted heteroalkyl, substituted carbocyclyl, substituted carbocyclylalkyl, substituted heterocycyl, substituted heterocyclylalkyl, substituted arylalkyl, substituted heteroarylalkyl, substituted carbocyclylheteroalkyl, substituted heterocyclyiheteroalkyl, substituted arylheteroalkyl, substituted heteroarylheteroalkyl, substituted alkylene, substituted heteroalkylene, 10 substituted alkenylene, substituted alkynylene, substituted carbocyclylene, or substituted heterocyclylene is independently substituted with one to four substituents selected from the group consisting of -halogen, -R, -0, =0, -OR, -SR, -S~, -NR 2 , -N(+)R 3 , =NR, -C(halogen) 3 , -CR(halogen) 2 , -CR 2 (halogen), -CN, -OCN, -SCN, -N=C=O, -NCS, -NO,
-NO
2 , =N 2 , -N 3 , -NRC(=O)R, -NRC(=O)OR, -NRC(=O)NRR, -C(=0)NRR, -C(=O)OR, 15 OC(=O)NRR, -OC(=O)OR, -C(=O)R, -S(=0) 2 0R, -S(=0) 2 R, -OS(=0) 2 0R, -S(=0) 2 NR, -S(=O)R, -NRS(=0) 2 R, -NRS(=0) 2 NRR, -NRS(=0) 2 0R, -OP(=OXOR) 2 , -P(=O)(OR) 2 , -P(O)(OR)(O)R, -C(=O)R, -C(=S)R, -C(=O)OR, -C(=S)OR, -C(=O)SR, -C(=S)SR, -C(=O)NRR, -C(=S)NRR, -C(=NR)NRR, and -NRC(=NR)NRR; wherein each R is independently H, alkyl, cycloalkyl, aryl, arylalkyl, or heterocyclyl. 20 While not wishing to be bound by theory, the inventors currently believe that the compounds of Formula Il are agonists of TLR-7 and may also be agonists of other TLRs. Another aspect of the present invention includes a method for treating a viral infection comprising administering a therapeutically effective amount of a compound of Formula If. The compound is administered to a human subject in need thereof, such as a 25 human being who is infected with a virus of the Flaviviridae family, such as hepatitis C virus. In one embodiment, the viral infection is acute or chronic HCV infection. In one embodiment, the treatment results in one or more of a reduction in viral load or clearance of RNA. Another aspect of the present invention includes the use of a compound of 30 Formula Il for the manufacture of a medicament for the treatment of a viral infection. Another aspect of the present invention includes a compound of Formula 11 for the use in treating a viral infection. In one embodiment, the viral infection is acute or chronic HCV infection. In one embodiment, the treatment results in one or more of a reduction in viral load or clearance of RNA. 6 WO 2010/077613 PCT/US2009/067002 In another aspect, a method for treating Flaviviridae viral infections is provided comprising administering an therapeutically effective amount of a compound of Formula it to a patient in need thereof. The compound of Formula 11 is administered to a human subject in need thereof, such as a human being who is infected with viruses of the 5 Flaviviridae family. In another embodiment, the compound of Formula 11 is administered to a human subject in need thereof, such as a human being who is infected with a HCV virus. In one embodiment, the treatment results in the reduction of one or more of the in viral loads or clearance of RNA in the patient. In another embodiment, provided is a method of treating and/or preventing a 10 disease caused by a viral infection wherein the viral infection is caused by a virus selected from the group consisting of dengue virus, yellow fever virus, West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus, Junjin virus, Murray Valley encephalitis virus, St Louis encephalitis virus, Omsk hemorrhagic fever virus, bovine viral disarrhea virus, Zika virus and Hepatitis C virus; by administering to a subject in need 15 thereof a therapeutically effective amount of a compound of Formula 11, or a pharmaceutically acceptable salt thereof. In another aspect, provided is the use of a compound of Formula I for the manufacture of a medicament for the treatment of Flaviviridae viral infections. In another aspect, provided is a compound of Formula 11 for use in treating a Flaviviridae viral 20 infection. In one embodiment, the Flaviviridae viral infection is acute or chronic HCV infection. In one embodiment of each aspect of use and compound, the treatment results in the reduction of one or more of the viral loads or clearance of RNA in the patient. In another aspect, provided is a method for treating or preventing HCV comprising administering an effective amount of a compound of Formula 11 to a patient in need 25 thereof. In another aspect, provided is the use of a compound of the present invention for the manufacture of a medicament for the treatment or prevention of HCV. In another aspect, provided is a pharmaceutical composition comprising a compound of Formula It and one or more pharmaceutically acceptable carriers or excipients. The pharmaceutical composition of Formula il may further comprise one or 30 more additional therapeutic agents. The one or more additional therapeutic agent may be, without limitation, selected from: interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, nucleoside or nucleotide inhibitors of HCV NS5B polymerase, non-nucleoside inhibitors of HCV NS5B polymerase, HCV NS5A inhibitors, TLR-7 agonists, cyclophilin inhibitors, HCV IRES 7 WO 2010/077613 PCT/US2009/067002 inhibitors, pharmacokinetic enhancers, and other drugs for treating HCV, or mixtures thereof. In another aspect, provided is a method for the treatment or prevention of the symptoms or effects of an HCV infection in an infected animal which comprises 5 administering to, i.e. treating, said animal with a pharmaceutical combination composition or formulation comprising an effective amount of a Formula II compound, and a second compound having anti-HCV properties. In another embodiment, provided are compounds of Formula Ii and pharmaceutically acceptable salts thereof and all racemates, enantiomers, 10 diastereomers, tautomers, polymorphs, pseudopolymorphs and amorphous forms thereof. In another aspect, provided are processes and novel intermediates disclosed herein which are useful for preparing Formula 11 compounds. In other aspects, novel methods for synthesis, analysis, separation, isolation, 15 purification, characterization, and testing of the compounds of Formula 11 are provided. The present invention includes combinations of aspects and embodiments, as well as preferences, as herein described throughout the present specification. 20 DETAILED DESCRIPTION Reference will now be made in detail to certain claims of the invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated claims, it will be understood that they are not intended to limit the invention to those claims. On the contrary, the invention 25 is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope of the present invention as defined by the claims. All documents referenced herein are each incorporated by reference in their entirety for all purposes. In one embodiment of Formula iI, L' is -NR 8 -. In another embodiment of Formula 30 11, L' is -O-. In another embodiment of Formula Ii, L' is -S-. In another embodiment of Formula 1l, L' is -N(R 8 )C(O)-. In another embodiment of Formula 11, L' is -S(O)-. In another embodiment of Formula 1l, L 1 is -S(0) 2 -. In another embodiment of Formula II, Ll is a covalent bond. In another embodiment of Formula 11, L' is -C(O)N(R)-. In another embodiment of Formula lI, L' is -N(R8)S(O)r. In another embodiment of Formula 11, L' is 35 -S(O) 2
N(R)
8 WO 2010/077613 PCT/US2009/067002 In one embodiment of Formula 11, R' is alkyl. In another embodiment of Formula II, R' is substituted alkyl. In another embodiment of Formula 11, R' is heteroalkyl. In another embodiment of Formula 11, R 1 is substituted heteroalkyl. In another embodiment of Formula 11, X 1 is alkylene. In another embodiment of 5 Formula 11, X 1 is substituted alkylene. In another embodiment of Formula 11, X' is heteroalkylene. In another embodiment of Formula Ii, X' is substituted heteroalkylene. In one embodiment of Formula 11, X' is C-C 6 alkylene. In another embodiment of Formula 11, X1 is substituted C-C6 alkylene. In another embodiment of Formula 11, X' is Cl-Ca heteroalkylene. In another embodiment of Formula 1I, X1 is substituted C1C6 10 heteroalkylene. In another embodiment of Formula it, X1 is -CH 2 -. In one embodiment of Formula I, D is carbocyclyl, substituted carbocycyl, heterocyclyl or substituted heterocyclyl wherein said carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6
R
7 . In another embodiment of Formula 11, D is a heterocyclyl or heteroaryl wherein said 15 heterocyclyl or heteroaryl comprises one to four nitrogen atoms. In another embodiment of Formula 1l, D is a 3- to 12-membered carbocyclyl or 3: to 12-membered heterocyclyl wherein said carbocyclyl or heterocyclyl is substituted with -L 2
-NR
6
R
T
. In another embodiment of Formula 11, D is phenyl, biphenyl or pyridinyl wherein said phenyl, biphenyl or pyridinyl is substituted with -L 2
-NR
6 R . In another embodiment of Formula Hi, D is a 20 heterocyclyl, substituted heterocycly, heteroaryl or substituted heteroaryl wherein said heterocyclyl, substituted heterocyclyl, heteroaryl or substituted heteroaryl comprises one to four nitrogen atoms. In another embodiment of Formula 11, D is a heterocyclyl, substituted heterocyclyl, heteroaryl or substituted heteroaryl wherein said heterocyclyl, substituted heterocyclyl, heteroaryl or substituted heteroaryl is optionally substituted 25 pyridinyl, optionally substituted piperidinyl, optionally substituted piperazinyl or optionally substituted 1,2,3,4-tetrahydroisoquinolinyl, In one embodiment of Formula 11, D is carbocyclyl, substituted carbocyclyl, heterocycly or substituted heterocyclyl wherein said carbocycly, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6
R
7 and R' 30 and R independently are H, alkyl, heteroalkyl, or, together with the nitrogen atom to which they are attached, form a substituted or unsubstituted heterocyclyl. In another embodiment of Formula 11, D is carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocycyl is substituted with one or two -L 2
-NR
6
R
7 and R 6 and R 7 taken 35 together with the nitrogen to which they are attached form a 4- to 1 0-membered mono- or 9 WO 2010/077613 PCT/US2009/067002 bicyclic, saturated, partially saturated, or unsaturated ring containing from 0 to 3 additional heteroatoms selected from N, 0, or S. In another embodiment of Formula 11, D is carbocycly, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said carbocyclyl, substituted carbocycly, heterocyclyl or substituted heterocyclyl is 5 substituted with one or two -L 2
-NR
6 R and R 7 taken together with L 2 , and the N to which they are both attached, forms a substituted or unsubstituted 3 to 8 membered heterocycle which may contain one or more additional heteroatoms selected from N, 0, S, or P. In one embodiment of Formula Ii, -Y-Z- is -CR 4
R
5 -. In another embodiment of Formula 11, -Y-Z- is -CR 4
R
5
-CR
4
R
6 -. In another embodiment of Formula 11, -Y-Z- is 10 CR 4
R
5 - wherein each R4 or R 5 is independently H or C-C 6 alkyl. In another embodiment of Formula 11, -Y-Z- is -CH 2 -. In another embodiment of Formula 11, -Y-Z- is -(CH 2
)
2 -. In another embodiment of Formula 11, -Y-Z- is -C(O)-. In one embodiment of Formula 11, -Y-Z- is -CR 4 R- or -CR 4
R
5
-CR
4
R
5 - and D is carbocyclyt, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said 15 carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6 R7. In another aspect of this embodiment, D is a 3- to 12 membered carbocyclyl or 3- to 12-membered heterocyclyl wherein said carbocyclyl or heterocyclyl is substituted with -L 2
-NR
6 R'. In another aspect of this embodiment, D is phenyl, biphenyl or pyridinyl wherein said phenyl, biphenyl or pyridinyl is substituted with 20 L 2
-NR
6
R
7 . In another aspect of this embodiment, R 6 and R 7 independently are H, alkyl, heteroalkyt, or, together with the nitrogen atom to which they are attached, form a substituted or unsubstituted heterocycyl. In another aspect of this embodiment, RC and
R
7 taken together with the nitrogen to which they are attached form a 4- to 10-membered mono- or bicyclic, saturated, partially saturated, or unsaturated ring containing from 0 to 3 25 additional heteroatoms selected from N, 0, or S. In another aspect of this embodiment,
R
7 taken together with L2, and the N to which they are both attached, forms a substituted or unsubstituted 3 to 8 membered heterocycle which may contain one or more additional heteroatoms selected from N, 0, S, or P. In another aspect of this embodiment, each of
R
6 and R 7 independently is H, alkyl, or heteroaryl. In another aspect of this embodiment, 30 R 6 and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, 0 or S. In another aspect of this embodiment, L' is -NH- or -0-. In another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclylalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted 35 carbocyclylalkyl. 10 WO 2010/077613 PCT/US2009/067002 In one embodiment of Formula 11, -Y-Z- is -CR 4
R
5 - or -CR 4
R
5
-CR
4
R
5 - and D is a heterocyclyl or heteroaryl wherein said heterocyclyl or heteroaryl comprises one to four nitrogen atoms. In another aspect of this embodiment, D is optionally substituted pyridinyl, optionally substituted piperidinyl, optionally substituted piperazinyl or optionally 5 substituted 1,2,3,4-tetrahydroisoquinolinyl. In another aspect of this embodiment, L' is NH- or -0-. In another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclylalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl. In one embodiment of Formula 11, -Y-Z- is -CR 4 R- wherein each R 4 or R 5 is 10 independently H or CH 3 and D is carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6 R. In another aspect of this embodiment, D is a 3- to 12-membered carbocyclyl or 3- to 12-membered heterocyclyl wherein said carbocyclyl or heterocycly is substituted with -L 2
-NR
6
R
7 . in 15 another aspect of this embodiment, D is phenyl, biphenyls or pyridinyl wherein said phenyl, biphenyls or pyridinyl is substituted with -L 2
-NR
6
R
T
. in another aspect of this embodiment,
R
6 and R' independently are H, alkyl, heteroalkyl, or, together with the nitrogen atom to which they are attached, form a substituted or unsubstituted heterocyclyl. In another aspect of this embodiment, R 6 and R 7 taken together with the nitrogen to which they are 20 attached form a 4- to 1 0-membered mono- or bicyclic, saturated, partially saturated, or unsaturated ring containing from 0 to.3 additional heteroatoms selected from N, 0, or S. In another aspect of this embodiment, R 7 taken together with L 2 , and the N to which they are both attached, forms a substituted or unsubstituted 3 to 8 membered heterocycle which may contain one or more additional heteroatoms selected from N, 0, S, or P. In 25 another aspect of this embodiment, each of R6 and R' independently is H, alkyl, or heteroaryl. In another aspect of this embodiment, R 6 and R t aken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, 0 or S. In another aspect of this embodiment, L' is -NH- or -0-. In another aspect of this embodiment, R' is alkyl, 30 substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclylalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl. In one embodiment of Formula 11, -Y-Z- is -CR 4
R
5 - wherein each R 4 or R 5 is independently H or CH 3 and D is a heterocyclyl or heteroaryl wherein said heterocyclyl or heteroaryl comprises one to four nitrogen atoms. In another aspect of this embodiment, 35 D is optionally substituted pyridinyl, optionally substituted piperidiny, optionally I I WO 2010/077613 PCT/US2009/067002 substituted piperazinyl or optionally substituted 1,2,3,4-tetrahydroisoquinolinyl. In another aspect of this embodiment, L' is -NH- or -O-. In another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalky, heterocyclyalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl, 5 In one embodiment of Formula 11, -Y-Z- is -CR 4
R
5 - wherein R 4 and R 5 taken together with the carbon to which they are attached is -C(O)-and D is carbocycfyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6
R
7 . In another aspect of this embodiment, D is a 3- to 12-membered 10 carbocyclyl or 3- to 12-membered heterocyclyl wherein said carbocyclyl or heterocyclyl is substituted with -L 2
-NR
6
R
7 . In another aspect of this embodiment, D is phenyl, biphenyl or pyridinyl wherein said phenyl, biphenyl or pyridinyl is substituted with -L 2
-NR
6
R
7 . In another aspect of this embodiment, R 6 and R 7 independently are H, alkyl, heteroalkyl, or, together with the nitrogen atom to which they are attached, form a substituted or 15 unsubstituted heterocyclyl. In another aspect of this embodiment, R 6 and R 7 taken together with the nitrogen to which they are attached form a 4- to 10-membered mono- or bicyclic, saturated, partially saturated, or unsaturated ring containing from 0 to 3 additional heteroatoms selected from N, 0, or S. In another aspect of this embodiment,
R
7 taken together with L 2 , and the N to which they are both attached, forms a substituted 20 or unsubstituted 3 to 8 membered heterocycle which may contain one or more additional heteroatoms selected from N, 0, S, or P. In another aspect of this embodiment, each of
R
6 and R 7 independently is H, alkyl, or heteroaryl. In another aspect of this embodiment,
R
6 and R taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from 25 N, 0 or S. In another aspect of this embodiment, L' is -NH- or -0-. In another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclyalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl, In one embodiment of Formula 11, -Y-Z- is -CR 4
R
5 - wherein R 4 and R 5 taken 30 together with the carbon to which they are attached is -C(O)- and D is a heterocyclyl or heteroaryl wherein said heterocyclyl or heteroaryl comprises one to four nitrogen atoms. In another aspect of this embodiment, D is optionally substituted pyridinyl, optionally substituted piperidinyl, optionally substituted piperazinyl or optionally substituted 1,2,3,4 tetrahydroisoquinoinyl. In another aspect of this embodiment, L' is -NH- or -O-. In 35 another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted 12 WO 2010/077613 PCT/US2009/067002 heteroalkyl, heterocyclylalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl. In one embodiment of Formula 11, -Y-Z- is -CH 2
CH
2 - and D is carbocyclyl, substituted carbocyclyl, heterocyclyl or substituted heterocyclyl wherein said carbocycyl, 5 substituted carbocyclyl, heterocyclyl or substituted heterocyclyl is substituted with one or two -L 2
-NR
6 R 7 In another aspect of this embodiment, D is a 3- to 12-membered carbocyclyl or 3- to 12-membered heterocyclyl wherein said carbocycly or heterocyclyl is substituted with -L 2
-NR
6
R
T
. In another aspect of this embodiment, D is phenyl, biphenyl or pyridinyl wherein said phenyl, biphenyl or pyridinyl is substituted with -L 2
-NR
6 R'. In 10 another aspect of this embodiment, R 6 and R independently are H, alkyl, heteroalkyl, or, together with the nitrogen atom to which they are attached, form a substituted or unsubstituted heterocyclyl. In another aspect of this embodiment, R6 and R 7 taken together with the nitrogen to which they are attached form a 4- to 1 0-membered mono- or bicyclic, saturated, partially saturated, or unsaturated ring containing from 0 to 3 15 additional heteroatoms selected from N, 0, or S. in another aspect of this embodiment,
R
7 taken together with L 2 , and the N to which they are both attached, forms a substituted or unsubstituted 3 to 8 membered heterocycle which may contain one or more additional heteroatoms selected from N, 0, S, or P. In another aspect of this embodiment, each of
R
6 and R 7 independently is H, alkyl, or heteroaryl. In another aspect of this embodiment, 20 R 6 and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, 0 or S. In another aspect of this embodiment, L' is -NH- or -0- In another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclylalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted 25 carbocyclylalkyl. In one embodiment of Formula 11, -Y-Z- is -CH 2
CH
2 - and D is a heterocyclyl or heteroaryl wherein said heterocyclyl or heteroaryl comprises one to four nitrogen atoms. In another aspect of this embodiment, D is optionally substituted pyridinyt, optionally substituted piperidinyl, optionally substituted piperazinyl or optionally substituted 1,2,3,4 30 tetrahydroisoquinolinyl. In another aspect of this embodiment, L' is -NH- or -0-. In another aspect of this embodiment, R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclylalkyl, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl. In one embodiment, the compound of Formula II is represented by Formula Ja: 13 WO 2010/077613 PCT/US2009/067002
NH
2 H NN NO N Ri R4 L' N N R5 D [a or a pharmaceutically acceptable salt thereof, wherein: L' is --NH- or -O-; 5 R' is alkyl, substituted alkyl, heteroalkyl, substituted heteroalkyl, heterocyclylalkyt, substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocyclylalkyl; each of R4 and R5 independently is H or Ce-Ce alkyl or R4 and R5 taken together with the carbon to which they are attached is -C(O)-; X' is Cj-Cs alkylene, Cr-C6 heteroalkylene or Cr-Cs substituted heteroalkylene; 10 D is phenyl, biphenyls or pyridinyl, wherein said phenyl, biphenyls or pyridinyl is substituted with -L2-NR6R7 ; or D is pyridinyl, piperidinyl, piperazinyl or 1,2,3,4-tetrahydroisoquinolinyl; n is 0 or 1; R3 is halogen, cyano, alkyl, carbocyclyl, carbocyclylalkyi, haloalkyl, -C(O)OR", 15 C(O)NR"R'o or -CHO; L 2 is Cl-Ce alkylene or a covalent bond; each of R 6 and R 7 independently is H, alkyl, or heteroaryl;or R 6 and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 20 heteroatoms selected from N, O or S. In one embodiment of Formula la, each of R4 and R' independently is H or Cr-C6 aikyl. In another embodiment of Formula la, each of R4 and R5 is H. In another embodiment of Formula la, R4 and R5 taken together with the carbon to which they are attached is -C(O)-. In another embodiment of Formula la, L' is -O-. In another 25 embodiment of Formula la, L' is -NH-. In another embodiment of Formula la, X, is C C6 alkylene. In another embodiment of Formula ia, X' is Cr-C6 heteroalkylene. In another embodiment of Formula la, X1 is Cr-Ce substituted heteroalkylene. In another embodiment of Formula la, X1 is -CHr-. In another embodiment of Formula la, D is 14 WO 2010/077613 PCT/US2009/067002 phenyl, biphenyl or pyridinyl, wherein said phenyl, biphenyl or pyridinyl is substituted with
-L
2
-NRR
7 . In another embodiment of Formula la, D is pyridinyl, piperidinyl, piperazinyl or 1,2,3,4-tetrahydroisoquinolinyl. In another embodiment of Fromula la, L2 is -CH 2 -. In another embodiment of Formula la, each of R' and R' independently is H, alky, or 5 heteroaryl. In another embodiment of Formula la, R and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, 0 or S. In one embodiment of Formula la, each of R and R 5 independently is H or CH 3 and D is phenyl, biphenyl or pyridinyl, wherein said phenyl, biphenyl or pyridinyl is 10 substituted with -L 2 -NRR. In another aspect of this embodiment, each of R and R' independently is H, alkyl, or heteroaryl. In another aspect of this embodiment, Re and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, o or S. In another aspect of this embodiment, L 2 is -CH 2 -. In another aspect of this 15 embodiment, X1 is -CH 2 -. In another aspect of this embodiment, L is -0-. In another aspect of this embodiment, L 1 is -NH-, In one embodiment of Formula la, each of R4 and R' independently is H or CH 3 and D is pyridinyl, piperidinyl, piperazinyl or 1,2,3,4-tetrahydroisoquinolinyl. in another aspect of this embodiment, X1 is -CH 2 -. In another aspect of this embodiment, X 1 is C 20 Ce alkylene. In another aspect of this embodiment, X 1 is C1C6 heteroalkylene. In another aspect of this embodiment, X' is Cl-r substituted heteroalkylene. In another aspect of this embodiment, L' is -0-. In another aspect of this embodiment, L' is -NH-. In one embodiment of Formula Ia, R' and R5 taken together with the carbon to which they are attached is -C(O)- and D is phenyl, biphenyl or pyridinyl, wherein said 25 phenyl, biphenyl or pyridinyl is substituted with -L 2
-NR
6 R. In another aspect of this embodiment, each of R 8 and R' independently is H, alkyl, or heteroaryl. In another aspect of this embodiment, R 6 and R7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, 0 or S. In another aspect.of this embodiment, L2 is -CH-. 30 In another aspect of this embodiment, X' is -CH 2 -. In another aspect of this embodiment, L' is -0-. In another aspect of this embodiment, Ll is -NH-. in one embodiment of Formula la, R 4 and R5 taken together with the carbon to which they are attached is -C(O)- and D is pyridinyl, piperidinyl, piperazinyl or 1,2,3,4 tetrahydroisoquinolinyl. In another aspect of this embodiment, X' is -CH 2 -. In another 35 aspect of this embodiment, X 1 is Cl-Cs alkylene. In another aspect of this embodiment, X 15 WO 2010/077613 PCT/US2009/067002 is Cl-Cs heteroalkylene. In another aspect of this embodiment, X 1 is 0 1
-C
6 substituted heteroalkylene. In another aspect of this embodiment, Ll is -0-. In another aspect of this embodiment, L' is -NH-. In one embodiment, the compound of Formula Il is represented by Formula la:
NH
2 H 0 N N R4, R1- R 5 L N N R 4 X' (R3)n D 5 ila or a pharmaceutically acceptable salt thereof, wherein: L' is -NH- or -0-; R' is alkyl, substituted alkyl, heteroakyl, substituted heteroalkyl, heterocyclylalkyl, 10 substituted heterocyclylalkyl, carbocyclylalkyl or substituted carbocycylalkyl; each of R 4 and R 5 independently is H or Cl-Ca alkyl or any R 4 and RW on the same carbon atom when taken together with the carbon to which they are attached is -C(O)-; X' is Cl-Cs alkylene, C-C 6 heteroalkylene or C-C 6 substituted heteroalkylene; 15 D is phenyl, biphenyl or pyridinyl, wherein said phenyl, biphenyl or pyridinyl is substituted with -L.-NRR 7 ; or D is pyridinyl, piperidinyl, piperazinyl or 1,2,3,4-tetrahydroisoquinolinyl; n is 0 or 1;
R
3 is halogen, cyano, alkyl, carbocyclyl, carbocyclylalkyl, haloalkyl, -C(O)OR, 20 C(O)NR 9 R" or -CHO; L2 is C-Cs alkylene or a covalent bond; each of R4 and R' independently is H, alkyl, or heteroaryl;or
R
6 and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 25 heteroatoms selected from N, 0 or S. In one embodiment of Formula Ila, each of R 4 and R 5 independently is H or C-C6 alkyl. In another embodiment of Formula la, each of R and R 5 is H. In another embodiment of Formula Ila, L' is -0-. In another embodiment of Formula Ila, L' is -NH-. 16 WO 2010/077613 PCT/US2009/067002 In another embodiment of Formula Ila, X' is C-C, alkylene. In another embodiment of Formula Ila, X 1 is C-C6 heteroalkylene. In another embodiment of Formula Ila, X 1 is C C6 substituted heteroalkylene. In another embodiment of Formula Ila, X' is -CH 2 -. In another embodiment of Formula Ila, D is phenyl, biphenyl or pyridinyl, wherein said 5 phenyl, biphenyl or pyridinyl is substituted with -L 2
-NR
6
R
7 . In another embodiment of Formula Ila, D is pyridinyl, piperidinyl, or piperazinyl. In another embodiment of Fromula Ila, L2 is -CH 2 -. In another embodiment of Formula Ila, each of R and R 7 independently is H, alkyl, or heteroaryl. In another embodiment of Formula Ila, R6 and R 7 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 10 membered heterocycle comprising 0 to 2 heteroatoms selected from N, 0 or S. In one embodiment of Formula Ila, each of R 4 and R 5 independently is H or CH 3 and D is phenyl, biphenyi or pyridinyl, wherein said phenyl, biphenyl or pyridinyl is substituted with -L 2
-NR
6
R
7 . In another aspect of this embodiment, each of R' and R' independently is H, alkyl, or heteroaryl. In another aspect of this embodiment, R 6 and R 7 15 taken together with the nitrogen to which they are attached form a substituted or unsubstituted 4-6 membered heterocycle comprising 0 to 2 heteroatoms selected from N, o or S. In another aspect of this embodiment, L 2 is -CHr. In another aspect of this embodiment, X' is -CHr. In another aspect of this embodiment L' is -0-. In another aspect of this embodiment, Ll is -NH-. 20 In one embodiment of Formula Ila, each of R 4 and R 5 independently is H or CH 3 and D is pyridinyl, piperidinyl, piperazinyl or 1,2,3,4-tetrahydroisoquinolinyl. In another aspect of this embodiment, X 1 is -CHr. In another aspect of this embodiment, X 1 is Cr C alkylene. In another aspect of this embodiment, X 1 is Cl-C heteroalkylene. In another aspect of this embodiment, X' is Cl-C substituted heteroalkylene. In another aspect of 25 this embodiment, L 1 is -0-. In another aspect of this embodiment, L 1 is -NH-, In another embodiment, provided are compounds of Formula 11 selected from the group consisting of 17 WO 2010/077613 PCT/US2009/067002
NH
2 H
NH
2 H S N N N N N N
NH
2 H
NH
2 N N 0' N> 0 ~ ~ N No
NH
2 N H 2 H N N ON N T OI - NI N
NH
2 NH 2 H N T N , N T NH2 H NH2H ON N< 0 N 0 N N N 5o")--N 18 WO 2010/077613 PCT/US2009/067002 NH2 H NH2 H ON N 0 N N N O _ N O MO No
NH
2 H
NH
2 H N N O N N V N NN NH2 H
NH
2 H HH ON OO ON N O OMe 0 No D
NH
2 N H 2 H N NO 0 N N 0 N CNN
NH
2
HNH
2 H ON N,N N O-j N- Cf- NO ' 5I
NH
2 H NH 2 H N NO NO 0N N 0IN N SNoN 19 WO 2010/077613 PCTJUS2009/067002 H
NH
2 H I N O NO NN
NH
2
HNH
2 H N 0 N~N0 HO, HO I T N- N 0)- O' N N '1[-rN ' rN
NH
2 H NH2 N N~ N NO 0 To ~No No
NH
2
HNH
2 H N N N NO OIN N N N N H
NH
2 H NH N N 0 ~N AN N NIN N H H I
NH
2
-HNH
2 H N N_ N N N N 20 WO 2010/077613 PCT/US2009/067002
NH
2 H
NH
2 H NN O N N O H N H N O N O NH2 H NH2H N N O N N N H H NIN- N N NH2 N H NH2 H NNN 0 NN N N N O N N HH O N N O~
H
2 H
NH
2 H NN o 21 "--NN N N N N 5 N No 0 No NH2 H NH 2H N N N N N CN NCNO 5 -A- -1 WO 2010/077613 PCT/US2009/067002
NH
2
HNH
2 H N No N~ NO K N N N 0 NH2 H0
NH
2 H N NN 0 ~-ON N AN N~ N NH2 H NH2 H O NN N ~ NH N
NH
2 H NlH2 H N NNO CNN No
NH
2 H N N- NNH 2 N NN '- No F3C-,-,ON, N 50 O-'C - No 22 WO 2010/077613 PCT/US2009/067002
NH
2 H
NH
2 H N N N
F
3 CO N N O N N v No IN
NH
2 H
NH
2 H MeN N Me NO O H NN1i rN 'CrN V-N
NH
2 NH 2 H MeN N Me N' N O N N O NO N N 5N NH2 H NH2 H N N HO N N
NH
2 H
NH
2 H NN 0 N~ N 0 0 N N 0 N N ~NHN NH2 H N HH2 N N 0l:_N 0N T~ 0 0,N_ o NN~ II NO ( NN N ~K N " 0I 0) No 23 WO 2010/077613 PCT/US2009/067002
NH
2 H NoN 0 0 N N
NH
2 NN 0
NH
2 H
NH
2 H N T N N- K-C I - ON O O N O?
NH
2 H N Nf NH2H N N o O NO -- o N N K NCN
NH
2 H NH2 H N N N N 0 O N N O N N N - N N
NH
2 H NHH 0NIN' N N H - N N N H N 5~No 5 24 WO 2010/077613 PCU1US2009/067002
NH
2 H N N T0 N N HI NH2H NJ N (N N N 0
NH
2 H N NO
H
2 H -N ~-N N N~r 0 SN NN H NH2 H NH2 H NH2 H N N 0 N (N 0 N NO0 ) NXOH -"' 0 O N NX )N NX O N N "-' NH 2H NH2 H NH2 H N N 0D N 0 14N N OI N j~ ON N NN NH2 H NH2 H NH2 H N N 0 IN 0 -4 _N-_r_ N XO - N N NX 0 )--''l N 0 NN N 25 WO 2010/077613 PCT/US2009/067002 NH2 H NH2 H NO N NH2 H
N
H2 H N NO N N O ONiN O NN N NH2H NH2 H N i Nt N NO NNH HIN O O N" O N N 26 NH2 H NH2 H N t0 Nt XN~ I- N C -- Oj NN NO NH N NN N N 0 NX N N N N 26 WO 2010/077613 PCT/US2009/067002 NH2H NH2 H O NOO NO NH N
NH
2 ON OO O -CON
NH
2 H N NH2 H O NH "- ON O N "t K N
NH
2 H NN NH2 H A ' N 0 I N. N N ONNH 2 H 0 N N N O N 0 N HN 27
NH
2 H N N N 0 N 'NH2 H NHZH N 0 N -NNr O, N NO N N _IN'. N NA~ Q NLO N N 27 WO 2010/077613 PCT/US2009/067002
NH
2 H NO OHNHN' N NH NH2 H NH2 H ON N O O N O NH22H NH2H 0O N F N NH2 H N NH 0 N N 0 N O HIN, N r NI&T f N
F
3 C' N
F
3 C
NH
2 H NH2H N N 0 N N O IN A F 3 C-O N N N 1\N\
NH
2
HNH
2 H NNN N N NO 5 CI N N H
NH
2 H N 0NH 2 H N No jIN N 5 cI C 28 WO 2010/077613 PCT/US2009/067002 NH2
NH
2 NN N 0 ON N N '~ N IH CF.
3
NH
2 HH NH2 H N N N T o N No NH2 H
NH
2 H 0 N NO N N N ' o 0 I o
NHNH
2 H 02 Nl, N N N \ O N N) ON N NoN No 29 WO 2010/077613 PCT/US2009/067002
NH
2 H 0
NH
2 H O N N N N" N N N N "-No N O. NH2 H 0 NH2 H 0 N N NoN
NH
2 N N N NH2 H 0 N N N ONN N CI
NH
2 H NH2 H 0 N I-N N O N N NNo 00 30
NH
2 NNH N> NH2 N N IN IN N IN N rj CH 3 ,an 0f H 50 or a pharmaceutically acceptable salt or ester thereof. Definitions 30 WO 2010/077613 PCT/US2009/067002 Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings. The fact that a particular term or phrase is not specifically defined should not be correlated to indefiniteness or lacking clarity, but rather terms herein are used within their ordinary meaning, When trade names are used herein, 5 applicants intend to independently include the tradename product and the active pharmaceutical ingredient(s) of the tradename product. The term "treating", and grammatical equivalents thereof, when used in the context of treating a disease, means slowing or stopping the progression of a disease, or ameliorating at least one symptom of a disease, more preferably ameliorating more than 10 one symptom of a disease. For example, treatment of a hepatitis C virus infection can include reducing the HCV viral load in an HCV infected human being, and/or reducing the severity of jaundice present in an HCV infected human being. As used herein, "a compound of the invention" or "a compound of formula la or formula It or formula Ila" means a compound of formula la or It or Ila, including alternative 15 forms thereof such as, solvated forms, hydrated forms, esterified forms, or physiologically functional derivatives thereof. Compounds of the invention also include tautomeric forms thereof, e.g., tautomeric "enols" as described herein. Similarly, with respect to isolatable intermediates, the phrase "a compound of formula (number)" means a compound of that formula and alternative forms thereof. 20 "Alkyl" is hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms. For example, an alkyl group can have 1 to 20 carbon atoms (i.e, C-C 20 alkyl), 1 to 10 carbon atoms (i.e., C 1
-C
10 alkyl), or 1 to 6 carbon atoms (i.e., C-C 6 alkyl). Examples of suitable alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, CH 2
CH
3 ), 1-propyl (n_-Pr, n-propyl, -CH 2
CH
2
CH
3 ), 2-propyl (i-Pr, i-propyl, -CH(CH 3
)
2 ), 1 25 butyl (n-Bu, n-butyl, -CH 2
CH
2
CH
2
CH
3 ), 2-methyl-1 -propyl (i-Bu, i-butyl, -CH 2
CH(CH
3
)
2 ), 2-butyl (s-Bu, s-butyl, -CH(CH 3
)CH
2
CH
3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH 3
)
3 ), 1 penty (n-pentyt -CH 2
CH
2
CH
2
CH
2
CH
3 ), 2-pentyi (-CH(CH 3
)CH
2
CH
2
CH
3 ), 3-pentyl
(-CH(CH
2
CH
3
)
2 ), 2-methyl-2-butyl (-C(CH 3
)
2
CH
2
CH
3 ), 3-methyl-2-butyl
(-CH(CH
3
)CH(CH
3
)
2 ), 3-methyl-1 -butyl (-CH 2
CH
2
CH(CH
3
)
2 ), 2-methyl-1 -butyl 30 (-CH 2
CH(CH
3
)CH
2
CH
3 ), I -hexyl (-CH 2
CH
2
CH
2
CH
2
CH
2
CH
3 ), 2-hexyl
(-CH(CH
3
)CH
2
CH
2
CH
2
CH
3 ), 3-hexyl (-CH(CH 2
CH
3
)(CH
2
CH
2 CH3)), 2-methyl-2-pentyl
(-C(CH
3
)
2
CH
2
CH
2
CH
3 ), 3-methyl-2-pentyl (-CH(CH 3
)CH(CH
3
)CH
2
CH
3 ), 4-methyl-2-pentyl
(-CH(CH
3
)CH
2
CH(CH
3
)
2 ), 3-methyl-3-pentyl (-C(CH3)(CH 2
CH
3 )2), 2-methyl-3-pentyl (
CH(CH
2
CH
3
)CH(CH
3
)
2 ), 2,3-dimethyl-2-butyl (-C(CH 3
)
2
CH(CH
3
)
2 ), 3,3-dimethyl-2-butyl ( 35 CH(CH 3
)C(CH
3
)
3 , and octyl (-(CH2)rCH 3 ) 31 WO 2010/077613 PCT/US2009/067002 "Alkoxy" means a group having the formula -0-alkyl, in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom. The alkyl portion of an alkoxy group can have I to 20 carbon atoms (i.e., C1C20 alkoxy), 1 to 12 carbon atoms (i.e., C-C12 alkoxy), or 1 to 6 carbon atoms(i.e., C-Ce alkoxy). Examples of 5 suitable alkoxy groups include, but are not limited to, methoxy (-O-CH 3 or -OMe), ethoxy
(-OCH
2
CH
3 or -OEt), t-butoxy (-O-C(CH 3
)
3 or -OtBu), and the like. "Haloalkyl" is an alkyl group, as defined above, in which one or more hydrogen atoms of the alkyl group is replaced with a halogen atom. The alkyl portion of a haloalkyl group can have 1 to 20 carbon atoms (i.e., C1C2o haloalkyl), I to 12 carbon atoms(i.e., 10 C1-C12 haloalkyl), or 1 to 6 carbon atoms (i.e., C,-0e alkyl). Examples of suitable haloalkyl groups include, but are not limited to, -CF 3 , -CHF 2 , -CFH 2 , -CH 2
CF
3 , and the like. "Alkenyl" is a hydrocarbon containing normal, secondary, tertiary, or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp2 double bond. For example, an alkenyl group can have 2 to 20 carbon atoms (i.e., C2-C20 alkenyl), 2 to 12 15 carbon atoms (i.e., C2-C12 alkenyl), or 2 to 6 carbon atoms (i.e., 02-C alkenyl). Examples of suitable alkenyl groups include, but are not limited to, ethylene, vinyl (-CH=CH 2 ), allyl
(-CH
2
CH=CH
2 ), cyclopentenyl (-C 5
H
7 ), and 5-hexenyl (-CH 2
CH
2
CH
2
CH
2
CH=CH
2 ). "Alkynyf" is a hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. For 20 example, an alkynyl group can have 2 to 20 carbon atoms (i.e., C2C20 alkynyl), 2 to 12 carbon atoms (i.e., C2-C12 alkyne), or 2 to 6 carbon atoms (i.e., C2C- alkynyl). Examples of suitable alkynyl groups include, but are not limited to, acetylenic (-CCH), propargyl
(-CH
2 CCH), and the like. "Alkylene" refers to a saturated, branched or straight chain or cyclic hydrocarbon 25 radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. For example, an alkylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkylene radicals include, but are not limited to, methylene (-CH 2 -), 1,1-ethylene (-CH(CH 3 )-), 1,2-ethylene (-CH 2
CH
2 -), 1,1-propylene (-CH(CH 2
CH
3 )-), 1,2 30 propylene (-CH 2
CH(CH
3 )-), 1,3-propylene (-CH 2
CH
2
CH
2 -), 1,4-butylene (-CH 2
CH
2
CH
2
CH
2 -), and the like. "Alkenylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. For 35 example, and alkenylene group can have I to 20 carbon atoms, I to 10 carbon atoms, or 1 32 WO 2010/077613 PCT/US2009/067002 to 6 carbon atoms. Typical alkenylene radicals include, but are not limited to, 1,2-ethylene (-CH=CH-). "Alkynylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two 5 hydrogen atoms from the same or two different carbon atoms of a parent alkyne. For example, an alkynylene group can have I to 20 carbon atoms, I to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkynylene radicals include, but are not limited to, acetylene (-C=C-), propargyl (-CH 2 C-C-), and 4-pentynyl (-CH 2
CH
2
CH
2 CEC-). "Aminoalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms 10 bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an amino radical. "Amidoalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a
-NR
2 CORb group where R 3 is hydrogen or aikyl and Rb is alkyl, substituted alkyi, aryl, or 15 substituted aryl as defined herein, e.g., -(CH 2
)
2
-NHC(O)CH
3 , -(CH 2 )rNH-C(O)-CHa, and the like. "Aryl" means a monovalent aromatic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. For example, an aryl group can have 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 12 20 carbon atoms. Typical aryl groups include, but are not limited to, radicals derived from benzene (e.g., phenyl), substituted benzene, naphthalene, anthracene, biphenyl, and the like. "Arylene" refers to an aryl as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms 25 of a parent aryl. Typical arylene radicals include, but are not limited to, phenylene. "Arylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an arl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2-naphthylethan-1-y!, naphthobenzyl, 2-naphthophenylethan-1-yi and the 30 like. The arylalkyI group can comprise 6 to 20 carbon atoms, e.g., the alkyl moiety is I to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. "Arylalkenyl" refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp2 carbon atom, is replaced with an aryl radical. The aryl portion of the arylalkenyl can 35 include, for example, any of the aryl groups disclosed herein, and the alkenyl portion of 33 WO 2010/077613 PCT/US2009/067002 the arylalkenyl can include, for example, any of the alkenyl groups disclosed herein. The arylalkenyl group can comprise 6 to 20 carbon atoms, e.g., the alkenyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. "Arylalkynyl" refers to an acyclic alkynyl radical in which one of the hydrogen 5 atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also an sp carbon atom, is replaced with an aryl radical. The aryl portion of the arylalkynyl can include, for example, any of the aryl groups disclosed herein, and the alkynyl portion of the arylalkynyl can include, for example, any of the alkynyl groups disclosed herein. The arylalkynyl group can comprise 6 to 20 carbon atoms, e.g., the alkynyl moiety is I to 6 10 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. "Halogen" refers to F, CI, Br, or I. As used herein the term "haloalkyl" refers to an alkyl group, as defined herein, that is substituted with at least one halogen. Examples of branched or straight chained "haloalkyl" groups as used herein include, but are not limited to, methyl, ethyl, propyl, 15 isopropyl, n-butyl, and t-buty substituted independently with one or more halogens, for example, fluoro, chloro, bromo, and iodo. The term "haloalkyl" should be interpreted to include such substituents as perfluoroalkyl groups such as -CF 3 . As used herein, the term "halcalkoxy" refers to a group -OR, where R' is a haloalkyl group as herein defined. As non-limiting examples, haloalkoxy groups include 20 -O(CH 2 )F, -O(CH)F 2 , and -OCF 3 . The term "substituted" in reference to alkyl, aryl, arylalkyl, carbocyclyl, heterocyclyl, and other groups used herein, for example, "substituted alkyl", "substituted aryl", "substituted arylalkyl", "substituted heterocyclyl", and "substituted carbocyclyl" means a group, alkyl, alkylene, aryl, arylalkyl, heterocyclyl, carbocyclyl respectively, in 25 which one or more hydrogen atoms are each independently replaced with a non hydrogen substituent. Typical substituents include, but are not limited to, -X, -R, -0-, =0, -OR, -SR, -S-, -NR 2 , -N(+)R 3 , =NR, -CX 3 , -CRX 2 , -CR 2 X, -CN, -OCN, -SCN, -N=C=O, -NCS, -NO, -NO 2 , =N 2 , -N 3 , -NRC(=O)R, -NRC(=O)OR, -NRC(=O)NRR, -C(=O)NRR, C(=O)OR, -OC(=O)NRR, -OC(=O)OR, -C(=O)R, -S(=O) 2 0R, -S(=0) 2 R, -OS(=O) 2 0R, 30 -S(=O) 2 NR, -S(=O)R, -NRS(=0) 2 R, -NRS(=0) 2 NRR, -NRS(=0) 2 0R, -OP(=O)(OR) 2 ,
-P(=O)(OR)
2 , -P(O)(OR)(O)R, -C(=O)R, -C(=S)R, -C(=O)OR, -C(=S)OR, -C(=O)SR, -C(=S)SR, -C(=O)NRR, -C(=S)NRR, -C(=NR)NRR, -NRC(=NR)NRR, where each X is independently a halogen: F, Cl, Br, or 1; and each R is independently H, alkyl, cycloalky, aryl, arylalkyl, a heterocycle, or a protecting group or prodrug moiety. Divalent groups may 35 also be similarly substituted. 34 WO 2010/077613 PCT/US2009/067002 Those skilled in the art will recognize that when moieties such as alkyll", "aryl", "heterocycyl", etc. are substituted with one or more substituents, they could alternatively be referred to as "alkylene", "arylene", "heterocyclylene", etc. moieties (i.e., indicating that at least one of the hydrogen atoms of the parent "alkyl", "aryl", "heterocyclyl" moieties has been 5 replaced with the indicated substituent(s)). When moieties such as "alkyl", "aryl", "heterocycly", etc. are referred to herein as "substituted" or are shown diagrammatically to be substituted (or optionally substituted, e.g., when the number of substituents ranges from zero to a positive integer), then the terms "alkyl", "aryl", "heterocyclyl", etc. are understood to be interchangeable with "alkylene", "arylene", "heterocyclylene", etc. 10 "Heteroalky" refers to an alkyl group where one or more carbon atoms have been replaced with a heteroatom, such as, 0, N, or S. For example, if the carbon atom of the alkyl group which is attached to the parent molecule is replaced with a heteroatom (e.g., 0, N, or S) the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g., -OCH 3 , etc.), an amine (e,g., -NHCH 3 , -N(CH 3
)
2 , and the like), or a thioalkyl group (e.g., -SCH 3 ). If a 15 non-terminal carbon atom of the alkyl group which is not attached to the parent molecule is replaced with a heteroatom (e.g., 0, N, or S) and the resulting heteroalkyl groups are, respectively, an alkyl ether (e.g., -CH 2
CH
2 -O-CHa, etc.), an alkyl amine (e.g., -CH 2
NHCH
3 ,
-CH
2
N(CH
3
)
2 , and the like), or a thioalkyl ether (e.g.,-CHr S-CH3). If a terminal carbon atom of the alkyl group is replaced with a heteroatom (e.g., 0, N, or S), the resulting heteroalkyl 20 groups are, respectively, a hydroxyalkyl group (e.g., -CH 2
CH
2 -OH), an aminoalkyl group (e.g., -CH 2
NH
2 ), or an alkyl thiol group (e.g., -CH 2
CH
2 SH). A heteroalkyl group can have, for example, 1 to 20 carbon atoms, I to 10 carbon atoms, or I to 6 carbon atoms. A C 1
-C
6 heteroalkyl group means a heteroalkyl group having 1 to 6 carbon atoms. "Heterocycle" or "heterocycyl" as used herein includes by way of example and not 25 limitation those heterocycles described in Paquette, Leo A.; Principles of Modern Heterocyclic Chemistry (WA. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. In one specific embodiment of the invention 30 "heterocycle" includes a "carbocycle" as defined herein, wherein one or more (e.g. 1, 2, 3, or 4) carbon atoms have been replaced with a heteroatom (e.g, 0, N, P or S). The terms "heterocycle" or "heterocyclyl" includes saturated rings, partially unsaturated rings, and aromatic rings (i.e., heteroaromatic rings). Heterocycles includes aromatic and non aromatic mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro. As used 35 herein, the term "heterocycle" encompasses, but is not limited to "heteroaryl." 35 WO 2010/077613 PCT/US2009/067002 Substituted heterocyclyls include, for example, heterocyclic rings substituted with any of the substituents disclosed herein including carbonyl groups. A non-limiting example of a carbonyl substituted heterocyclyl is: N rNH 0 5 Examples of heterocycles include by way of example and not limitation pyridyl, dihydroypyridyl, tetrahyd ropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, 10 benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, azetidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H-1,2,5-thiadiazinyl, 2H,6H-1 ,5,2-dithiazinyl, thienyl, thianthrenyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathinyl, 2H pyrrolyl, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, 15 1H-indazoly, purinyl, 4H-quinolizinyl, phthalaziny, naphthyridiny, quinoxalinyl, quinazolinyi, cinnolinyl, pteridinyl, 4aH-carbazoly, carbazolyl, 9-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, 20 oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, isatinoyl, and bis tetrahydrofuranyl: 0 36 WO 2010/077613 PCT/US2009/067002 By way of example and not limitation, carbon bonded heterocycles are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an 5 oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3 pyridazinyl, 4-pyridazinyl, 5-pyridazinyt, 6-pyridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5 10 pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyt, 3-pyrazinyt, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4 thiazolyl, or 5-thiazolyl. By way of example and not limitation, nitrogen bonded heterocycles are bonded at position I of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 15 3-pyrazoline, piperidine, piperazine, indole, indoline, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or a carboline. Still more typically, nitrogen bonded heterocycles include 1-aziridyl, 1 azetedyl, I-pyrrolyl, 1-imidazolyt, 1-pyrazolyl, and 1-piperidinyl. "Heterocyclylene" refers to a heterocyclyl, as defined herein, derived by replacing 20 a hydrogen atom from a carbon atom or heteroatom of a heterocyclyl, with an open valence. Similarly, "heteroarylene" refers to an aromatic heterocyclylene. "Heterocyclylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclyl-alkylene- moiety). Typical heterocyclyl alkyl 25 groups include, but are not limited to heterocyclyl-CH 2 -, 2-(heterocyclyl)ethan-1-yl, and the like, wherein the "heterocyclyl" portion includes any of the heterocyclyl groups described above, including those described in Principles of Modern Heterocyclic Chemistry. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkyl portion of the heterocyclyl alkyl by means of a carbon-carbon bond 30 or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkyl group comprises 2 to 20 carbon atoms, e.g., the alkyl portion of the arylalkyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 carbon atoms. Examples of heterocyclylalkyls include by way of example and not limitation 5-membered sulfur, oxygen, and/or nitrogen containing 35 heterocycles such as thiazolylmethyl, 2-thiazolylethan-1-yl, imidazolylmethyl, 37 WO 2010/077613 PCT/US2009/067002 oxazolylmethyl, thiadiazolylmethyl, and the like, 6-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyridinylmethyl, pyridizylmethyl, pyrimidylmethyl, pyrazinylmethyl, and the like. 5 "Heterocyclylalkenyl" refers to an acyclic alkenyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but also a sp2 carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclyl alkenylene- moiety). The heterocyclyl portion of the heterocyclyl alkenyl group includes any of the heterocyclyl groups described herein, including those described in Principles of 10 Modern Heterocyclic Chemistry, and the alkenyl portion of the heterocyclyl alkenyl group includes any of the alkenyl groups disclosed herein. One skilled in the art will also understand that the heterocyclyl group can be attached to the alkenyl portion of the heterocyclyl alkenyl by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkenyl 15 group comprises 2 to 20 carbon atoms, e.g., the alkenyl portion of the heterocyclyl alkenyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 carbon atoms. "Heterocyclylalkynyl" refers to an acyclic alkynyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, but 20 also an sp carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyclyl alkynylene- moiety). The heterocyclyl portion of the heterocycly alkynyl group includes any of the heterocyclyl groups described herein, including those described in Principles of Modern Heterocyclic Chemistry, and the alkynyl portion of the heterocyclyl alkyny group includes any of the alkynyl groups disclosed herein. One skilled in the art will also 25 understand that the heterocyclyl group can be attached to the alkynyl portion of the heterocyclyl alkynyl by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemically stable. The heterocyclyl alkyny group comprises 2 to 20 carbon atoms, e.g., the alkynyl portion of the heterocyclyl alkynyl group comprises 1 to 6 carbon atoms and the heterocyclyl moiety comprises 1 to 14 30 carbon atoms. "Heteroaryl" refers to a monovalent aromatic heterocyclyl having at least one heteroatom in the ring. Non-limiting examples of suitable heteroatoms which can be included in the aromatic ring include oxygen, sulfur, and nitrogen. Non-limiting examples of heteroaryl rings include all of those listed in the definition of "heterocycly", including 35 pyridinyl, pyrrolyl, oxazolyl, indolyl, isoindolyl, purinyl, furanyl, thienyl, benzofuranyl, 38 WO 2010/077613 PCT/US2009/067002 benzothiophenyl, carbazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, quinolyl, isoquinolyl, pyridazyl, pyrimidyl, pyrazyl, and the like. Heteroaryl also includes monovalent aromatic heterocyclyl comprising an aryl moiety and a heteroaryl group. Non limiting examples of these heteroaryls are: N - N N NH HN 0 S,/ 5 S NN 5 "Carbocycle" or "carbocycly" refers to a saturated, partially unsaturated or aromatic ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a polycycle. Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bicyclic carbocycles have 7 to 12 10 ring atoms, e.g., arranged as a bicyclo (4,5), (5,5), (5,6) or (6,6) system, or 9 or 10 ring atoms arranged as a bicyclo (5,6) or (6,6) system. Carbocycles includes aromatic and non-aromatic mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro. Non limiting examples of monocyclic carbocycles include the cycloalkyls group such as cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent 15 3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl or aryl groups such as phenyl, and the like. Thus, "carbocycle," as used herein, encompasses but is not limited to "aryl", "phenyl" and "bipheny." "Carbocyclyiene" refers to a carbocyclyl or carbocycle as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or 20 two different carbon atoms of a parent carbocyclyl Typical carbocyclylene radicals include, but are not limited to, phenyiene. Thus, "carbocyclylene," as used herein, encompasses but is not limited to "arylene." "Carbocyclylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with 39 WO 2010/077613 PCTUS2009/067002 a carbocyclyi radical as defined above. Typical carbocyclyalkyl groups include, but are not limited to the arylalkyl groups such as benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2 naphthylethan-1 -yl, naphthobenzyl, 2-naphthophenylethan-1 -yI or the cycloalkylalkyl groups such as cyclopropylmethyl, cyclobutylethyl, cyclohexylmethyl and the like. The 5 arylalkyl group can comprise 6 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 carbon atoms and the aryl moiety is 6 to 14 carbon atoms. The cycloalkylalkyl group can comprise 4 to 20 carbon atoms, e.g., the alkyl moiety is 1 to 6 caron atoms and the cycloalkyl group is 3 to 14 carbon atoms. "Arylheteroalkyl" refers to a heteroalkyl as defined herein, in which a hydrogen 10 atom, which may be attached either to a carbon atom or a heteroatom, has been replaced with an aryl group as defined herein. The aryl groups may be bonded to a carbon atom of the heteroalkyl group, or to a heteroatom of the heteroalkyl group, provided that the resulting aryiheteroalkyl group provides a chemically stable moiety. For example, an arylheteroalkyl group can have the general formulae -arkylene-O-aryl, -alkylene-O 15 alkylene-aryl, -alkylene-NH-aryl, -alkylene-NH-alkylene-aryl, -alkylene-S-aryl, alkylene-S-alkylene-aryl, and the like. In addition, any of the alkylene moieties in the general formulae above can be further substituted with any of the substituents defined or exemplified herein. "Heteroarylalky" refers to an alkyl group, as defined herein, in which a hydrogen 20 atom has been replaced with a heteroaryl group as defined herein. Non-limiting examples of heteroaryl alkyl include -CH 2 -pyridinyl, -CH 2 -pyrrolyl, -CH 2 -oxazolyl, -CH 2 indolyl, -CH 2 -isoindoly, -CH 2 -purinyl, -CH-furanyl, -CH 2 -thienyl, -CH 2 -benzofuranyl, -CH 2 benzothiophenyl, -CH 2 -carbazolyl, -CH 2 -imidazolyl, -CH 2 -thiazolyl, -CH 2 -isoxazolyl, -CH 2 pyrazolyl, -CH 2 -isothiazoly, -CH 2 -quinoly, -CH 2 -isoquinolyl, -CH 2 -pyridazyl, -CH 2 25 pyrimidyl, -CHrpyrazyl, -CH(CH 3 )-pyridinyl, -CH(CH 3 )-pyrrolyl, -CH(CH 3 )-oxazolyl, CH(CH 3 )-indolyl, -CH(CH 3 )-isoindolyl, -CH(CH 3 )-purinyl, -CH(CH 3 )-furanyl, -CH(CH 3
)
thienyl, -CH(CH 3 )-benzofurany, -CH(CH 3 )-benzothiophenyl, -CH(CH 3 )-carbazoly, CH(CH 3 )-imidazolyl, -CH(CH 3 )-thiazoly, -CH(CH 3 )-isoxazolyl, -CH(CH 3 )-pyrazolyl, CH(CH 3 )-isothiazolyl, -CH(CH 3 )-quinolyl, -CH(CH 3 )-isoquinolyl, -CH(CH 3 )-pyridazy, 30 CH(CH3)-pyrimidyl, -CH(CH 3 )-pyrazy, and the like. The term "optionally substituted" in reference to a particular moiety of the compound of the Formulae of the invention, for example an optionally substituted aryl group, refers to a moiety having 0, 1, or more substituents. As will be appreciated by those skilled in the art, the compounds of the present 35 invention are capable of existing in solvated or hydrated form. The scope of the present 40 WO 2010/077613 PCT/US2009/067002 invention includes such forms. Again, as will be appreciated by those skilled in the art, the compounds may be capable of esterification. The scope of the present invention includes esters and other physiologically functional derivatives. The scope of the present invention also includes tautomeric forms, namely, tautomeric "enols" as herein described. 5 In addition, the scope of the present invention includes prodrug forms of the compound herein described. "Ester" means any ester of a compound in which any of the --COOH functions of the molecule is replaced by a -C(O)OR function, or in which any of the -OH functions of the molecule are replaced with a -OC(O)R function, in which the R moiety of the ester is 10 any carbon-containing group which forms a stable ester moiety, including but not limited to alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heterocyclyl, heterocyclylalkyl and substituted derivatives thereof. Esters can also include esters - as described above - of "tautomeric enois", e.g. as shown below:
NH
2
NH
2 H N OH N> NO N> O N N OOXO N N NN 15 The term "ester thereof" includes but is not limited to pharmaceutically acceptable esters thereof. The term "prodrug" as used herein refers to any compound that when administered to a biological system generates the drug substance, i.e., active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemical reaction(s), photolysis, 20 and/or metabolic chemical reaction(s). A prodrug is thus a covalently modified analog or latent form of a therapeutically active compound. One skilled in the art will recognize that substituents and other moieties of the compounds of Formula I or if should be selected in order to provide a compound which is sufficiently stable to provide a pharmaceutically useful compound which can be formulated 25 into an acceptably stable pharmaceutical composition. Compounds of Formula I or 11 which have such stability are contemplated as falling within the scope of the present invention. As will be appreciated by those skilled in the art, the compounds of the present invention may contain one or more chiral centers. The scope of the present invention includes such forms. Again, as will be appreciated by those skilled in the art, the 41 WO 2010/077613 PCT/US2009/067002 compound is capable of esterification. The scope of the present invention includes esters and other physiologically functional derivatives. The scope of the present invention also includes tautomeric forms, namely, tautomeric "enols" as herein described. In addition, the scope of the present invention includes prodrug forms of the compound herein 5 described. A compound of Formula la, Ila, or Il and its pharmaceutically acceptable salts may exist as different polymorphs or pseudopolymorphs. As used herein, crystalline polymorphism means the ability of a crystalline compound to exist in different crystal structures. Polymorphism generally can occur as a response to changes in temperature, 10 pressure, or both. Polymorphism can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility, and melting point. The crystalline polymorphism may result from differences in crystal packing (packing polymorphism) or differences in packing between different conformers of the same molecule 15 (conformational polymorphism). As used herein, crystalline pseudopolymorphism means the ability of a hydrate or solvate of a compound to exist in different crystal structures. The pseudopolymorphs of the instant invention may exist due to differences in crystal packing (packing pseudopolymorphism) or due to differences in packing between different conformers of the same molecule (conformational pseudopolymorphism). The 20 instant invention comprises all polymorphs and pseudopolymorphs of the compounds of Formula I-II and their pharmaceutically acceptable salts. A compound of Formula la, lIla, or il and its pharmaceutically acceptable salts may also exist as an amorphous solid. As used herein, an amorphous solid is a solid in which there is no long-range order of the positions of the atoms in the solid. This definition 25 applies as well when the crystal size is two nanometers or less. Additives, including solvents, may be used to create the amorphous forms of the instant invention. The instant invention comprises all amorphous forms of the compounds of Formula la, Ila, or 11 and their pharmaceutically acceptable salts. Certain of the compounds described herein contain one or more chiral centers, or 30 may otherwise be capable of existing as multiple stereoisomers. The scope of the present invention includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures. Also included within the scope of the invention are the individual isomers of the compounds represented by the formulae of the present invention, as well as any wholly or partially equilibrated mixtures thereof. The 35 present invention also includes the individual isomers of the compounds represented by 42 WO 2010/077613 PCT/US2009/067002 the formulas above as mixtures with isomers thereof in which one or more chiral centers are inverted. The term "chiral" refers to molecules which have the property of non superimposability of the mirror image partner, while the term "achiral" refers to molecules 5 which are superimposable on their mirror image partner. The term "stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. "Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different 10 physical properties, e.g., melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography. "Enantiomers" refer to two stereoisomers of a compound which are non superimposable mirror images of one another. 15 Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionarv of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In 20 describing an optically active compound, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and I or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these 25 stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic 30 mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity. The present invention includes a salt or solvate of the compounds herein described, including combinations thereof such as a solvate of a salt. The compounds of the present invention may exist in solvated, for example hydrated, as well as unsolvated 35 forms, and the present invention encompasses all such forms. 43 WO 2010/077613 PCT/US2009/067002 Typically, but not absolutely, the salts of the present invention are pharmaceutically acceptable salts. Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention. 5 Examples of suitable pharmaceutically acceptable salts include inorganic acid addition salts such as chloride, bromide, sulfate, phosphate, and nitrate; organic acid addition salts such as acetate, galactarate, propionate, succinate, lactate, glycolate, malate, tartrate, citrate, maleate, fumarate, methanesulfonate, p-toluenesulfonate, and ascorbate; salts with acidic amino acid such as aspartate and glutamate; alkali metal salts 10 such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; ammonium salt; organic basic salts such as trimethylamine salt, triethylamine salt, pyridine salt, picoline salt, dicyclohexylamine salt, and N,N' dibenzylethylenediamine salt; and salts with basic amino acid such as lysine salt and arginine salt. The salts may be in some cases hydrates or ethanol solvates. 15 Protecting Groups In the context of the present invention, protecting groups include prodrug moieties and chemical protecting groups. Protecting groups are available, commonly known and used, and are optionally used to prevent side reactions with the protected group during synthetic procedures, ie. 20 routes or methods to prepare the compounds of the invention. For the most part the decision as to which groups to protect, when to do so, and the nature of the chemical protecting group "PG" will be dependent upon the chemistry of the reaction to be protected against (e.g., acidic, basic, oxidative, reductive or other conditions) and the intended direction of the synthesis. The PG groups do not need to be, and generally are 25 not, the same if the compound is substituted with multiple PG. In general, PG will be used to protect functional groups such as carboxyl, hydroxyl, thio, or amino groups and to thus prevent side reactions or to otherwise facilitate the synthetic efficiency. The order of deprotection to yield free, deprotected groups is dependent upon the intended direction of the synthesis and the reaction conditions to be encountered, and may occur in any order 30 as determined by the artisan. Various functional groups of the compounds of the invention may be protected. For example, protecting groups for -OH groups (whether hydroxyl, carboxylic acid, phosphonic acid, or other functions) include "ether- or ester-forming groups". Ether- or ester-forming groups are capable of functioning as chemical protecting groups in the 44 WO 2010/077613 PCT/US2009/067002 synthetic schemes set forth herein. However, some hydroxyl and thio protecting groups are neither ether- nor ester-forming groups, as will be understood by those skilled in the art, and are included with amides, discussed below. A very large number of hydroxyl protecting groups and amide-forming groups and 5 corresponding chemical cleavage reactions are described in Protective Groups in Organic Synthesis, Theodora W. Greene and Peter G. M. Wuts (John Wiley & Sons, Inc., New York, 1999, ISBN 0-471-16019-9) ("Greene"). See also Kocienski, Philip J.; Protecting Groups (Georg Thieme Verlag Stuttgart, New York, 1994), which is incorporated by reference in its entirety herein. In particular Chapter 1, Protecting Groups: An Overview, 10 pages 1-20, Chapter 2, Hydroxyl Protecting Groups, pages 21-94, Chapter 3, Diol Protecting Groups, pages 95-117, Chapter 4, Carboxyl Protecting Groups, pages 118 154, Chapter 5, Carbonyl Protecting Groups, pages 155-184. For protecting groups for carboxylic acid, phosphonic acid, phosphonate, sulfonic acid and other protecting groups for acids see Greene as set forth below. Such groups include by way of example and not 15 limitation, esters, amides, hydrazides, and the like. Ether- and Ester-forming protecting groups Ester-forming groups include: (1) phosphonate ester-forming groups, such as phosphonamidate esters, phosphorothioate esters, phosphonate esters, and phosphon 20 bis-amidates; (2) carboxyl ester-forming groups, and (3) sulphur ester-forming groups, such as sulphonate, sulfate, and sulfinate. Metabolites of the Compounds of the Invention Also falling within the scope of this invention are the in vivo metabolic products of 25 the compounds described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such 30 products typically are identified by preparing a radiolabelled (e.g., C" or H 3 ) compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These 35 products are easily isolated since they are labeled (others are isolated by the use of 45 WO 2010/077613 PCT/US2009/067002 antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g.b by MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so 5 long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they possess no anti infective activity of their own. Compounds of Formula la or I1 or Ila 10 The definitions and substituents for various genus and subgenus of the present compounds are described and illustrated herein. It should be understood by one skilled in the art that any combination of the definitions and substituents described above should not result in an inoperable species or compound. "Inoperable species or compounds" means compound structures that violates relevant scientific principles (such as, for 15 example, a carbon atom connecting to more than four covalent bonds) or compounds too unstable to permit isolation and formulation into pharmaceutically acceptable dosage forms. Pharmaceutical Formulations 20 The compounds of this invention are formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in 25 the Handbook of Pharmaceutical Excipients (1986), herein incorporated by reference in its entirety. Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10. 30 While it is possible for the active ingredients to be administered alone it may be preferable to present them as pharmaceutical formulations. The formulations of the invention, both for veterinary and for human use, comprise at least one active ingredient, together with one or more acceptable carriers and optionally other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the 35 other ingredients of the formulation and physiologically innocuous to the recipient thereof. 46 WO 2010/077613 PCT/US2009/067002 The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack 5 Publishing Co., Easton, Pa.), herein incorporated by reference in its entirety. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the 10 product. Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion 15 or a water-in-oil liquid emulsion. The active ingredient may also be administered as a bolus, electuary or paste. A tablet is made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or 20 granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient. 25 For administration to the eye or other external tissues e.g., mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1% and 20% in increments of 0.1% w/w such as 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w. 30 When formulated in an ointment, the active ingredients may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with an oil-in-water cream base, If desired, the aqueous phase of the cream base may include, for example, at least 30% w/w of a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups 35 such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene 47 WO 2010/077613 PCT/US2009/067002 glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulphoxide and related analogs. 5 The oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is 10 also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations. Emulgents and emulsion stabilizers suitable for use in the formulation of the 15 invention include Tween@ 60, Span@ 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate. The choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties. The cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other 20 containers. Straight or branched chain, mono- or dibasic alkyl esters such as di isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethyhexyl patmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the 25 properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils are used. Pharmaceutical formulations according to the present invention comprise one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical 30 formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of 35 pharmaceutical compositions and such compositions may contain one or more agents 48 WO 2010/077613 PCT/US2009/067002 including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert 5 diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated 10 by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed. Formulations for oral use may be also presented as hard gelatin capsules where 15 the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin; or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil. Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients 20 include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain 25 aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as 30 sucrose or saccharin. Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth 49 WO 2010/077613 PCT/US2009/067002 herein, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid. Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture 5 with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. The pharmaceutical compositions of the invention may also be in the form of oil 10 in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and 15 condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent. 20 The pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned herein. The sterile injectable preparation may also be a sterile injectable solution or suspension 25 in a non-toxic parenteraily acceptable diluent or solvent, such as a solution in 1,3-butane diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono 30 or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral 35 administration to humans may contain approximately 1 to 1000 mg of active material 50 WO 2010/077613 PCT/US2009/067002 compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may 5 contain from about 3 to 500 pg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur. Formulations suitable for administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is preferably present in such 10 formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w. Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and 15 glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier. Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate. Formulations suitable for intrapulmonary or nasal administration have a particle 20 size for example in the range of 0.1 to 500 pm (including particle sizes in a range between 0.1 and 500 pm in increments such as 0.5 pm, 1 pm, 30 pm, 35 pm, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder 25 administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of infections as described herein. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the 30 active ingredient such carriers as are known in the art to be appropriate. Formulations suitable for parenteral administration include aqueous and non aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending 35 agents and thickening agents, 51 WO 2010/077613 PCT/US2009/067002 The formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are 5 prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient. It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art 10 having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents. Compounds of the invention can also be formulated to provide controlled release of the active ingredient to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the active ingredient. Accordingly, the invention also provided 15 compositions comprising one or more compounds of the invention formulated for sustained or controlled release. The effective dose of an active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active disease or condition, the method of delivery, and the 20 pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. The effective dose can be expected to be from about 0.0001 to about 10 mg/kg body weight per day, typically from about 0.001 to about 1 mg/kg body weight per day, more typically from about 0.01 to about 1 mg/kg body weight per day, even more typically from about 0.05 to about 0.5 mg/kg body weight per day. For 25 example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from about 0.05 mg to about 100 mg, or between about 0.1 mg and about 25 mg, or between about 0.4 mg and about 4 mg, and may take the form of single or multiple doses. In yet another embodiment, the present application discloses pharmaceutical 30 compositions comprising a compound of Formula I or 11 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or exipient. Routes of Administration One or more compounds of the invention (herein referred to as the active 35 ingredients) are administered by any route appropriate to the condition to be treated. 52 WO 2010/077613 PCT/US2009/067002 Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient. An advantage of the compounds of 5 this invention is that they are orally bioavailable and can be dosed orally. Combination Therapy In one embodiment, the compounds of the present invention are used in combination with an additional active therapeutic ingredient or agent. 10 In one embodiment, combinations of the compounds of Formula la, 11, or Ila and additional active agents may be selected to treat patients with a viral infection, for example, HBV, HCV, or HIV infection. Useful active therapeutic agents for HBV include reverse transcriptase inhibitors, such as lamivudine (Epivir@), adefovir.(Hepsera@), tenofovir (Viread@), telbivudine 15 (Tyzeka@), entecavir (Baraclude®), and Clevudine@. Other useful active therapeutic agents include immunomodulators, such as interferon alpha-2b (Intron A@), pegylated interferon alpha-2a (Pegasys@), interferon alpha 2a (Roferon®), interferon alpha N1, prednisone, predinisolone, Thymalfasin@, retinoic acid receptor agonists, 4 methylumbelliferone, Alamifovir@, Metacavir@, Albuferon@, agonists of TLRs (e.g., TLR-7 20 agonists), and cytokines. With regard to treatment for HCV, other active therapeutic ingredients or agents are interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, nucleoside or nucleotide inhibitors of HCV NS5B polymerase, non-nucleoside inhibitors of HCV NS5B polymerase, HCV NS5A inhibitors, 25 TLR-7 agonists, cyclophillin inhibitors, HCV IRES inhibitors, pharmacokinetic enhancers, and other drugs for treating HCV, or mixtures thereof. Combinations of the compounds are typically selected based on the condition to be treated, cross-reactivities of ingredients and pharmaco-properties of the combination. For example, when treating an infection (e.g., HCV), the compositions of the invention are 30 combined with other active agents (such as those described herein). Suitable active agents or ingredients which can be combined with the compounds of Formula I or 11 or a salt thereof, can include one or more compounds selected from the group consisting of: (1) interferons selected from the group consisting of pegylated rIFN-alpha 2b 35 (PEG-Intron), pegylated rIFN-alpha 2a (Pegasys), rIFN-alpha 2b (Intron A), rIFN-alpha 2a 53 WO 2010/077613 PCT/US2009/067002 (Roferon-A), interferon alpha (MOR-22, OPC-18, Alfaferone, Alfanative, Multiferon, subalin), interferon alfacon-1 (Infergen), interferon alpha-n1 (Wellferon), interferon alpha n3 (Alferon), interferon-beta (Avonex, DL-8234), interferon-omega (omega DUROS, Biomed 510), albinterferon alpha-2b (Albuferon), IFN alpha-2b XL, BLX-883 (Locteron), 5 DA-3021, glycosylated interferon alpha-2b (AVI-005), PEG-Infergen, PEGylated interferon lambda-i (PEGylated IL-29), belerofon, and mixtures thereof; (2) ribavirin and its analogs selected from the group consisting of ribavirin (Rebetol, Copegus), taribavirin (Viramidine), and mixtures thereof; (3) HCV NS3 protease inhibitors selected from the group consisting of boceprevir 10 (SCH-503034 , SCH-7), telaprevir (VX-950), TMC435350, B1-1 335, B-1230, MK-7009, VBY-376, VX-500, BMS-790052, BMS-605339, PHX-1766, AS-101, YH-5258, YH5530, YH5531, ITMN-191, and mixtures thereof; (4) alpha-glucosidase 1 inhibitors selected from the group consisting of celgosivir (MX-3253), Miglitol, UT-231B, and mixtures thereof; 15 (5) hepatoprotectants selected from the group consisting of IDN-6556, ME 3738, LB-84451, silibilin, MitoQ, and mixtures thereof; (6) nucleoside or nucleotide inhibitors of HCV NS5B polymerase selected from the group consisting of R1626, R7128 (R4048), IDX184, IDX-102, BCX-4678, valopicitabine (NM-283), MK-0608, and mixtures thereof; 20 (7) non-nucleoside inhibitors of HCV NS5B polymerase selected from the group consisting of PF-868554, VCH-759, VCH-916, JTK-652, MK-3281, VBY-708, VCH-222, A848837, ANA-598, GL60667, GL59728, A-63890, A-48773, A-48547, BC-2329, VCH 796 (nesbuvir), GSK625433, BILN-1941, XTL-2125, GS-9190, and mixtures thereof; (8) HCV NS5A inhibitors selected from the group consisting of AZD-2836 (A-831), 25 A-689, and mixtures thereof; (9) TLR-7 agonists selected from the group consisting of ANA-975, SM-360320, and mixtures thereof; (10) cyclophillin inhibitors selected from the group consisting of DEBIO-025, SCY 635, NIM81 1, and mixtures thereof; 30 (11) HCV IRES inhibitors selected from the group consisting of MCl-067, (12) pharmacokinetic enhancers selected from the group consisting of BAS-1 00, SPI-452, PF-4194477, TMC-41629, roxythromycin, and mixtures thereof; and (13) other drugs for treating HCV selected from the group consisting of thymosin alpha 1 (Zadaxin), nitazoxanide (Alinea, NTZ), BIVN-401 (virostat), PYN-17 (altirex), 35 KPE02003002, actilon (CPG-1 0101), KRN-7000, civacir, GI-5005, XTL-6865, BIT225, 54 WO 2010/077613 PCT/US2009/067002 PTX-1 11, ITX2865, TT-033i, ANA 971, NOV-205, tarvacin, EHC-18, VGX-41 0C, EMZ 702, AVI 4065, BMS-650032, BMS-791325, Bavituximab, MDX-1106 (ONO-4538), Oglufanide, VX-497 (merimepodib), and mixtures thereof, In addition, the compounds of the invention may be employed in combination with 5 other therapeutic agents for the treatment or prophylaxis of AIDS and/or one or more other diseases present in a human subject suffering from AIDS (e.g., bacterial and/or fungal infections, other viral infections such as hepatitis B or hepatitis C, or cancers such as Kaposi's sarcoma). The additional therapeutic agent(s) may be coformulated with one or more salts of the invention (e.g., coformulated in a tablet). 10 Examples of such additional therapeutic agents include agents that are effective for the treatment or prophylaxis of viral, parasitic or bacterial infections, or associated conditions, or for treatment of tumors or related conditions, include 3'-azido-3' deoxythymidine (zidovudine, AZT), 2'-deoxy-3'-thiacytidine (3TC), 2',3'-dideoxy-2',3' didehydroadenosine (D4A), 2',3'-dideoxy-2',3'-didehydrothymidine (D4T), carbovir 15 (carbocyclic 2',3'-dideoxy-2',3'-didehydroguanosine), 3-azido-2',3'-dideoxyuridine, 5 fluorothymidine, (E)-5-(2-bromovinyl)-2'-deoxyuridine (BVDU), 2-chlorodeoxyadenosine, 2-deoxycoformycin, 5-fluorouracil, 5-fluorouridine, 5-fluoro-2'-deoxyuridine, 5 trifluorom ethyl-2'-deoxyu ridi ne, 6-azauridine, 5-fluoroorotic acid, methotrexate, triacetyluridine, 1-(2'-deoxy-2'-fluoro-1-p-arabinosyl)-5-iodocytidine (FIAC), tetrahydro 20 imidazo(4,5, 1-jk)-(1,4)-benzodiazepin-2(1 H)-thione (TIBO), 2'-nor-cyclicGMP, 6 methoxypurine arabinoside (ara-M), 6-methoxypurine arabinoside 2'-O-valerate; cytosine arabinoside (ara-C), 2',3'-dideoxynucleosides such as 2',3'-dideoxycytidine (ddC), 2',3' dideoxyadenosine (ddA) and 2',3'-dideoxyinosine (ddl); acyclic nucleosides such as acyclovir, penciclovir, famciclovir, ganciclovir, HPMPC, PMEA, PMEG, PMPA, PMPDAP, 25 FPMPA, HIPMPA, HPMPDAP, (2R, 5R)-9->tetrahydro-5-(phosphonomethoxy)-2 furanyladenine, (2R, 5R)-1->tetrahydro-5-(phosphonomethoxy)-2-furanylthymine; other antivirals including ribavirin (adenine arabinoside), 2-thio-6-azauridine, tubercidin, aurintricarboxylic acid, 3-deazaneoplanocin, neoplanocin, rimantidine, adamantine, and foscarnet (trisodium phosphonoformate); antibacterial agents including bactericidal 30 fluoroquinolones (ciprofloxacin, pefloxacin and the like); aminoglycoside bactericidal antibiotics (streptomycin, gentamicin, amicacin and the like); p-lactamase inhibitors (cephalosporins, penicillins and the like); other antibacterials including tetracycline, isoniazid, rifampin, cefoperazone, claithromycin and azithromycin, antiparasite or antifungal agents including pentamidine (1,5-bis(4'-aminophenoxy)pentane), 9-deaza 35 inosine, sulfamethoxazole, sulfadiazine, quinapyramine, quinine, fluconazole, 55 WO 2010/077613 PCT/US2009/067002 ketoconazole, itraconazole, Amphotericin B, 5-fluorocytosine, clotrimazole, hexadecylphosphocholine and nystatin; renal excretion inhibitors such as probenicid; nucleoside transport inhibitors such as dipyridamole, dilazep and nitrobenzylthioinosine, immunomodulators such as FK506, cyclosporin A, thymosin a-1; cytokines including TNF 5 and TGF-p; interferons including IFN-ct, IFN-p, and IFN-y; interleukins including various interleukins, macrophage/granulocyte colony stimulating factors including GM-CSF, G CSF, M-CSF, cytokine antagonists including anti-TNF antibodies, anti-interleukin antibodies, soluble interleukin receptors, protein kinase C inhibitors and the like. Examples of suitable active therapeutic agents or ingredients which can be 10 combined with the compounds of the invention, and which have activity against HIV, include 1) HIV protease inhibitors, e.g., amprenavir, atazanavir, fosamprenavir, indinavir, lopinavir, ritonavir, lopinavir + ritonavir, nelfinavir, saquinavir, tipranavir, brecanavir, darunavir, TMC-126, TMC-1 14, mozenavir (DMP-450), JE-2147 (AG1776), AG1859, DG35, L-756423, R00334649, KNI-272, DPC-681, DPC-684, and GW640385X, DG17, 15 PPL-100, 2) a HIV non-nucleoside inhibitor of reverse transcriptase, e.g., capravirine, emivirine, delaviridine, efavirenz, nevirapine, (+) calanoride A, etravirine, GW5634, DPC 083, DPC-961, DPC-963, MIV-150, and TMC-120, TMC-278 (rilpivirine), efavirenz, BILR 355 BS, VRX 840773, UK-453,061, RDEA806, 3) a HIV nucleoside inhibitor of reverse transcriptase, e.g., zidovudine, emtricitabine, didanosine, stavudine, zalcitabine, 20 lamivudine, abacavir, amdoxovir, elvucitabine, alovudine, MIV-210, racivir ( -FTC), D d4FC, emtricitabine, phosphazide, fozivudine tidoxil, fosalvudine tidoxil, apricitibine (AVX754), amdoxovir, KP-1461, abacavir + lamivudine, abacavir + lamivudine + zidovudine, zidovudine + lamivudine, 4) a HIV nucleotide inhibitor of reverse transcriptase, e.g., tenofovir, tenofovir disoproxil fumarate + emtricitabine, tenofovir 25 disoproxil fumarate + emtricitabine + efavirenz, and adefovir, 5) a HIV integrase inhibitor, e.g., curcumin, derivatives of curcumin, chicoric acid, derivatives of chicoric acid, 3,5 dicaffeoylquinic acid, derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxylic acid, derivatives of aurintricarboxylic acid, caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin, derivatives of tyrphostin, quercetin, derivatives of 30 quercetin, S-1360, zintevir (AR-177), L-870812, and L-870810, MK-0518 (raltegravir), BMS-707035, MK-2048, BA-011, BMS-538158, GSK364735C, 6) a gp41 inhibitor, e.g., enfuvirtide, sifuvirtide, FBO06M, TRI-1144, SPC3, DES6, Locus gp4l, CovX, and REP 9, 7) a CXCR4 inhibitor, e.g., AMD-070, 8) an entry inhibitor, e.g., SP01A, TNX-355, 9) a gp120 inhibitor, e.g., BMS-488043 and BlockAide/CR, 10) a G6PD and NADH-oxidase 35 inhibitor, e.g., immunitin, 10) a CCR5 inhibitor, e.g., aplaviroc, vicriviroc, INCB9471, PRO 56 WO 2010/077613 PCT/US2009/067002 140, INCB15050, PF-232798, CCR5mAb004, and maraviroc, 11) an interferon, e.g., pegylated rIFN-alpha 2b, pegylated rlFN-alpha 2a, rIFN-alpha 2b, IFN alpha-2b XL, rIFN alpha 2a, consensus IFN alpha, infergen, rebif, iocteron, AVI-005, PEG-infergen, pegylated IFN-beta, oral interferon alpha, feron, reaferon, intermax alpha, r-IFN-beta, 5 infergen + actimmune, IFN-omega with DUROS, and albuferon, 12) ribavirin analogs, e.g., rebetol, copegus, levovirin, VX-497, and viramidine (taribavirin) 13) NS5a inhibitors, e.g., A-831 and A-689, 14) NS5b polymerase inhibitors, e.g., NM-283, valopicitabine, R1626, PSI-6130 (R1656), HIV-796, BILB 1941, MK-0608, NM-107, R7128, VCH-759, PF-868554, GSK625433, and XTL-2125, 15) NS3 protease inhibitors, e.g., SCH-503034 10 (SCH-7), VX-950 (Telaprevir), ITMN-191, and BILN-2065, 16) alpha-glucosidase 1 inhibitors, e.g., MX-3253 (celgosivir) and UT-231B, 17) hepatoprotectants, e.g., IDN 6556, ME 3738, MitoQ, and LB-84451, 18) non-nucleoside inhibitors of HIV, e.g., benzimidazole derivatives, benzo-1,2,4-thiadiazine derivatives, and phenylalanine derivatives, 19) other drugs for treating HIV, e.g., zadaxin, nitazoxanide (alinea), BIVN 15 401 (virostat), DEBIO-025, VGX-410C, EMZ-702, AVI 4065, bavituximab, oglufanide, PYN-17, KPE02003002, action (CPG-10101), KRN-7000, civacir, GI-5005, ANA-975 (isatoribine), XTL-6865, ANA 971, NOV-205, tarvacin, EHC-18, and NIM811, 19) pharmacokinetic enhancers, e.g., BAS-100 and SP1452, 20)RNAse H inhibitors, e.g., ODN-93 and ODN-1 12, 21) other anti-HIV agents, e.g., VGV-1, PA-457 (bevirimat), 20 ampligen, HRG214, cytolin, polymun, VGX-410, KD247, AMZ 0026, CYT 99007, A-221 HIV, BAY 50-4798, MDX010 (iplimumab), PBS1 19, ALG889, and PA-1050040. Again by way of example, the following list discloses exemplary HIV antivirals, with their corresponding U.S. Patent numbers, incorporated by reference with regard to the preparation of such antivirals, which can be combined with the compounds of the 25 present invention. Exemplary HIV Antivirals and Patent Numbers Ziagen (Abacavir sulfate, US 5,034,394) Epzicom (Abacavir sulfate/lamivudine, US 5,034,394) Hepsera (Adefovir dipivoxit, US 4,724,233) 30 Agenerase (Amprenavir, US 5,646,180) Reyataz (Atazanavir sulfate, US 5,849,911) Rescriptor (Delavirdine mesilate, US 5,563,142) Hivid (Dideoxycytidine; Zalcitabine, US 5,028,595) Videx (Dideoxyinosine; Didanosine, US 4,861,759) 35 Sustiva (Efavirenz, US 5,519,021) 57 WO 2010/077613 PCT/US2009/067002 Emtriva (Emtricitabine, US 6,642,245) Lexiva (Fosamprenavir calcium, US 6,436,989) Virudin; Triapten; Foscavir (Foscarnet sodium, US 6,476,009) Crixivan (Indinavir sulfate, US 5,413,999) 5 Epivir (Lamivudine, US 5 047,407) Combivir (Lamivudine/Zidovudine, US 4,724,232) Aluviran (Lopinavir) Kaletra (Lopinavir/ritonavir, US 5,541,206) Viracept (Nelfinavir mesilate, US 5,484,926) 10 Viramune (Nevirapine, US 5,366,972) Norvir (Ritonavir, US 5,541,206) Invirase; Fortovase (Saquinavir mesilate, US 5,196,438) Zerit (Stavudine, US 4,978,655) Truvada (Tenofovir disoproxil fumarate/emtricitabine, US 5,210,085) 15 Aptivus (Tipranavir) Retrovir (Zidovudine; Azidothymidine, US 4,724,232) Where the disorder is cancer, combination with at least one other anticancer therapy is envisaged. In particular, in anti-cancer therapy, combination with other anti neoplastic agent (including chemotherapeutic, hormonal or antibody agents) is envisaged 20 as well as combination with surgical therapy and radiotherapy. Combination therapies according to the present invention thus comprise the administration of at least one compound of formula (1) or a salt or solvate thereof, and the use of at least one other cancer treatment method. Preferably, combination therapies according to the present invention comprise the administration of at least one compound of formula (1) or a salt or 25 solvate thereof, and at least one other pharmaceutically active agent, preferably an anti neoplastic agent. The compound(s) of formula (I)) and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately this may occur simultaneously or sequentially in any order (including administration on different days according to the therapy regimen) and by any convenient route. The 30 amounts of the compound(s) of formula (11) and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. In one embodiment, the further anti-cancer therapy is at least one additional antineoplastic agent. Any anti-neoplastic agent that has activity versus a susceptible 35 tumor being treated may be utilized in the combination. Typical anti-neoplastic agents 58 WO 2010/077613 PCT/US2009/067002 useful include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase I inhibitors 5 such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; nonreceptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic agents; proapoptotic agents; and cell cycle signaling inhibitors. 10 Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle. Examples of anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids. Diterpenoids, which are derived from natural sources, are phase specific anti cancer agents that operate at the G 2 1M phases of the cell cycle. It is believed that the 15 diterpenoids stabilize the B-tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel. Paclitaxel, 59,20-epoxy-I ,2a,4,7B,108,13a-hexa-hydroxytax-1 1-en-9-one 4,10 20 diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes. Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et a., Yale Journal of Biology and 25 Medicine, 64:583, 1991 ; McGuire et al., Ann. Intern, Med., 11 1 :273,1989) and for the treatment of breast cancer (Holmes et aL, J. Nat. Cancer Inst., 83:1797,1991.) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. a!., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. ai., Sem. Oncol., 20:56, 1990). The compound also shows potential for the treatment of polycystic kidney disease 30 (Woo et. aL., Nature, 368:750. 1994), lung cancer and malaria. Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, RJ. et. al, Cancer Chemotherapy Pocket GuideA 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, CM. et. a., Seminars in Oncology, 3(6) p.16-23, 1995). 59 WO 2010/077613 PCT/US2009/067002 Docetaxel, (2R,3S)- N-carboxy-3-phenylisoserine,N-tef-butyi ester, 13-ester with SB- 20-epoxy-i ,2a,4,7B,105,13a-hexahydroxytax-1 1-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE@. Docetaxel is indicated for the treatment of breast cancer. Docetaxel is a semisynthetic derivative of 5 paclitaxel q.v., prepared using a natural precursor, 10- deacetyl-baccatin IlIl, extracted from the needle of the European Yew tree. Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize 10 into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine. Vinblastine, vincaleukoblastine sulfate, is commercially available as VELBAN@ as an injectable solution. Although, it has possible indication as a second line therapy of 15 various solid tumors, it is primarily indicated in the treatment of testicular cancer and various lymphomas including Hodgkin's Disease; and lymphocytic and histiocytic lymphomas. Myelosuppression is the dose limiting side effect of vinblastine. Vincristine, vincaleukoblastine, 22-oxo-, sulfate, is commercially available as ONCOVIN@ as an injectable solution. Vincristine is indicated for the treatment of acute leukemias and has 20 also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas. Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur. Vinorelbine, 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R', R)-2,3 25 dihydroxybutanedioate (1 :2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE@), is a semisynthetic vinca alkaloid. Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the 30 most common dose limiting side effect of vinorelbine. Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA. The platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor. Examples of platinum coordination complexes include, but are not 35 limited to, oxaliplatin, cisplatin and carboplatin. Cisplatin, cis-diamminedichloroplatinum, 60 WO 2010/077613 PCT/US2009/067002 is commercially available as PLATINOL@ as an injectable solution. Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer. Carboplatin, platinum, diammine [1 ,1 -cyclobutane-dicarboxylate(2-) 0,0], is commercially available as PARAPLATIN® as an injectable solution. Carboplatin 5 is primarily indicated in the first and second line treatment of advanced ovarian carcinoma. Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, 10 hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine. Cyclophosphamide, 2-[bis(2-chloroethyl)amino)tetrahydro-2H-1 3,2 15 oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN@. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias. Melphalan, 4-[bis(2-chloroethyl)amino]-L phenylalanine, is commercially available as an injectable solution or tablets as 20 ALKERAN@. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan. Chlorambucil, 4-[bis(2 chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN@ tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic 25 leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease. Busuifan, 1 4-butanediol dimethanesulfonate, is commercially available as MYLERAN@ TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Carmustine, 1 ,3-[bis(2-chloroethyl)-1 -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU@. Carmustine is 30 indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non-Hodgkin's lymphomas. Dacarbazine, 5-(3,3-dimethyl-1-triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome@. Dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the 35 second line treatment of Hodgkin's Disease. 61 WO 2010/077613 PCT/US2009/067002 Antibiotic anti-neoplasties are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death. Examples of antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as 5 dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins. Dactinomycin, also know as Actinomycin D, is commercially available in injectable form as COSMEGEN@. Dactinomycin is indicated for the treatment of Wilm's tumor and rhabdomyosarcoma. Daunorubicin, (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy-a-L lyxo- hexopyranosyl)oxy]-7,8,9, 10-tetrahydro-6,8, 1 1 -trihydroxy-1 -methoxy-5, 12 10 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME@ or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma. Doxorubicin, (8S, 10S)-10-[(3-amino-2,3,6-trideoxy-a-L lyxo-hexopyranosyl)oxy]-8- glycoloyl, 7,8,9, 10-tetrahydro-6, 8,1 1-trihydroxy-1-methoxy 15 5,12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX@ or ADRIAMYCIN RDF@. Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblasts leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Bleomycin, a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is 20 commercially available as BLENOXAN E@. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas. Topoisomerase 11 inhibitors include, but are not limited to, epipodophyllotoxins. Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake 25 plant. Epipodophyllotoxins typically affect cells in the S and G 2 phases of the cell cycle by forming a ternary complex with topoisomerase I and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide. Etoposide, 4'-demethyl epipodophyllotoxin 9[4,6-0-(R )-ethylidene-B-D- glucopyranoside], is commercially 30 available as an injectable solution or capsules as VePESID@ and is commonly known as VP-16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell rung cancers. Teniposide, 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-thenylidene-R-D glucopyranoside, is commercially available as an injectable solution as VUMON@ and is 62 WO 2010/077613 PCT/US2009/067002 commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children. Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting 5 purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows. Examples of antimetabolite anti neoplastic agents include, but are not limited to, fluorouracit, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine. 5-fluorouracil, 5-fluoro-2,4- (1 H,3H) pyrimidinedione, is commercially available as fluorouracil. Administration of 5-fluorouracil 10 leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death. 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Other fluoropyrimidine analogs include 5 fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate. 15 Cytarabine, 4-amino-1 -B-D-arabinofuranosyl-2 (I H)-pyrimidinone, is commercially available as CYTOSAR-U@ and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of 20 acute leukemia. Other cytidine analogs include 5- azacytidine and 2',2' difluorodeoxycytidine (gemcitabine). Mercaptopurine, 1 ,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL®. Mercaptopurine exhibits cell phase specificity at S- phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Mercaptopurine is indicated as a single agent or in combination with other 25 chemotherapy agents in the treatment of acute leukemia. A useful mercaptopurine analog is azathioprine. Thioguanine, 2-amino-1,7-dihydro-6H-purine-6-thione, is commercially available as TABLOID®. Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of 30 acute leukemia. Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine. Gemcitabine, 2'-deoxy-2', 2'-difluorocytidine monohydrochloride (S-isomer), is commercially available as GEMZAR@. Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1/S boundary. Gemcitabine is indicated in combination with cisplatin in the treatment of 35 locally advanced non-small cell lung cancer and alone in the treatment of locally 63 WO 2010/077613 PCT/US2009/067002 advanced pancreatic cancer. Methotrexate, N-[4[[(2,4-diamino-6-pteridinyl) methyl]methylamino] benzoyl]-L- glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or-replication through the inhibition of dyhydrofolic acid reductase 5 which is required for synthesis of purine nucleotides and thymidylate. Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder. Camptothecins, including, camptothecin and camptothecin derivatives are 10 available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)-1 0,11 -ethylenedioxy-20- camptothecin described below. Irinotecan HCI, (4S)-4,11-diethyl-4-hydroxy-9-[(4-piperidinopiperidino) 15 carbonyloxy]- 1 H-pyrano[3',4',6,7]indolizino[1 ,2-b]quinoline-3,14(4H,12H)-dione hydrochloride, is commercially available as the injectable solution CAMPTOSAR@. Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I - DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I 20 DNA : irintecan or SN-38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum. Topotecan HCi, (S) 1 0-[(dimethylamino)methyl}-4-ethyl-4,9-dihydroxy-1 H- pyrano[3',4',6,7]indolizino[1 ,2 b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTI N@. Topotecan is a derivative of camptothecin which binds 25 to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule. Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer. Hormones and hormonal analogues are useful compounds for treating cancers in 30 which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer. Examples of hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children ; aminoglutethimide and other aromatase inhibitors such as 35 anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical 64 WO 2010/077613 PCT/US2009/067002 carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrot acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5a-reductases such 5 as finasteride and dutasteride, useful in the treatment of prostatic carcinoma and benign prostatic hypertrophy; anti-estrogens such as tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, as well as selective estrogen receptor modulators (SERMS) such those described in U.S. Patent Nos. 5,681 ,835, 5,877,219, and 6,207,716, useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and 10 gonadotropin-releasing hormone (GnRH) and analogues thereof which stimulate the release of leutinizing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin acetate and luprolide. Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a 15 chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation. Signal tranduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domain blockers, serine/threonine kinases, phosphotidyl inositol-3 kinases, myo-inositol signaling, and Ras oncogenes. 20 Several protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth. Such protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases. Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. 25 Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e. aberrant kinase growth factor receptor activity, for example by over expression or mutation, has been shown to result in uncontrolled cell growth. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growth. 30 Consequently, inhibitors of such kinases could provide cancer treatment methods. Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, ret, vascular endothelial growth factor receptor (VEGFr), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growth factor -1 (IGFI) receptor, 35 macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor 65 WO 2010/077613 PCT/US2009/067002 (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene. Several inhibitors of growth receptors are under development and include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides. Growth factor receptors and agents that inhibit growth factor receptor 5 function are described, for instance, in Kath, John C, Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al, "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London. Tyrosine kinases, which are not growth factor receptor kinases are termed 10 nonreceptor tyrosine kinases. Non-receptor tyrosine kinases useful in the present invention, which are targets or potential targets of anti-cancer drugs, include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-Abl. Such non-receptor kinases and agents which inhibit non-receptor tyrosine kinase function are described in Sinh, S. and Corey, S. J., (1999) Journal of Hematotherapy and Stem 15 Cell Research 8 (5): 465 - 80; and Bolen, J. B., Brugge, J. S., (1997) Annual review of Immunology. 15: 371-404. SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, P13-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP. SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T. E. 20 (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32. Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, 25 iota, zeta). IkB kinase family (IKKa, IKKb), PKB family kinases, akt kinase family members, and TGF beta receptor kinases. Such Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1 101-1107; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 30 27:41-64; Philip, P.A., and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226; U.S. Patent No. 6,268,391 ; and Martinez- lacaci, L., et al, Int. J. Cancer (2000), 88(1 ), 44-52. Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of P13-kinase, ATM, DNA-PK, and Ku are also useful in the present invention. Such kinases 35 are discussed in Abraham, RT. (1996), Current Opinion in Immunology. 8 (3) 412-8; 66 WO 2010/077613 PCT/US2009/067002 Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301-3308; Jackson, S. P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al, Cancer res, (2000) 60(6), 1541-1545. Also useful in the present invention are Myo-inositol signaling inhibitors such as 5 phospholipase C blockers and Myoinositol analogues. Such signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, London. Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene. Such inhibitors include inhibitors of farnesyltransf erase, geranyl-geranyl 10 transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy. Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras , thereby acting as antiproliferation agents. Ras oncogene inhibition is discussed in Scharovsky, 0. G., Rozados, V.R., Gervasoni, S.I. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M.N. (1998), Current Opinion 15 in Lipidology. 9 (2) 99 - 102; and BioChim. Biophys. Acta, (19899) 1423(3):19-30. As mentioned above, antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors. This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases. For example Imclone C225 EGFR specific antibody 20 (see Green, M. C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat. Rev., (2000), 26(4), 269-286); Herceptin ® erbB2 antibody (see Tyrosine Kinase Signalling in Breast cancep erbB Family Receptor Tyrosine Kniases, Breast cancer Res., 2000, 2(3), 176-183); and 2CB VEGFR2 specific antibody (see Brekken, R.A. et al, Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks 25 tumor growth in mice, Cancer Res. (2000) 60, 51 17-5124). Anti-angiogenic agents including non-receptorkinase angiogenesis inhibitors may alo be useful. Anti-angiogenic agents such as those which inhibit the effects of vascular edothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM], and compounds that work by other mechanisms (for 30 example linomide, inhibitors of integrin av&3 function, endostatin and angiostatin). Agents used in immunotherapeutic regimens may also be useful in combination with the compounds of formula (1). Immunotherapy approaches, including for example ex vivo and in-vivo approaches to increase the immunogenecity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte 35 macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches 67 WO 2010/077613 PCT/US2009/067002 using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies. Agents used in proapoptotic regimens (e.g., bcl-2 antisense oligonucleotides) may 5 also be used in the combination of the present invention. Cell cycle signalling inhibitors inhibit molecules involved in the control of the cell cycle. A family of protein kinases called cyclin dependent kinases (CDKs) and their interaction with a family of proteins termed cyclins controls progression through the eukaryotic cell cycle. The coordinate activation and inactivation of different cyclin/CDK 10 complexes is necessary for normal progression through the cell cycle. Several inhibitors of cell cycle signalling are under development. For instance, examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania et al, Exp. Opin. Ther. Patents (2000) 10(2):215-230. For the treatment or prophylaxis of pulmonary disorders, anticholinergics of 15 potential use in treating asthma, COPD, bronchitis, and the like, and therefore useful as an additional therapeutic agent include antagonists of the muscarinic receptor (particularly of the M3 subtype) which have shown therapeutic efficacy in man for the control of cholinergic tone in COPD (Witek, 1999); 1-{4-Hydroxy-1-[3,3,3-tris-(4-fluoro phenyl)-propionyl)-pyrrolidine-2-carbonyl}-pyrrolidine-2-carboxylic acid (1-methyl 20 piperidin-4-ylmethyl)-amide; 3-[3-(2-Diethylamino-acetoxy)-2-pheny-propionyloxy]-8 isopropyl-8-methyl-8-azonia-bicyclo[3.2. Octane (Ipratropium-N,N-diethylglycinate); 1 Cyclohexyl-3,4-dihydro-1H-isoquinoline-2-carboxylic acid 1 -aza-bicyclo[2.2.2]oct-3-yl ester (Solifenacin); 2-Hydroxymethyl-4-methanesulfiny-2-phenyl-butyric acid 1-aza bicyclo[2.2.2]oct-3-yl ester (Revatropate); 2-{1-[2-(2,3-Dihydro-benzofuran-5-yl)-ethyl] 25 pyrrolidin-3-yl)-2,2-diphenyl-acetamide (Darifenacin); 4-Azepan-1-yl-2,2-diphenyl butyramide (Buzepide); 7-[3-(2-Diethylarmino-acetoxy)-2-pheny-propionyloxy]-9-ethyl-9-methyl-3-oxa-9-azonia tricyclo[3.3.1.02,4]nonane (Oxitropium-N,N-diethylglycinate); 7-[2-(2-Diethylamino acetoxy)-2,2-di-thiophen-2-yl-acetoxy]-9,9-dimethyl-3-oxa-9-azonia 30 tricyclo[3.3.1.02,4]nonane (Tiotropium-N,N-diethylglycinate); Dimethylamino-acetic acid 2-(3-diisopropylamino-1 -phenyl-propyl)-4-methyl-phenyl ester (Tolterodine-N,N dimethylglycinate); 3-[4,4-Bis-(4-fluoro-phenyl)-2-oxo-imidazolidin-1-yl]-1-methyl-i-(2-oxo 2-pyridin-2-yl-ethyl)-pyrrolidinium; 1-[1-(3-Fluoro-benzyl)-piperidin-4-yl]-4,4-bis-(4-fluoro-phenyl)-imidazolidin-2-one; 35 1-Cyclooctyl-3-(3-methoxy-1-aza-bicyclo[2.2.2]oct-3-yl)-1-phenyl-prop-2-yn-1-ol; 68 WO 2010/077613 PCT/US2009/067002 3-{2-(2-Diethylamino-acetoxy)-2,2-di-thiophen-2-y-acetoxy]-1 -(3-phenoxy-propyl)-1 azonia-bicyclo2.2.2]octane (Aclidinium-N,N-diethylglycinate); or (2-Diethylamino acetoxy)-di-thiophen-2-yI-acetic acid 1-methy-1 -(2-phenoxy-ethyl)-piperidin-4-yl ester; beta-2 agonist used to treat broncho-constriction in asthma, COPD and bronchitis include 5 salmeterol and albuterol; anti-inflammatory signal transduction modulators for asthma. With regard to the pulmonary condition of asthma, those skilled in the art appreciate that asthma is a chronic inflammatory disease of the airways resulting from the infiltration of pro-inflammatory cells, mostly eosinophils and activated T-lymphocytes into the bronchial mucosa and submucosa. The secretion of potent chemical mediators, 10 including cytokines, by these proinflammatory cells alters mucosal permeability, mucus production, and causes smooth muscle contraction. All of these factors lead to an increased reactivity of the airways to a wide variety of irritant stimuli (Kaliner, 1988). Targeting signal transduction pathways is an attractive approach to treating inflammatory diseases, as the same pathways are usually involved in several cell types and regulate 15 several coordinated inflammatory processes, hence modulators have the prospect of a wide spectrum of beneficial effects. Multiple inflammatory signals activate a variety of cell surface receptors that activate a limited number of signal transduction pathways, most of which involve cascades of kinases. These kinases in turn may activate transcription factors that regulate multiple inflammatory genes. Applying "anti-inflammatory signal 20 transduction modulators" (referred to in this text as AISTM), like phosphodiesterase inhibitors (e.g. PDE-4, PDE-5, or PDE-7 specific), transcription factor inhibitors (e.g. blocking NFKB through IKK inhibition), or kinase inhibitors (e.g. blocking P38 MAP, JNK, P13K, EGFR or Syk) is a logical approach to switching off inflammation as these small molecules target a limited number of common intracellular pathways - those signal 25 transduction pathways that are critical points for the anti-inflammatory therapeutic intervention (see review by P.J. Barnes, 2006). Additional therapeutic agents include: 5-(2,4-Difluoro-phenoxy)-1-isobutyl-1H indazole-6-carboxylic acid (2-dimethylamino-ethyl)-amide (P38 Map kinase inhibitor ARRY-797); 3-Cyclopropylmethoxy-N-(3,5-dichloro-pyridin-4-y)-4-difluorormethoxy 30 benzamide (PDE-4 inhibitor Roflumilast); 4-[2-(3-cyclopentyloxy-4-methoxyphenyl)-2 phenyl-ethyl]-pyridine (PDE-4 inhibitor CDP-840); N-(3,5-dichloro-4-pyridinyl)-4 (difluoromethoxy)-8-(methylsulfonyl)amino]-1-dibenzofurancarboxamide (PDE-4 inhibitor Oglemilast); N-(3,5-Dichloro-pyridin-4-y)-2-[1-(4-fluorobenzyl)-5-hydroxy-1H-indol-3-yl]-2 oxo-acetamide (PDE-4 inhibitor AWD 12-281); 8-Methoxy-2-trifluoromethyl-quinoline-5 35 carboxylic acid (3,5-dichloro-1-oxy-pyridin-4-yl)-amide (PDE-4 inhibitor Sch 351591); 4-[5 69 WO 2010/077613 PCT/US2009/067002 (4-Fluorophenyl)-2-(4-methanesulfinyl-phenyl)-1H-imidazol-4-yi]-pyridine (P38 inhibitor SB-203850); 4-[4-(4-Fluoro-phenyl)-1 -(3-phenyl-propyl)-5-pyridin-4-yI-1 H-imidazol-2-yl] but-3-yn-1-ol (P38 inhibitor RWJ-67657); 4-Cyano-4-(3-cyclopentyloxy-4-methoxy phenyf)-cyclohexanecarboxylic acid 2-diethylamino-ethyl ester (2-diethyl-ethyl ester 5 prodrug of Cilomilast, PDE-4 inhibitor); (3-Chloro-4-fluorophenyl)47-methoxy-6-(3 morpholin-4-yl-propoxy)-quinazolin-4-y]-amine (Gefitinib, EGFR inhibitor); and 4-(4 Methyl-piperazin-1-ylmethyl)-N-[4-methyl-3-(4-pyridin-3-y-pyrimidin-2-ylamino)-phenyl] benzamide (Imatinib, EGFR inhibitor). Moreover, asthma is a chronic inflammatory disease of the airways produced by 10 the infiltration of pro-inflammatory cells, mostly eosinophils and activated T-lymphocytes (Poston, Am. Rev. Respir. Dis., 145 (4 Pt 1), 918-921, 1992; Walker, J. Allergy Clin. Immunot, 88 (6), 935-42, 1991) into the bronchial mucosa and submucosa. The secretion of potent chemical mediators, including cytokines, by these proinflammatory cells alters mucosal permeability, mucus production, and causes smooth muscle 15 contraction. All of these factors lead to an increased reactivity of the airways to a wide variety of irritant stimuli (Kaliner, "Bronchial asthma, Immunologic diseases" E. M. Samter, Boston, Little, Brown and Company: 117-118. 1988). Glucocorticoids, which were first introduced as an asthma therapy in 1950 (Carryer, Journal of Allergy, 21, 282-287, 1950), remain the most potent and consistently 20 effective therapy for this disease, although their mechanism of action is not yet fully understood (Morris, J. Allergy Clin. ImmunoL, 75 (1 Pt) 1-13, 1985). Unfortunately, oral glucocorticoid therapies are associated with profound undesirable side effects such as truncal obesity, hypertension, glaucoma, glucose intolerance, acceleration of cataract formation, bone mineral loss, and psychological effects, all of which limit their use as 25 long-term therapeutic agents (Goodman and Gilman, 10th edition, 2001). A solution to systemic side effects is to deliver steroid drugs directly to the site of inflammation. Inhaled corticosteroids (ICS) have been developed to mitigate the severe adverse effects of oral steroids. While ICS are very effective in controlling inflammation in asthma, they too are not precisely delivered to the optimal site of action in the lungs and produce 30 unwanted side effects in the mouth and pharynx (candidiasis, sore throat, dysphonia). Combinations of inhaled f2-adrenoreceptor agonist bronchodilators such as formoterol or salmeterol with ICS's are also used to treat both the bronchoconstriction and the inflammation associated with asthma and COPD (Symbicort@ and Advair@, respectively). However, these combinations have the side effects of both the iCS's and the p2 35 adrenoreceptor agonist because of systemic absorption (tachycardia, ventricular 70 WO 2010/077613 PCT/US2009/067002 dysrhythmias, hypokalemia) primarily because neither agent is delivered to the optimal sites of actions in the lungs. In consideration of all problems and disadvantages connected with the adverse side effect profile of ICS and of R2-agonists it would be highly advantageous to provide mutual steroid-p2-agonist prodrug to mask the pharmacological 5 properties of both steroids and f2-agonists until such a prodrug reaches the lungs, thereby mitigating the oropharyngeal side effects of ICS and cardiovascular side-effects of p2-agonists. In one aspect, such a mutual steroid-p2-agonist prodrug would be effectively delivered to the endobronchial space and converted to active drugs by the action of lung enzymes, thereby delivering to the site of inflammation and 10 bronchoconstriction a therapeutic amount of both drugs. An anti-inflammatory agent for combination therapy includes dexamethasone, dexamethasone sodium phosphate, fluorometholone, fluorometholone acetate, loteprednol, loteprednol etabonate, hydrocortisone, prednisolone, fludrocortisones, triamcinolone, triamcinolone acetonide, betamethasone, beclomethasone diproprionate, methylprednisolone, fluocinolone, 15 fluocinolone acetonide, flunisolide, fluocortin-21-butylate, flumethasone, flumetasone pivalate, budesonide, halobetasol propionate, mometasone furoate, fluticasone propionate, ciclesonide; or a pharmaceutically acceptable salt thereof. The immune response to certain antigens can be enhanced through the use of immune potentiators, known as vaccine adjuvants. A discussion of immunological 20 adjuvants can be found in "Current Status of Immunological Adjuvants", Ann. Rev. Immunol, 1986, 4, pp. 369-388 and "Recent Advances in Vaccine Adjuvants and Delivery Systems" by D. T. O'Hagan and N. M. Valiante. The disclosures of U. S. 4,806,352; 5,026,543; and 5,026,546 describe various vaccine adjuvants appearing in the patent literature. Each of these references is hereby incorporated by reference in their 25 entireties. In one embodiment of the instant invention, provided are methods of administering a vaccine by administering a compound of Formula 11 alone or in combination with antigens and/or other agents. In another embodiment, immune responses to vaccines using antigenic epitopes from sources such as synthetic peptides, bacterial, or viral 30 antigens are enhanced by co-administration of the compounds of Formula 11. In other embodiments, the instant invention provides immunogenic compositions comprising one or more antigens and a compound of Formula I effective to stimulate a cell mediated response to said one or more antigens. In another embodiment, compounds of Formula Il can be used in the manufacture 35 of a medicament for enhancing the immune response to an antigen. Other embodiments 71 WO 2010/077613 PCT/US2009/067002 provide the use of the compound of Formula 11 in the manufacture of a medicament for immune stimulation, and another agent, such as an antigen, for simultaneous, separate or sequential administration. In another embodiment, provided is a pharmaceutical preparation comprising (a) a 5 compound of Formula 11 and (b) an antigen, wherein (a) and (b) are either in admixture or are separate compositions. These embodiments are for simultaneous, separate or sequential administration. When in separate compositions, the compound of Formula Il may be administered may be administered enterally, orally, parenterally, sublingually, intradermally, by inhalation spray, rectally, or topically in dosage unit formulations that 10 include conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. For example, suitable modes of administration include oral, subcutaneous, transdernal, transmucosal, iontophoretic, intravenous, intramuscular, intraperitoneal, intranasal, subdermal, rectal, and the like. Topical administration may also include the use of transdermal administration such as transdermal patches or 15 ionophoresis devices. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques. orally, topically, nasally, rectally, by inhalation or by injection. In another embodiment, compounds of Formula 11 are used as polyclonal activators for the production of antigens. More particularly the invention relates to a 20 method of preparing monoclonal antibodies with a desired antigen specificity comprising contacting a compound of Formula with immortalized memory B cells. The monoclonal antibodies produced therefrom, or fragments thereof, may be used for the treatment of disease, for the prevention of disease or for the diagnosis of disease. Vaccines or immunogenic compositions of the instant invention comprising a 25 compound of Formula Il may be administered in conjunction with one or more immunoregulatory agents. In particular, compositons can include another adjuvant. Adjuvants for use with the invention include, but are not limited to, mineral containing compositions such as calcium or aluminium salts, for example AIK(SO4) 2 , AI(OH) 3 , AIPO 4 , or combinations thereof. Other adjuvants include oil-emulsions, particularly submicron 30 oil-in-water emulsions such as those described in W090/1 4837, US 6,299,884 and US 6,452,325. Other adjuvants include saponin formulations such as QS7, QS17, QS18, QS21, QH-A, QH-B and QH-C, see US 5,057,540 and Barr, et al. Advanced Drug Delivery Reviews (1998), 32:247-271. Other adjuvants include virosomes and virus like particles (VLPs) (Gluck, et al., Vaccine (2002) 20:B10-B16, US 20090263470); bacterial 35 or microbial derivatives, Lipid A derivatives, immunostimulartory oligonucleotides, ADP 72 WO 2010/077613 PCT/US2009/067002 ribosylating toxins and detoxified derivaties thereof, bioadhesives and mucoadhesives, microparticles, liposomes, polyphasphazene (PCPP), and other small molecule immunopotentiators. One or more of the above named adjuvants may be used in a vaccine combination with a compound of Formula 11. 5 The invention is also directed to methods of administering the immunogenic compositions of the invention, wherein the immunogenic composition includes in one embodiment one or more adjuvants and antigens as described herein in combination with a compound of Formula 11. In some embodiments, the immunogenic composition is administered to the subject in an amount effective to stimulate an immune response. The 10 amount that constitutes an effective amount depends, inter alia, on the particular immunogenic composition used, the particular adjuvant compound being administered and the amount thereof, the immune response that is to be enhanced (humoral or cell mediated), the state of the immune system (e.g., suppressed, compromised, stimulated), and the desired therapeutic result. Accordingly it is not practical to set forth generally the 15 amount that constitutes an effective amount of the immunogenic composition. Those of ordinary skill in the art, however, can readily determine the appropriate amount with due consideration of such factors. The immunogenic compositions of the present invention can be used in the manufacture of a vaccine. Suitable vaccines include, but are not limited to, any material 20 that raises either or both humoral or cell mediated immune response. Suitable vaccines can include live viral and bacterial antigens and inactivated viral, tumor-derived, protozoal, organism-derived, fungal, and bacterial antigens, toxoids, toxins, polysaccharides, proteins, glycoproteins, peptides, and the like. Compositions of a compound of Formula i may be administered in conjunction 25 with one or more antigens for use in therapeutic, prophylactic, or diagnostic methods of the instant invention. In another aspect of this embodiment, these compositions may be used to treat or prevent infections caused by pathogens. In another aspect of this embodiment, these compostions may also be combined with an adjuvant as described supra. 30 Antigens for use with the invention include, but are not limited to, one or more of the antigens comprising bacterial antigens, viral antigens, fungal antigens, antigens from sexually transmitted diseases (STD), respiratory antigens, pediatric vaccine antigens, antigens suitable for use in elderly or immunocompromised individuals, antigens suitable for use in adolescent vaccines, and tumor antigens. 73 WO 2010/077613 PCT/US2009/067002 In yet another embodiment, the present application discloses pharmaceutical compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, in combination with at least one additional active agent, and a pharmaceutically acceptable carrier or excipient. In yet another embodiment, the present 5 application provides a combination pharmaceutical agent with two or more therapeutic agents in a unitary dosage form. Thus, it is also possible to combine any compound of the invention with one or more other active agents in a unitary dosage form. The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two 10 or more administrations. Co-administration of a compound of the invention with one or more other active agents generally refers to simultaneous or sequential administration of a compound of the invention and one or more other active agents, such that therapeutically effective amounts of the compound of the invention and one or more other active agents are both 15 present in the body of the patient. Co-administration includes administration of unit dosages of the compounds of the invention before or after administration of unit dosages of one or more other active agents, for example, administration of the compounds of the invention within seconds, minutes, or hours of the administration of one or more other active agents. For example, 20 a unit dose of a compound of the invention can be administered first, followed within seconds or minutes by administration of a unit dose of one or more other active agents. Alternatively, a unit dose of one or more other active agents can be administered first, followed by administration of a unit dose of a compound of the invention within seconds or minutes. In some cases, it may be desirable to administer a unit dose of a compound 25 of the invention first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more other active agents. In other cases, it may be desirable to administer a unit dose of one or more other active agents first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound of the invention. The combination therapy may provide "synergy" and "synergistic effect", i.e, the 30 effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation 35 therapy, a synergistic effect may be attained when the compounds are administered or 74 WO 2010/077613 PCT/US2009/067002 delivered sequentially, e.g., in separate tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together. 5 Methods of Treatment As used herein, an "agonist" is a substance that stimulates its binding partner, typically a receptor. Stimulation is defined in the context of the particular assay, or may be apparent in the literature from a discussion herein that makes a comparison to a factor 10 or substance that is accepted as an "agonist" or an "antagonist" of the particular binding partner under substantially similar circumstances as appreciated by those of skill in the art. Stimulation may be defined with respect to an increase in a particular effect or function that is induced by interaction of the agonist or partial agonist with a binding partner and can include allosteric effects. 15 As used herein, an "antagonist" is a substance that inhibits its binding partner, typically a receptor. Inhibition is defined in the context of the particular assay, or may be apparent in the literature from a discussion herein that makes a comparison to a factor or substance that is accepted as an "agonist" or an "antagonist" of the particular binding partner under substantially similar circumstances as appreciated by those of skill in the 20 art. Inhibition may be defined with respect to a decrease in a particular effect or function that is induced by interaction of the antagonist with a binding partner, and can include allosteric effects. As used herein, a "partial agonist" or a "partial antagonist" is a substance that provides a level of stimulation or inhibition, respectively, to its binding partner that is not 25 fully or completely agonistic or antagonistic, respectively. It will be recognized that stimulation, and hence, inhibition is defined intrinsically for any substance or category of substances to be defined as agonists, antagonists, or partial agonists. As used herein, "intrinsic activity" or "efficacy" relates to some measure of biological effectiveness of the binding partner complex. With regard to receptor 30 pharmacology, the context in which intrinsic activity or efficacy should be defined will depend on the context of the binding partner (e.g., receptor/ligand) complex and the consideration of an activity relevant to a particular biological outcome. For example, in some circumstances, intrinsic activity may vary depending on the particular second messenger system involved. Where such contextually specific evaluations are relevant, 35 and how they might be relevant in the context of the present invention, will be apparent to 75 WO 2010/077613 PCT/US2009/067002 one of ordinary skill in the art. As used herein, modulation of a receptor includes agonism, partial agonism, antagonism, partial antagonism, or inverse agonism of a receptor. As will be appreciated by those skilled in the art, when treating a viral infection 5 such as HCV, HBV, or HIV, such treatment may be characterized in a variety of ways and measured by a variety of endpoints. The scope of the present invention is intended to encompass all such characterizations. In one embodiment, the method can be used to induce an immune response against multiple epitopes of a viral infection in a human. Induction of an immune 10 response against viral infection can be assessed using any technique that is known by those of skill in the art for determining whether an immune response has occurred. Suitable methods of detecting an immune response for the present invention include, among others, detecting a decrease in viral load or antigen in a subject's serum, detection of IFN-gamma-secreting peptide specific T cells, and detection of elevated levels of one 15 or more liver enzymes, such as alanine transferase (ALT) and aspartate transferase (AST). In one embodiment, the detection of IFN-gamma-secreting peptide specific T cells is accomplished using an ELISPOT assay. Another embodiment includes reducing the viral load associated with HBV infection, including a reduction as measured by PCR testing. 20 in another aspect, the present invention provides methods for treating a hepatitis B viral infection or a hepatitis C viral infection, wherein each of the methods includes the step of administering to a human subject infected with hepatitis B virus or hepatitis C virus a therapeutically effective amount a compound of Formula la, ll, or Ila or a pharmaceutically acceptable salt thereof. Typically, the human subject is suffering from 25 a chronic hepatitis B infection or a chronic hepatitis C infection, although it is within the scope of the present invention to treat people who are acutely infected with HBV or HCV. Treatment in accordance with the present invention typically results in the stimulation of an immune response against HBV or HCV in a human being infected with HBV or HCV, respectively, and a consequent reduction in the viral load of HBV or HCV in 30 the infected person. Examples of immune responses include production of antibodies (e.g., IgG antibodies) and/or production of cytokines, such as interferons, that modulate the activity of the immune system. The immune system response can be a newly induced response, or can be boosting of an existing immune response. In particular, the immune system response can be seroconversion against one or more HBV or HCV 35 antigens. 76 WO 2010/077613 PCT/US2009/067002 The viral load can be determined by measuring the amount of HBV DNA or HCV DNA present in the blood. For example, blood serum HBV DNA can be quantified using the Roche COBAS Amplicor Monitor PCR assay (version 2.0; lower limit of quantification, 300 copies/mL [57 IU/mL]) and the Quantiplex bDNA assay (lower limit of quantification, 5 0.7 MEq/mL; Bayer Diagnostics, formerly Chiron Diagnostics, Emeryville, CA). The amount of antibodies against specific HBV or HCV antigens (e.g., hepatitis B surface antigen (HBsAG)) can be measured using such art-recognized techniques as enzyme linked immunoassays and enzyme-linked immunoabsorbent assays. For example, the amount of antibodies against specific HBV or HCV antigens can be measured using the 10 Abbott AxSYM microparticle enzyme immunoassay system (Abbott Laboratories, North Chicago, IL). A compound of Formula 11 can be administered by any useful route and means, such as by oral or parenteral (e.g., intravenous) administration. Therapeutically effective amounts of Formula 11 are from about 0.00001 mg/kg body weight per day to about 10 15 mg/kg body weight per day, such as from about 0.0001 mg/kg body weight per day to about 10 mg/kg body weight per day, or such as from about 0.001 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.01 mg/kg body weight per day to about 1 mg/kg body weight per day, or such as from about 0.05 mg/kg body weight per day to about 0.5 mg/kg body weight per day, or such as from about 0.3 pg to 20 about 30 mg per day, or such as from about 30 pg to about 300 pg per day. The frequency of dosage of Formula 11 will be determined by the needs of the individual patient and can be, for example, once per day or twice, or more times, per day. Administration of Formula 1l continues for as long as necessary to treat the HBV or HCV infection. For example, Formula 11 can be administered to a human being infected with 25 HBV or HCV for a period of from 20 days to 180 days or, for example, for a period of from 20 days to 90 days or, for example, for a period of from 30 days to 60 days. Administration can be intermittent, with a period of several or more days during which a patient receives a daily dose of Formula 11, followed by a period of several or more days during which a patient does not receive a daily dose of Formula 11. For 30 example, a patient can receive a dose of Formula II every other day, or three times per week. Again by way of example, a patient can receive a dose of Formula I each day for a period of from 1 to 14 days, followed by a period of 7 to 21 days during which the patient does not receive a dose of Formula 11, followed by a subsequent period (e.g., from 1 to 14 days) during which the patient again receives a daily dose of Formula lH. 77 WO 2010/077613 PCT/US2009/067002 Alternating periods of administration of Formula 11, followed by non-administration of Formula 1l, can be repeated as clinically required to treat the patient. As described more fully herein, Formula I can be administered with one or more additional therapeutic agent(s) to a human being infected with HBV or HCV. The 5 additional therapeutic agent(s) can be administered to the infected human being at the same time as Formula 11, or before or after administration of Formula 1l. In another aspect, the present invention provides a method for ameliorating a symptom associated with an HBV infection or HCV infection, wherein the method comprises administering to a human subject infected with hepatitis B virus or hepatitis C 10 virus a therapeutically effective amount of Formula 11, or a pharmaceutically acceptable salt thereof, wherein the therapeutically effective amount is sufficient to ameliorate a symptom associated with the HBV infection or HCV infection. Such symptoms include the presence of HBV virus particles (or HCV virus particles) in the blood, liver inflammation, jaundice, muscle aches, weakness and tiredness. 15 In a further aspect, the present invention provides a method for reducing the rate of progression of a hepatitis B viral infection, or a hepatitis C virus infection, in a human being, wherein the method comprises administering to a human subject infected with hepatitis B virus or hepatitis C virus a therapeutically effective amount of Formula 11, or a pharmaceutically acceptable salt thereof, wherein the therapeutically effective amount is 20 sufficient to reduce the rate of progression of the hepatitis B viral infection or hepatitis C viral infection. The rate of progression of the infection can be followed by measuring the amount of HBV virus particles or HCV virus particles in the blood. In another aspect, the present invention provides a method for reducing the viral load associated with HBV infection or HCV infection, wherein the method comprises 25 administering to a human being infected with HBV or HCV a therapeutically effective amount of Formula 11, or a pharmaceutically acceptable salt thereof, wherein the therapeutically effective amount is sufficient to reduce the HBV viral load or the HCV viral load in the human being. In a further aspect, the present invention provides a method of inducing or 30 boosting an immune response against Hepatitis B virus or Hepatitis C virus in a human being, wherein the method comprises administering a therapeutically effective amount of Formula 11, or a pharmaceutically acceptable salt thereof, to the human being, wherein a new immune response against Hepatitis B virus or Hepatitis C virus is induced in the human being, or a preexisting immune response against Hepatitis B virus or Hepatitis C 35 virus is boosted in the human being. Seroconversion with respect to HBV or HCV can be 78 WO 2010/077613 PCT/US2009/067002 induced in the human being. Examples of immune responses include production of antibodies, such as IgG antibody molecules, and/or production of cytokine molecules that modulate the activity of one or more components of the human immune system. Induction of seroconversion against HCV or HBV in patients chronically infected 5 with either of these viruses is an unexpected property of Formula I. In clinical practice, an HBV patient, or HCV patient, is treated with Formula I, alone or in combination with one or more other therapeutic agents, until an immune response against HBV or HCV is induced or enhanced and the viral load of HBV or HCV is reduced. Thereafter, although the HBV or HCV virus may persist in a latent form in the patient's body, treatment with 10 Formula 11 can be stopped, and the patient's own immune system is capable of suppressing further viral replication. In patients treated in accordance with the present invention and who are already receiving treatment with an antiviral agent that suppresses replication of the HBV virus or HCV virus, there may be little or no detectable viral particles in the body of the patient during treatment with the antiviral agent(s). In these 15 patients, seroconversion will be evident when the antiviral agent(s) is no longer administered to the patient and there is no increase in the viral load of HBV or HCV. In the practice of the present invention, an immune response is induced against one or more antigens of HBV or HCV. For example, an immune response can be induced against the HBV surface antigen (HBsAg), or against the small form of the HBV 20 surface antigen (small S antigen), or against the medium form of the HBV surface antigen (medium S antigen), or against a combination thereof. Again by way of example, an immune response can be induced against the HBV surface antigen (HBsAg) and also against other HBV-derived antigens, such as the core polymerase or x-protein. Induction of an immune response against HCV or HBV can be assessed using 25 any technique that is known by those of skill in the art for determining whether an immune response has occurred. Suitable methods of detecting an immune response for the present invention include, among others, detecting a decrease in viral load in a subject's serum, such as by measuring the amount of HBV DNA or HCV DNA in a subject's blood using a PCR assay, and/or by measuring the amount of anti-HBV antibodies, or anti-HCV 30 antibodies, in the subject's blood using a method such as an ELISA. Additionally, the compounds of this invention are useful in the treatment of cancer or tumors (including dysplasias, such as uterine dysplasia). These includes hernatological malignancies, oral carcinomas (for example of the lip, tongue or pharynx), digestive organs (for example esophagus, stomach, small intestine, colon, large intestine, 35 or rectum), liver and biliary passages, pancreas, respiratory system such as larynx or 79 WO 2010/077613 PCT/US2009/067002 lung (small cell and non-small cell), bone, connective tissue, skin (e.g., melanoma), breast, reproductive organs (uterus, cervix, testicles, ovary, or prostate), urinary tract (e.g., bladder or kidney), brain and endocrine glands such as the thyroid. In summary, the compounds of this invention are employed to treat any neoplasm, including not only 5 hematologic malignancies but also solid tumors of all kinds. Hematological malignancies are broadly defined as proliferative disorders of blood cells and/or their progenitors, in which these cells proliferate in an uncontrolled manner. Anatomically, the hematologic malignancies are divided into two primary groups: lymphomas - malignant masses of lymphoid cells, primarily but not exclusively in lymph 10 nodes, and leukemias - neoplasm derived typically from lymphoid or myeloid cells and primarily affecting the bone marrow and peripheral blood. The lymphomas can be sub divided into Hodgkin's Disease and Non-Hodgkin's lymphoma (NHL). The later group comprises several distinct entities, which can be distinguished clinically (e.g. aggressive lymphoma, indolent lymphoma), histologically (e.g. follicular lymphoma, mantle cell 15 lymphoma) or based on the origin of the malignant cell (e.g. B lymphocyte, T lymphocyte). Leukemias and related malignancies include acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL). Other hematological malignancies include the plasma cell dyscrasias including multiple myeloma, and the myelodysplastic syndromes. 20 Synthetic Examples Certain abbreviations and acronyms are used in describing the experimental details. Although most of these would be understood by one skilled in the art, Table 1 contains a list of many of these abbreviations and acronyms. 25 Table 1. List of abbreviations and acronyms. Abbreviation Meaning Ac 2 O acetic anhydride AIBN 2,2'-azobis(2-methylpropionitrile) Bn benzyl BnBr benzytbromide BSA bis(trimethylsilyl)acetamide BzCI benzoyl chloride CDI carbonyl diimidazole 80 WO 2010/077613 PCT/US2009/067002 DABCO 1,4-diazabicyclo[2.22octane DBN 1,5-diazabicyclo[4.3.0)non-5-ene DDQ 2,3-dichloro-5,6-dicyano-1,4-benzoquinone DBU 1,5-diazabicyclo[5.4.0]undec-5-ene DCA dichloroacetamide DCC dicyclohexylcarbodiimide DCM dichioromethane DMAP 4-dimethylaminopyridine OME 1,2-dimethoxyethane DMTCl dimethoxytrityl chloride DMSO dimethylsulfoxide DMTr 4, 4'-dimethoxytrityl DMF dimethylformamide EtOAc ethyl acetate ESI -electrospray ionization HMDS hexamethyldisilazane HPLC High pressure liquid chromatography LDA lithium diisopropylamide LRMS low resolution mass spectrum MCPBA meta-chloroperbenzoic acid MeCN acetonitrile MeOH methanol MMTC mono methoxytrityl chloride m/z or m/e mass to charge ratio MH' mass plus 1 MH' mass minus 1 MsOH methanesulfonic acid MS or ms mass spectrum NBS N-bromosuccinimide Ph phenyl rt or r.t. room temperature TBAF tetra butylammonium fluoride TMSCI chlorotrimethylsilane TMSBr bromotrimethylsilane 81 WO 2010/077613 PCT/US2009/067002 TMS1 iodotrimethylsilane I5SOTf (trimethylsilyl)trifluoromethylsulfonate TEA triethylamine TBA tributylamine TBAP tributylammonium pyrophosphate TBSCI t-butyldimethylsilyl chloride TEAB triethylammonium bicarbonate TFA trifluoroacetic acid TLC or tic thin layer chromatography Tr triphenylmethyl Tol 4-methylbenzoylJ Turbo Grignard 1:1 mixture of isopropylmagnesium chloride and lithium chloride parts per million down field from tetramethylsilane General Scheme Pteridinone Derivatives 82 WO 20101077613 PCT/US20091067002 C0 2 CH
NH
2 R4
NO
2 HN R 5 Step 1 N CN D
(L
2
-R
2 )m
NH
2 NH N N 2
CO
2 CH NO 0 2 0H 3 S Step 2 N
NO
2 S N N R S N N- R5 X1 I X (RS), X1()
(L
2
-R
2 )m
(L
2
-R
2 )m
NH
2 NH
N
2
CO
2
GH
3 NH 2 H~ Step3 R1 L Nz C~R Step 4 R 1 N A
L
1 N N- R5 Li) N N R 5 (R3), (R-)I D D
(L
2
-R
2 )m (L 2
-R
2 )m 83 WO 2010/077613 PCT/US2009/067002 Scheme 1
CO
2 Et CI NH 2 NHN0N NO2 NO 2
H
3 CS N CI H 3 CS N C1 C A B
NH
2 NO 02Et
NH
2 N
H
3 CS N N Fe T
H
3 CS N N> HOAc D Example 1
CO
2 Et
NH
2 N HN N BrCH2CO 2 Et E C Compound B 5 To a solution of compound A (2.46 g, 10.2 mmol) in THF (34 mL) at - 20 OC was added Et 3 N (3.14 mL, 22.5 mmol) followed by a solution of NH 3 (2.0 M in MeOH, 5.4 mL, 11 mmol). The mixture was stirred while warming to 0 IC for 1.5 h (LCIMS indicated consumption of starting materials). The reaction mixture was taken forward without work up. 10 Compound C 84 WO 2010/077613 PCT/US2009/067002 To a solution of 3-((1-pyrrolidinylmethyl)phenyl)methanamine E (1.95 9, 10.2 mmol) in THF (34 mL) at 0 *C was added Et 3 N (3.14 mmol, 22.5 mmol) followed by methyl bromoacetate (1.04 mL, 22.3 mmol) dropwise. The reaction mixture was stirred until LC/MS indicated consumption of starting materials, approximately 2 h. The mixture was 5 taken forward to the synthesis of compound D without work up. Compound D The above reaction mixture containing compound C was added to the reaction mixture containing compound B at 0 0 C. The reaction mixture was stirred until LC/MS indicated 10 the consumption of compound B, approximately 45 min. A saturated solution of NH 4 CI (50 mL) was added. The layers were separated, and the aqueous layer was extracted with EtOAc (2 x 30 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum. Purification by silica gel chromatography provided 2.11 g (46% from A) of compound D. 'H NMR (CD 3 0D, 300 MHz): d (ppm) 7.32-7.16 (m, 4H), 15 4.69 (s, 2H), 4.19 (q, J = 7 Hz, 2H), 4.07 (s, 2H), 3.60 (s, 2H), 2.49 (m, 4H), 2.40 (s, 3H), 1.78 (m, 4H), 1.23 (t, 3 H, J = 7 Hz). LCMS-ESI*: calc'd for C 21
H
29
N
6 0 4 S: 461.2 (M+H*); Found: 461.0 (M+H*). Example 1 20 A solution of compound 4 (50 mg) and Fe dust (117 mg) in AcOH (2 mL) was stirred at rt for 13 h. The reaction was filtered through Celite and purified by HPLC on a C18 column, eluting with a gradient of 2-98% acetonitrile in H 2 0 to provide Example I in 13% yield. 1 H NMR (CD30D): 6 7.40-7.22 (m, 4H), 4.82 (s, 2H), 3.93 (s, 2H), 3.73 (s, 2H), 2.70-2.60 (m, 4H), 2.41 (s, 3H), 1.90-1.78 (m, 4 H); MS: 385.2 (M + H*). 25 Scheme 2 85 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2 NO2 :1NO 2 1 OxoneN NO N N0 2 O N N CO2Et
CH
3 S N N/CO2Et 2. 2-methoxyethano0 SN2CO3 F N
NH
2 H Fe, AcOH N XONA OAN' N N N Example 2 Compound F Compound D was dissolved in methanol (2 mL), and to this was added a solution of 5 Oxone (1.08 g) in H 2 0 (3 mL). The mixture was stirred for 30 min, after which the oxidation was almost complete. The mixture was added to water and extracted with
CH
2
CI
2 . The organic phase was dried over Na 2
SO
4 , filtered, and concentrated under vacuum to give the desired sulfone intermediate, which was carried on to the next step. The sulfone and Cs 2 C0 3 (384 mg) were taken up in CH 2
CI
2 (4 mL) and to this was added 10 2-methoxyethanol (880 LL) dropwise. After stirring for one hour, some sulfone starting material remained as indicated by LC/MS, and another 200 piL of 2-methoxyethanol was added and the reaction was stirred for an additional 30 min. The reaction mixture was diluted with CH 2
CI
2 and washed with water. The organic layer was dried over Na 2
SO
4 , filtered, and concentrated under vacuum. The product was purified by flash 15 chromatography on silica gel, eluting with 20% MeOH in CH 2
CI
2 , to give compound F in 40% yield. 1 H NMR (CD 3 OD): 5 7.40-7.15 (m, 4H), 4.69 (br s, 2H), 4.33 (t, J = 4.8 Hz, 2H), 4.17 (q, J = 6.9 Hz, 2H), 4.04 (s, 2H), 3.68 (s, 2H), 3.03 (t, J = 4.2 Hz, 2H), 3.68 (s, 3H), 2.60 (s, 4H), 1.81 (s, 4H), 1.24 (t, J = 7.2 Hz, 3H); MS: 489.2 (M + H*). 20 Example 2 A mixture of compound F (33 mg), iron dust (56 mg), and acetic acid (1 mL) was stirred at rt for 4 h. After this time conversion was incomplete, so another portion of iron dust (20 mg) was added and the reaction was stirred for another 6 h. A third portion of iron dust (30 mg) was added and the mixture was stirred another 12 h. The mixture was filtered 25 through silica gel, and the solvent was removed under vacuum. The product was purified 86 WO 2010/077613 PCT/US2009/067002 from the remaining material by preparative HPLC on a C18 column, eluting with a gradient of 2-98% acetonitrile in H 2 0, providing Example 2. 1 H NMR (CD30D): 5 7.62 (s, 1H), 7.50 (s, 3H), 4.95 (s, 2H), 4.60-4.53 (m, 2H), 4.39 (s, 2H), 4.15 (s, 2H), 3.95-3.67 (m, 2H), 3.60-3.42 (m, 2H), 3.68 (s, 3H), 3.25-3.12 (m, 2H), 2.23-1.95 (m, 4H); MS: 413.2 (M 5 + H*). Scheme 3 Br N K 2 CO3, pyrrolidine N N N EtOH, 65 *C (74% yield) Method I: 3-(pyrroldin-1'-yl)-methyl benzonitrile: To a solution of 3-(bromomethyl) 10 benzonitrile (30.0 g, 1.00 equiv) in absolute EtOH (600 mL) was added pyrrolidine (13.3 mL, 1.00 equiv), followed by K2CO3 (anhydrous, 63.5 g, 3.00 equiv). The reaction was stirred vigorously at 65 0C until consumption of the bromide was complete (Reaction is monitored on Merck 254nm silica-coated TLC plates using a combination of EtOAc/hexane as eluent). The reaction (which may be orange-colored) was cooled to 23 15 *C and filtered over coarse glass frits, and the filtrate was concentrated. The resulting residue was partitioned between H 2 0 and EtOAc (300 mL each) and the organic phase collected. The aqueous layer was extracted (2 x 200 mL EtOAc). All of the resulting organic layers were combined, dried (Na 2 SO4), filtered, and concentrated in vacuo, giving the title nitrile (21.1 g, 74% yield) as an orange residue. 'H NMR (CDC13, 300 MHz): d 20 (ppm) 7.65 (s, 1H), 7.59 (d, J = 7.7 Hz, 1H), 7.54 (d, J = 7.6 Hz, 1H), 7.41 (dd, J = 7.7 Hz, 7.6 Hz, 1H), 3.65 (s, 2H), 2.52 (m, 4H), 1.81 (m, 4H). LCMS-ESI': calc'd for CH 5
N
2 : 187.1 (M+H*); Found: 187.1 (M+H*). Scheme 4 Br 0 K 2 C0s N 0 pyrroldine 25 EtOH Method II: 3-(pyrroldin-1'-yl)-methyl benzaldehyde: A suspension of K 2
CO
3 (2.09 g, 15.2 mmol, 3.00 equiv) in absolute ethanol (20 mL) was treated with pyrroldine (439 pL, 5.05 mmol, 1.00 equiv). 3-(bromomethyl)-benzaldehyde (1.00 g, 5.05 mmol, 1.00 equiv) 87 WO 2010/077613 PCT/US2009/067002 was introduced, and the reaction was heated to 65 *C for 1 h. The reaction was cooled and filtered. The cake was washed with more ethanol. The filtrate was concentrated to a cloudy oil and partitioned between DCM (50 mL) and 2% wlv aq NaHCO 3 (50 mL). The organic phase was collected, and the aq layer was extracted with DCM (2 x 50 mL). All 5 organic layers were combined, dried (Na 2 SO4), filtered, and concentrated, giving 3 (pyrrolidin-1-ylmethyl)-benzaldehyde (846 mg, 88% yield) as a pale yellow oil, which was used without further purification. 'H-NMR: 300 MHz, (CDC 3 ) d: 10.00 (s, IH), 7.84 (s, IH), 7.76 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.47 (dd, J = 7.6 Hz, 7.6 Hz, 1H), 3.69 (s, 2H), 2.52 (m, 4H), 1.79 (m, 4H). LCMS-ESl*: calc'd for C 1 2
H
18 NO: 190.1 (M+H*); 10 Found: 190.1 (M+H*). Scheme 5 N LiAIH 4 NH N Et20, 0 "C N NH2 (90% yield) Method Il: 3-(pyrrolidin-1'-yl)methyl benzylamine: A 1 Liter round-bottom flask was 15 charged with LiAIH 4 (7.55 g) and anhydrous Et 2 0 (230 mL). After cooling to 0 "C, 3 (pyrrolidin-1-ylmethyl)-benzonitrile (18.55 g) in THF (30 mL) was added slowly over a 5 min period. Rxn transitioned from orange to green. Once the reaction was complete (as indicated by TLC using Merck 254nm silica-coated plates with DCM/MeOH/aq. NH 4 0H eluent or by LCMS), it was slowly treated first with H 2 0 (7.5 mL) with sufficient time to 20 allow gas evolution to cease, second (after a 5 min wait past the end of gas evolution) with 15% w/v aq. NaOH (7.5 mL) (again allowing gas evolution to stop, followed by a 5 min wait), and finally with more H 2 0 (26.5 mL). The reaction was filtered over glass frits to remove all of the solids present, and the filter cake was washed with Et 2 0 (100 mL). The filtrate was dried with copious MgSO 4 , filtered, and concentrated, affording the title amine 25 (17.0 g, 90% yield) as an oil. 1 H NMR (CDC 3 , 300 MHz): d (ppm) 7.32-7.17 (m, 4H), 3.86 (s, 2H), 3.62 (s, 2H), 2.52 (m, 4H), 1.79 (m, 4H), 1.61 (s, broad, 2H). LCMS-ESl*: calc'd for C 1 2
H
1
N
2 : 191.1 (M+H*); Found: 191.0 (M+H*). Scheme 6 88 WO 2010/077613 PCT/US2009/067002 N Brk 0 -AOx N ~' N 2 Et 3 N '- N"-o- NH2 THFt23*C H H Method IV: Ethyl-Na-[3-(pyrrodin-1'-ylmethyl)-benzyl]-glycinate: A solution of 3 (pyrrolidin-1-ylmethyl)-benzylamine (17.0 g, 1.00 equiv) in THF (160 mL) was treated with Et 3 N (27.4 mL, 2.20 equiv). Ethyl bromoacetate (9.90 mL, 1.00 equiv) was added 5 dropwise to this solution at 23 0C over a 10 min period. After 24 hrs, the reaction was diluted with H 2 0 (600 mL) and extracted with EtOAc (3 x 150 mL). The organic layers were combined, dried (MgSO4), filtered, and concentrated, giving the title product as a yellow oil (21.2 g, 86%). 'H. NMR (CDCls, 300 MHz): d (ppm) 7.32-7.18 (m, 4H), 4.19 (q, J = 7.0 Hz, 2H), 3.80 (s, 2H), 3.61 (s, 2H), 2.51 (m, 4H), 1.79 (m, 4H), 1.28 (t, J = 7.0 Hz, 10 3H). LCMS-ESl*: calc'd for C 16
H
25
N
2 0 2 : 277.2 (M+H*); Found: 277.1 (M+H'). Scheme 7 OH OH N ~HN0 3 N N0 SI TFA '_1. OH S N OH Method V: 4,6-Dihydroxy-2-methylthio-5-nitropyrimidine: A solution of 4,6-dihydroxy 15 2-methylthiopyrimidine (42 g, 0.257 mol) in trifluoroacetic acid (91 ml, 1.186 mol) was stirred at 23*C and warmed until all solid had gone into solution. The reaction was stirred for five hours at 23*C. Next, fuming HNO 3 (15 ml, 350 mmol) was added portion wise to the reaction mixture over 25 minutes at 0 'C. The reaction was stirred for twenty hours at 23 *C, and treated with H 2 0 (at 23*C) at 80% conversion (according LC-MS). The solid 20 precipitate was captured via filteration giving 4,6-dihydroxy-2-methythio-5-nitropyrimidine as a tan-colored solid. The crude solid was azeotroped with toluene to give 35 g of pale tan powdery solid. 'H-NMR: 300 MHz, (CD 3 0D, 300 MHz) d (ppm) 2.63 (s, 3H). LCMS ES: calc'd for C 5
H
4
N
3 0 4 S: 202.0 (M-H-); Found: 202.0 (M-H-). 25 Scheme 8 OH Cl N l NO 2 CN3 NO 2 N,N-dimethyl I I N OH aniline S N Cl 89 WO 2010/077613 PCT/US2009/067002 Method VI: 4,6-Dichloro-2-methylthio-5-nitropyrimidine: A 500 mL round bottom flask was charged with POC 3 (89.5 mL, 0.960 mol, 5.00 equiv), and N,N-dimethylaniline (73.0 mL, 0.576 mol, 3.00 equiv). The reaction was cooled to 0 *C, and 4,6-dihydroxy-2 methylthio-5-nitropyrimidine (39.0 g, 0.192 mol, 1.00 equiv) was added portionwise in a 5 manner to control exotherm. Once the exotherm had subsided, the reaction was carefully warmed to 100 *C for 2 h. Reaction was then transferred to the upper reservoir of a continuous lower-density phase continuous extractor and extracted continuously with hot hexanes, which pooled in the lower reservoir. The lower reservoir was at 140 "C during extraction. After UV activity (254 nm) in the upper reservoir hexane phase was at its 10 minimum, the system was cooled. The hexane phase was concentrated to an oil in vacuo. The residue was purified via silica gel chromatography (1 g residue/ 3 g silica)(Eluent: DCM). During loading (20 mL DCM was added to residue to aid fluidity) onto the column, there was a mild exotherm. After chromatography, crystalline 4,6 dichloro-2-methylthio-5-nitropyrimidine 34.9 g (76% yield) was obtained. 1 H-NMR: 300 15 MHz, (CDC 3 ) d (ppm): 2.62 (s, 3H). LCMS-ESl*: compound does not ionize. Scheme 9 Cl
NH
2 N NO 2
NH
3 , Et 3 N N NO 2
CH
3 S N Cl CH 3 S N C Method VII, Part 1: 4-Amino-6-chloro-2-methylthio-5-nitropyrimidine: To a solution of 20 above dichloride (2.46 g, 10.2 mmol) in THF (34 mL) at - 20 *C was added Et 3 N (3.14 mL, 22.5 mmol) followed by a solution of NH 3 (2.0 M in MeOH, 5.4 mL, 11 mmol). The mixture was stirred while warming to 0 "C for 1.5 h (LC/MS indicated consumption of starting materials. Some bis-addition is observed). The reaction mixture was taken forward without work-up. 25 Scheme 10 HN- CO 2 Et NH2 NO2 N N NO 2 N
CH
3 S N N CO 2 Et
CH
3 S N Ct Et 3 N N> 90 WO 2010/077613 PCT/US2009/067002 Method Vil, Part 2: Ethyl-Na-[4-amino-2-methylthio-5-nitropyrimidin-6-y],Na-[3' (pyrrolidin-1"-ylmethyl)-benzyl]-glycinate: To the previous reaction mixture at 0 *C was added the secondary amine (2.82 g, 10.2 mmol) in THF (10 mL) over 5 min. The reaction mixture was stirred until LC/MS indicated the consumption of starting material, 5 approximately 30 min. The reaction was filtered over glass frits; the filter cake was washed with EtOAc. The filtrate was concentrated and partitioned between EtOAc (30 mL) and 5% aq Na 2
CO
3 (30 mL). The organic phase was collected, and the aqueous phase extracted twice more with EtOAc (30 mL each), The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum. Absolute EtOH (30 mL) was 10 added, and the material was concentrated again. The residue was taken up in a minimum of absolute EtOH at 70 *C (-12 mL), then the solution was allowed to cool gradually to 23 0 C. Crystals were filtered over glass frits and washed with hexane, then dried in vacuo. Product is a yellowish-green solid. 'H NMR (CDCI 3 , 300 MHz): d (ppm) 7.32-7.16 (m, 4H), 4.69 (s, 2H), 4.19 (q, J = 7 Hz, 2H), 4.07 (s, 2H), 3.60 (s, 2H), 2.49 (m, 4H), 2.40 (s, 3H), 15 1.78 (m, 4H), 1.23 (t, 3 H, J = 7 Hz). LCMS-ES*: calc'd for C 2 zH 2 9
NO
4 S: 461.2 (M+H'); Found: 461.0 (M+H*). Scheme 11
NH
2
NH
2 N NO 2
H
2 0 2 , HOAc N NO 2
CHI
3 S N N-'CO 2 Et Na 2 W0 4 *2H 2 0 MeO 2 S N N-^'CO 2 Et N - N 20 Method Vill: Ethyl-N,-[4-amino-2-methanesulfonyl-5-nitropyrimidin-6-y],N
.
-[3' (pyrrolidin-1"-ylmethyl)-benzyl]-glycinate: To a solution a suspension of the sulfide (3.68 g, 8.00 mmol) in EtOH (40 mL) at 0 'C was added sodium tungstate dihydrate (792 mg, 2.40 mmol), acetic acid (4.6 mL, 80 mmol), and hydrogen peroxide (3.4 mL, -40 mmol, 35% w/w in H 2 0) sequentially. After 3 h, additional acetic acid (4.6 mL) and 25 hydrogen peroxide (3.4 mL) were added. The reaction was maintained at 0 *C for 16 h. A saturated solution of Na 2
SO
3 (50 mL) was added carefully while at 0 0 C followed by
CH
2
CI
2 (75 mL). The layers were separated, and the aqueous layer was extracted with
CH
2
C
2 (4 x 50 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum and used without further purification. 30 Scheme 12 91 WO 2010/077613 PCT/US2009/067002 0 H 2 N GCO 2 Me HN 7 C0 2 Me N Na(OAc)3BH, N HOAc, MeOH Method IX: Methyl-a,a-(1"',2m'-ethylidene),N-[3-(pyrrodin-1'-ylmethyl)-benzyl] glycinate: To a solution of 3-(pyrrolidin-1'-yimethyl)-benzaldehyde (284 mg, 1.50 mmol) in MeOH (5 mL) was added acetic acid (258 tL, 4.50 mmol), sodium 5 triacetoxyborohydride (636 mg, 3.00 mmol), and methyl 1-aminocyclopropanecarboxylate hydrochloride (250 mg, 1.65 mmol) sequentially. The reaction mixture was stirred at room temperature for 2 h and was then poured onto brine (15 mL) and CH 2
CI
2 (15 mL). The layers were separated, and the aqueous layer was extracted with CH 2 CI2 (3 x 10 mL). The combined organic layers were dried (Na 2 SO4, filtered, and concentrated in vacuo 10 and the title product was taken on without further purification as in Method XV, Parts 1 and 2 (below). LCMS-ESI*: calc'd for C 17
H
2 5
N
2 0 2 : 289.4 (M+H*); Found: 289.1 (M+H). Scheme 13
NH
2 NH 2 N NO2 TFA N NO2 MeO 2 S N N CO 2 Et R'OH R N N CO2Et N 15 Method X: To a solution of sulfone (1.0 g, 2.0 mmol) in alcohol (R-OH) (10 mL) was added TFA (470 pL, 6.1 mmol). The reaction was stirred at 100 0C for 1 h. The reaction mixture was poured onto a saturated solution of NaHCO 3 (20 mL) and CH 2 Cl 2 (30 mL). The layers were separated, and the aqueous layer was extracted with CH 2
C
2 (30 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under 20 vacuum. Purification was conducted by silica gel chromatography (1 g substrate/10 g SiO 2 ) (2-15% MeOH/CH 2
C
2 ). Scheme 14 92 WO 2010/077613 PCT/US2009/067002
NH
2 NH 2 N - NO 2 TFA I DMF N NO 2 MeO 2 S N N-'~CO 2 Et R, ROIN N CO2Et N OHNN Method XI: To a solution of sulfone (1.0 g, 2.0 mmol) in alcohol (R-OH) (10 mL) was added DMF (1.0 mL) and TFA (470 pL, 6.1 mmol). The reaction was stirred at 90-100 0C for 1 h. The reaction mixture was poured onto a saturated solution of NaHCO 3 (20 mL) 5 and CH 2
CI
2 (30 mL). The layers were separated, and the aqueous layer was extracted with CH 2
CI
2 (30 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum. Purification was conducted by silica gel chromatography (1 g substrate/10 g SiO 2 ) (2-15% MeOH/CH 2
C
2 ). 10 Scheme 15
NH
2
NH
2 H N NO 2 N N 0 R' N N-CO2Et Raney Ni Rs N -N OIN N INXN N H 2 , MeOH N Method XII: To a solution of nitro compound (730 mg, 1.5 mmol) in MeOH (10 mL) was added a Raney Nickel (-200 ptL, slurry in H 2 0). The reaction vessel was flushed with H 2 and then stirred under an H 2 atmosphere for 1.5 h. The mixture was filtered through celite 15 with CH 2 C1 2 and MeOH (1:1). The filtrate was concentrated under vacuum and left on lyophilizer overnight. The title product was obtained as a free base is a white solid. Scheme 16
NH
2 NH 2 MeO2S' NO2 -- CO2Et R H RIsN -CO2Et 2 R2 R2 N 20 Method XAll: A suspension of the sulfone (50 mg), THF (1 .0 mL), and the amine
(R'R
2 NH) (100 pL was heated to 60 OC for 3 h. The reaction was cooled to 23 C and directly loaded to a C18-reversed phase column (50 mg / 4 g packing material) and 93 WO 2010/077613 PCT/US2009/067002 purified by LC (Eluent: neutral H 2 0/CH 3 CN 95:5 -- 0:100 -+ neutral CH 3 CN/MeOH 100:0 50:50) to provide the product. Scheme 17
NH
2 NH 2 H N , NO 2 N1_ N R1' N Raney Ni R N_ O N!N N__CO 2 Et N NN N H2, MeOH Method XIV: A solution of the nitro compound (50 mg) in MeOH (4.0 mL) was treated with Raney Nickel (-200 RL, slurry in H 2 0). The reaction vessel was flushed with H 2 and then stirred under an H 2 atmosphere for 1.5 h. The mixture was filtered through celite with
CH
2 Cl 2 and MeOH (1:1). The filtrate was concentrated and dried in vacuo, giving the 10 product as a free base. Occasionally, 1.0 M aq HCI (200 pL) was added to the filtrate prior to concentrating. This gave an HCl salt, which usually had sharper 1 H NMR resonances. Scheme 18 CI 1H Nt NO 2
NH
3 , MeOH N O 2 02 MeS N Cl MeS N Cl 15 Method XV, Part 1: 4-Amino-6-chloro-2-methylthio-5-nitropyrimidine: To a solution of 4,6-dichloro-2-(methylthio)-5-nitropyrimidine (327 mg, 1.36 mmol) in THF (5.4 mL) at - 10 *C was added Et 3 N (474 tL, 3.40 mmol) followed by a solution of NH 3 (2.0 M in MeOH, 750 gL, 1.5 mmol). The mixture was stirred while warming to 0 0 C for 1.5 h (LC/MS 20 indicated consumption of starting materials.). The reaction mixture was taken forward without work-up. Scheme 19 94 WO 2010/077613 PCT/US2009/067002 - NH2O HN CO 2 Me N NO N MeS N N CO 2 Me 0- 1 No Method XV, Part 2: Methyl-a,a -(1 "',2"'-ethylidene),N-[4-amino-2-methylthio-5 nitropyrimidin-6-yl],Na-[3'-(pyrrolidin-1"-yImethyl)-benzyl]-glycinate: To the previous reaction mixture at 0 0C was added the crude secondary amine (-1.5 mmol) in THF (1.5 5 mL). The reaction mixture was stirred at rt for 18 h then 60 *C for 6 h. A saturated solution of NH 4 CI (10 mL) was added. The layers were separated, and the aqueous layer was extracted with EtOAc (2 x 10 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum. Purification by silica gel chromatography (-1 g substrate/15 g SiO 2 ) (2-20% MeOH/DCM) provided the product. LCMS-ESt*: calc'd for 10 C 22
H
2 9 Ne0 4 S: 473.6 (M+H*); Found: 473.1 (M+H). Scheme 20 Methyl N Bromoacetate N
NH
2 Et 3 N N 0x ,: rTHF H0 Method XVI: To a solution of 3-((1-pyrrolidinylmethyl)phenyl)methanamine (1.95 g, 10.2 15 mmol) in THF (34 mL) at 0 IC was added Et 3 N (3.14 mmol, 22.5 mmol) followed by methyl bromoacetate (1.04 mL, 22.3 mmol) dropwise. The reaction mixture was stirred until LOIMS indicated consumption of starting materials, approximately 2 h. The product mixture was taken forward without work up. LCMS-ESl*: calc'd for C 15
H
2 3
N
2 0 2 : 263.4 (M+H*); Found: 263.1 (M+H). 20 Compound G: Prepared using method Vill
NH
2 N NO 2 MeO 2 S N) N CO2Me N 95 WO 2010/077613 PCT/US2009/067002 Methyl-a,a-(1 "',2"'-ethylidene),Na-[4-amino-2-methanesulfonyl-5-nitropyrimidin-6 yl],N.-[3'-(pyrrolidin-l"-ylmethyl)-benzyl]-glycinate: LCMS-ES*: calc'd for C22H 29
N
6
O
6 S: 505.6 (M+H 4 ); Found: 505.2 (M+H). 5 Compound H: Prepared using Method X
NH
2 N N N CO2Me N Methyl-a,a -(I "',2"'-ethylidene),N.-[4-amino-2-n-butoxy-5-nitropyrimidin-6-y],N-[3' (pyrrolidin-l"-ylmethyl)-benzyl]-glycinate: LCMS-ESI*: calcd for C 25
H
35
N
6 0 5 : 499.6 (M+H*); Found: 499.2 (M+H). 10 Example 3: Prepared using Method Xli
NH
2 H O"-1 N No 4-Amino-2-n-butoxy-7-(1 "',2"'-ethylidene)-8-[3'-(pyrrolidin-1"-ylmethyl)-benzyl] 5,6,7,8-tetrahydropteridine-6-one: 'H-NMR: 300 MHz, (CD3OD) d: 7.39-7.60 (m, 4H), 15 4.91 (s, 2H), 4.30-4.41 (m, 4H), 3.47 (m, 2H), 3.18 (m, 2H), 2.18 (m, 2H), 2.03 (m, 2H), 1.65 (m, 2H), 1.42 (m, 2H), 0.79-0.98 (m, 7H) - [HCI salt]. LCMS-ESI*: calc'd for
C
24
H
33
N
6 0 2 : 437.6 (M+H*); Found: 437.2 (M+H). Compound I: Prepared using Method XV, Parts 1 and 2
NH
2 N NO 2 MeS NIN CQ 2 Et 20 N 96 WO 2010/077613 PCT/US2009/067002 Ethyl-Na[4-amino-2-methylthio-5-nitropyrimidin-6-y,N1[4'-(pyrrolidin-1 "-ylmethyl) benzyl]-glycinate: 'H-NMR: 300 MHz, (DMSO-d 6 ) d: 7.22-7.25 (m, 4H), 4.64 (s, 2H), 4.08 (m, 2H), 3.54 (s, 2H), 3.31 (s, 2H), 2.39 (s, 3H), 2.32 (m, 4H), 1.66 (m, 4H), 1.16 (t, J = 7 Hz, 3H). LCMS-ES*: calc'd for C 21
H
2 9N 6
O
4 S: 461.6 (M+H 4 ); Found: 461.2 (M+H). 5 Compound J: Prepared using method Vill
NH
2 N
NO
2 MeO 2 S N O CO 2 Et N Ethyl-N 8 -[4-amino-2-methanesulfonyl-5-nitropyrimidin-6-y],N-[4'-(pyrrolidin-1" ylmethyl)-benzyll-glycinate: LCMS-ESI*: calc'd for C 21
H
2 9
N
6 OS: 493.6 (M+H*); Found: 10 493.2 (M+H). Compound K: Prepared using Method X:
NH
2 N NOCO 2 Et N Ethyl-N[4-amino-2-n-butoxy-5-nitropyrimidin-6-yt],N-[4'-(pyrrolidin-1"-ylmethyl) 15 benzyl]-glycinate: 'H-NMR: 300 MHz, (CD 3 0D) d: 7.32 (m, 4H), 4.75 (s, 2H), 4.13-4.24 (m, 6H), 3.67 (s, 2H), 2.59 (m, 4H), 1.82 (m, 4H), 1.66 (m, 2H), 1.40 (m, 2H), 1.25 (t, J = 7 Hz, 3H), 0.92 (m, 3H). LCMS-ESI*: calc'd for C 2 4
H
38
N
6 0 5 : 487.6 (M+H*); Found: 487.3 (M+H). 20 Example 4: Prepared using Method XII:
NH
2 H O NN o 97 WO 2010/077613 PCT/US2009/067002 4-amino-2-n-butoxy-8-[4'.(pyrrolidin-1"-ylmethyl)-benzyl]-5,6,7,8-tetrahydropteridin 6-one: 1 H-NMR: 300 MHz, (CD 3 0D) d: 7.47-4.62 (m, 4H), 4.94 (s, 2H), 4.38-4.46 (m, 4H), 4,13 (s, 2H), 3.48 (m, 2H), 3.20 (m, 2H), 2.17 (m, 2H), 2.02 (m, 2H), 1.75 (m, 2H), 1.43 (m, 2H), 0.94 (t, J = 7 Hz, 3H). LCMS-ESI t : calc'd for C 22
H
31
N
6 0 2 : 411.5 (M+H*); 5 Found: 411.2 (M+H). Compound L: Prepared using Method X:
NH
2 SN NO CO 2 Me NN 10 Methyl-Na-[4-amino-2-{(cyclopropyl)methoxy}-5-nitropyrimidin-6-yll,Na-[3' (pyrrolidin-1"-ylmethyl)-benzyl]-glycinate: 1 H NMR (CD 3 0D, 300 MHz): 5 7.22-7.32 (m, 4H), 4.76 (s, 2H), 4.16 (s, 2H), 4.02 (d, J = 7 Hz, 2H), 3.73 (s, 3H), 3.64 (s, 2H), 2.53 (m, 4H), 1.80 (m, 4H), 1.16 (m, IH), 0.55 (m, 2H), 0.28 (m, 2H). LCMS-ESF*: calc'd for
C
23
H
31
N
6 0 5 : 471.5 (M+H*); Found: 471.2 (M+H*). 15 Example 5: Prepared using Method Xii:
NH
2 H N O1N N N 4-amino-2-{(cyclopropyl)methoxy)-8-[3'-(pyrrolidin-1"-ylmethyl)-benzyl]-5,6,7,8 tetrahydropteridin-6-one: 1 H NMR (CD 3 0D, 300 MHz): 5 7.64 (s, 1H), 7.50 (m, 3H), 20 4.95 (s, 2H), 4.39 (s, 2H), 4.26 (d, J = 7 Hz, 2H), 4.15 (s, 2H), 3.47 (m, 2H), 3.19 (m, 2H), 2.17 (m, 2H), 2.04 (m, 2H), 1.13 (m, IH), 0.59 (m, 2H), 0.34 (m, 2H) - [HCI salt]. LCMS ESI: calc'd for C 22
H
29
N
6 0 2 : 409.5 (M+H*); Found: 409.2 (M+H*). Compound M: Prepared using Method X: 98 WO 2010/077613 PCT/US2009/067002
NH
2 OIN N CO 2 Me ~- N Methyl-N"[4-amino-2-{(1'-methylcycloprop-1"'-yl)methoxy}-5-nitropyrimidin-6 yl],Ng[3'-(pyrrolidin-l"-ylmethyl)-benzyl]-glycinate: 'H NMR (CD 3 0D, 300 MHz): 6 7.25-7.33 (m, 4H), 4.75 (s, 2H), 4.16 (s, 2H), 3.99 (s, 2H), 3.73 (s, 3H), 3.67 (s, 2H), 2.57 5 (m, 4H), 1.81 (m, 4H), 1.16 (s, 3H), 0.48 (m, 2H), 0.39 (m, 2H). LCMS-ESl*: calc'd for
C
24 H3-N 6
O
5 : 485.6 (M+H*); Found: 485.2 (M+H*). Example 6: Prepared using Method XII:
NH
2 H N NjO N N 01N,- N T 10 4-amino-2-{(1"'-methylcycloprop-1'"-yl)methoxy}-8-[3'-(pyrrolidin-1"-ylmethyl) benzyl]-5,6,7,8-tetrahydropteridin-6-one: 'H NMR (CD 3 0D, 300 MHz): 6 7.63 (s, 1H), 7.51 (m, 3H), 4.94 (s, 2H), 4.39 (s, 2H), 4.24 (s, 2H), 4.14 (s, 2H), 3.48 (m, 2H), 3.18 (m, 2H), 2.17 (m, 2H), 2.04 (m, 2H), 1.19 (s, 3H), 0.56 (m, 2H), 0.43 (m, 2H) - [HCI salt]. LCMS-ESI*: calc'd for C 23
H
3 oN 6 0 2 : 423.5 (M+H*); Found: 423.1 (M+H*). 15 Compound N: Prepared using Method X:
NH
2 O N N CO 2 Me ~- N Methyl-Na-[4-amino-2-{(cyclobutyl)methoxy}-5-nitropyrimidin-6-yl],N-[3'-(pyrrolidin 1"-ylmethyl)-benzyl]-glycinate: 'H NMR (CD 3 0D, 300 MHz): 6 7.22-7.32 (m, 4H), 4.77 20 (s, 2H), 4.16 (m, 4H), 3.74 (s, 3H), 3.64 (s, 2H), 2.67 (m, 1H), 2.54 (m, 4H), 2.08 (m, 2H), 1.95 (m, 2H), 1.83 (m, 6H). LCMS-ESl*: calc'd for C 24
H
33
N
6 0 5 : 485.6 (M+H*); Found: 485.2 (M+H*). 99 WO 2010/077613 PCT/US2009/067002 Example 7: Prepared using Method Xil:
NH
2 H NTO Oj N 4-amino-2-{(cyclobutyl)methoxy}-8-[3'-(pyrrolidin-1"-ylmethyl)-benzyl]-5,6,7,8 5 tetrahydropteridin-6-one: 'H NMR (CD 3 OD, 300 MHz): 5 7.63 (s, 1 H), 7.50 (m, 3H), 4,96 (s, 2H), 4.39 (m, 4H), 4.16 (s, 2H), 3.47 (m, 2H), 3.19 (m, 2H), 1.85-2.17 (m, 11H) [HCl salt]. LCMS-ESI*: calc'd for C 23
H
31
N
6 0 2 : 423.5 (M+H*); Found: 423.2 (M+H*). Compound 0: Prepared using Method X:
NH
2 N NO 2 o N N CO 2 Me 10 Methyl-Na-[4-amino-2-{(cyclopentyl)methoxy}-5-nitropyrimidin-6-y],N-[3' (pyrrolidin-1"-ylmethyl)-benzyl]-glycinate: 1 H NMR (CD 3 0D, 300 MHz): 6 7.21-7.31 (m, 4H), 4.76 (s, 2H), 4.15 (s, 2H), 4.06 (d, J = 7 Hz, 2H), 3.73 (s, 3H), 3.61 (s, 2H), 2.51 (m, 4H), 2.26 (m, 1H), 1.79 (m, 4H), 1.58 (m, 4H), 1.29 (m, 4H). LCMS-ESl*: calc'd for 15 C 25
H
3 5N 6 O5: 499.6 (M+H*); Found: 499.2 (M+H 4 ). Example 8: Prepared using Method XII:
NH
2 H N 1 N N 4-amino-2-{(cyclopentyl)methoxy}-8-[3'-(pyrrolidin-1"-ylmethyl)-benzyl]-5,6,7,8 20 tetrahydropteridin-6-one: 'H NMR (CD 3 OD, 300 MHz): & 7.65 (s, 1H), 7.50 (m. 3H), 4.95 (s, 2H), 4.39 (s, 2H), 4.31 (d, J = 7 Hz, 2H), 4.16 (s, 2H), 3.47 (m, 2H), 3.19 (m, 2H), 100 WO 2010/077613 PCTIUS2009/067002 2.33 (m, 1 H), 2.17 (m, 2H), 2.03 (m, 2H), 1.77 (m, 2H), 1.60 (m, 4H), 1.33 (m, 2H) - {HCI salt]. LCMS-ESI*: calo'd for C 24
H
33
N
6 0 2 : 437.6 (M+H*); Found: 437.2 (M+H*). Compound P: Prepared using Method X:
NH
2 O NO CO 2 Me 5 r N Methyl-N.-[4-amino-2-{2'"-(cyclopropyl)ethoxy}-5-nitropyrimidin-6-yIL],Na-[3' (pyrrolidin-1"-ylmethyl)-benzyl}-glycinate: 1 H NMR (CD 3 0D, 300 MHz): 6 7,21-7.31 (m, 4H), 4.76 (s, 2H), 4.26 (t, J = 7 Hz, 2H), 4.16 (s, 2H), 3.73 (s, 3H), 3.62 (s, 2H), 2.50 (m, 4H), 1.79 (m, 4H), 1.56 (q, 2 H, 7 Hz), 0.76 (m, 1H), 0.44 (m, 2H), 0.08 (m, 2H). 10 LCMS-ESl*: calc'd for C 2 4
H
33
N
6 0 5 : 485.6 (M+H*); Found: 485.2 (M+H*). Example 9: Prepared using Method XII:
NH
2 H N O N NN 'c No 4-amino-2-{2"'-(cyclopropyl)ethoxy}-8-[3'-(pyrrolidin-I "-ylmethyl)-benzyl]-5,6,7,8 15 tetrahydropteridin-6-one: 1 H NMR (CD 3 0D, 300 MHz): 5 7.67 (s, 1H), 7.50 (m, 3H), 4.95 (s, 2H), 4.50 (t, J = 7 Hz, 2H), 4.40 (s, 2H), 4.17 (s, 2H), 3.49 (m, 2H), 3.19 (m, 2H), 2.17 (m, 2H), 2.04 (m, 2H), 1.63 (q, J = 7 Hz, 2H), 0.80 (m, 1H), 0.44 (m, 2H), 0.05 (m, 2H) - [HCI salt]. LCMS-ESI*: calc'd for C 23
H
31
N
6 0 2 : 423.5 (M+H*); Found: 423.2 (M+H*). 20 Compound Q: Prepared using Method X:
NH
2 O N N O 2 Me 10 101 WO 2010/077613 PCT/US2009/067002 'H NMR (CD 3 OD, 300 MHz): § 7.32-7.39 (m, 4H), 4.77 (s, 2H), 4.19 (s, 2H), 3.96 (d, J = 7 Hz, 2H), 3.89 (s, 2H), 3.74 (s, 3H), 2.81 (m, 4H), 2.00 (m, 1 H), 1.92 (m, 4H), 0.95 (d, 6 H, J = 7 Hz). LCMS-ESl*: calc'd for C 23
H
33
N
6 0 5 : 473.5 (M+H*); Found: 473.2 (M+H*). 5 Example 10: Prepared using Method XII:
NH
2 H N N O O N N . 'o N 'H NMR (CD 3 OD, 300 MHz): 8 7.64 (s, 1H), 7.49 (m, 3H), 4.96 (s, 2H), 4.39 (s, 2H), 4.20 (d, J = 7 Hz, 2H), 4.15 (s, 2H), 3.47 (m, 2H), 3.19 (m, 2H), 2.16 (m, 2H), 2.04 (m, 3H), 0.97 (d, 6 H, J= 6 Hz) - [HCI salt]. LCMS-ESl*: calc'd for C 22
H
31
N
6 0 2 : 411.5 (M+H*); 10 Found: 411.2 (M+H'). Compound R: Prepared using Method X:
NH
2
NO
2 O N N CO 2 Me ~-N 'H NMR (CD 3 OD, 300 MHz): 8 7.22-7.32 (m, 4H), 4.77 (s, 2H), 4.22 (t, J = 7 Hz, 2H), 4.16 15 (s, 2H), 3.73 (s, 3H), 3.64 (s, 2H), 2.54 (m, 4H), 1.80 (m, 4H), 1.75 (m, 1 H), 1.56 (q, J = 7 Hz, 2H), 0.92 (d, 6 H, J = 7 Hz). LCMS-ESI*: calc'd for C 24
H
35
N
6 Og: 487.6 (M+H*); Found: 487.2 (M+H*). Example 11: Prepared using Method XII:
NH
2 H NT ON N 20 N 'H NMR (CD 3 OD, 300 MHz): 6 7.67 (s, 1 H), 7.49 (m, 3H), 4.95 (s, 2H), 4.46 (t, J = 7 Hz, 2H), 4.40 (s, 2H), 4.16 (s, 2H), 3.47 (m, 2H), 3.17 (m, 2H), 2.16 (m, 2H), 2.02 (m, 2H), 102 WO 2010/077613 PCT/US2009/067002 1.72 (m, 1H), 1.64 (q, J = 7 Hz, 2H), 0.91 (d, 6 H, J= 7 Hz) - {HCI salt]. LCMS-ESI*: calc'd for C 23
H
33
N
6
O
2 : 425.5 (M+H'); Found: 425.3 (M+H*). Compound S: Prepared using Method X:
NH
2
NNXNO
2 NO NCO 2 Me 5 N 5 H NMR (CD 3 0D, 300 MHz): 6 7.25-7.33 (m, 4H), 4.77 (s, 2H), 4.16-4.22 (m, 4H), 3.73 (s, 3H), 3.66 (s, 2H), 2.56 (m, 4H), 1.82 (m, 4H), 1.70 (m, 2H), 1.37 (m, 4H), 0.92 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 24
H
35
N
6 0 5 : 487.6 (M+H'); Found: 487.2 (M+H*). 10 Example 12: Prepared using Method XII:
NH
2 H N N O N NN 'H NMR (CD 3 0D, 300 MHz): 8 7.65 (s, 1H), 7.50 (m, 3H), 4.96 (s, 2H), 4.40 (m, 4H), 4.16 (s, 2H), 3.48 (m, 2H), 3.19 (m, 2H), 2.18 (m, 2H). 2.03 (m, 2H), 1.76 (m, 2H), 1.36 (m, 4H), 0.91 (t, J = 7 Hz, 3H) - {HCI salt]. LCMS-ESIl: calc'd for C 23
H
33
N
6
O
2 : 425.5 (M+H*); 15 Found: 425.3 (M+H*). Compound T: Prepared using Method X:
NH
2 O1 NO C2Me N N 'H NMR (CD 3 OD, 300 MHz): 6 7.24-7.32 (m, 4H), 4.77 (s, 2H), 4.16 (s, 2H), 3.99 (d, J = 7 20 Hz, 2H), 3.74 (s, 3H), 3.63 (s, 2H), 2.52 (m, 4H), 1.67-1.82 (m, 9H), 1.25 (m, 4H), 1.00 (m, 2H). LCMS-ESI': calc'd for C 26
H
37 Nr0 5 : 513.6 (M+H*); Found: 513.2 (M+H*). 103 WO 2010/077613 PCT/US2009/067002 Example 13: Prepared using Method XII:
NH
2 H 0 A,<- N - N k-/ 'H NMR (CD 3 0D, 300 MHz): 5 7.65 (s, 1H), 7.50 (m, 3H), 4.95 (s, 2H), 4.40 (s, 2H), 4.22 (d, J = 7 Hz, 2H), 4.16 (s, 2H), 3.47 (m, 2H), 3.19 (m, 2H), 2.17 (m, 2H), 2.03 (m, 2H), 5 1.76 (m, 5H), 1.23 (m, 4H), 1.04 (m, 2H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 5
H
3 5
N
6 0 2 : 451.6 (M+H*); Found: 451.3 (M+H*). Compound U: Prepared using Method X:
NH
2 O N N CO 2 Me ' j N 10 'H NMR (CD 3 0D, 300 MHz): 5 7.27-7.34 (m, 4H), 4.76 (s, 2H), 4.17 (s, 2H), 3.88 (s, 2K), 3.74 (s, 3H), 3.65 (s, 2H), 2.54 (m, 4H), 1.80 (m, 4H), 0.97 (s, 9H). LCMS-ESI*: calc'd for C24H34N605: 487.6 (M+H*); Found: 487.2 (M+H*). Example 14: Prepared using Method XII:
NH
2 H N0 O N N 15 N 'H NMR (CD 3 OD, 300 MHz): 5 7.65 (s, 1H), 7.50 (m, 3H), 4.96 (s, 2H), 4.39 (s, 2H), 4.16 (s, 2H), 4.11 (s, 2H), 3.48 (m, 2H), 3.19 (m, 2H), 2.17 (m, 2H), 2.04 (m, 2H), 1.00 (s, 9H) [HCI salt]. LCMS-ESIl: caic'd for C 2 3
H
3 3
N
8 0 2 : 425.5 (M+H*); Found: 425.2 (M+H*). 20 Compound V: Prepared using Method X: 104 WO 2010/077613 PCT/US2009/067002
NH
2 N
~NO
2 0 N N CO 2 Et ~NN 'H NMR (CD 3 0D, 300 MHz): {all resonances were rather broad] 8 7.33 (9H), 5.26 (2H), 4.78 (2H), 4.17 (4H), 3.94 (2H), 2.86 (4H), 1.90 (4H), 1.23 (3H). LCMS-ESI*: calc'd for
C
2 7
H
33
N
6 0 5 : 521.6 (M+H*); Found: 521.2 (M+H*). 5 Example 15: Prepared using Method XII:
NH
2 H S NN 0 ~ N (N~ 11H NMR (CD 3 0D, 300 MHz): 8 7.31-7.59 (m, 9H), 5.46 (s, 2H), 4.97 (s, 2H), 4.35 (s, 2H), 4.14 (s, 2H), 3.44 (m, 2H), 3.13 (m, 2H), 2.14 (m, 2H), 2.00 (m, 2H) - [HCI salt]. LCMS 10 ESI*: calc'd for C 2 6
H
2 gN 6
O
2 : 445.5 (M+H*); Found: 445.2 (M+H*). Compound W: Prepared using Method X:
NH
2 0 N N CO 2 Et N c N 'H NMR (CD 3 OD, 300 MHz): [all resonances were rather broad] 6 8.54 (2H), 7.87 (1H), 15 7.43 (1H), 7.27 (4H), 5.33 (2H), 4.77 (2H), 4.15 (4H), 3.64 (2H), 2.54 (4H), 1.79 (4H), 1.23 (3H). LCMS-ESl*: calc'd for C 26
H
32
N
7 0 5 : 522.6 (M+H*); Found: 522.2 (M+H*). Example 16: Prepared using Method XIl: 105 WO 2010/077613 PCT/US2009/067002
NH
2 H NyO S0 N N NN N 'H NMR (CD 3 0D, 300 MHz): [all resonances were rather broad] 5 9.04 (1 H), 8.78 (2H), 8.06 (1H), 7.62 (1H), 7.48 (3H), 5.77 (2H), 4.91 (2H), 4.38 (2H), 4.12 (2H), 3.45 (2H), 3.16 (2H), 2.14 (2H), 2.01 (2H) - [HCI salt]. LCMS-ESI*: calc'd for C 24
H
28
N
7 0 2 : 446.5 5 (M+H*); Found: 446.2 (M+H*). Compound X: Prepared using Method X: NH2 N '-NO 2 J o N N CO 2 Et 0 No 'H NMR (CD30D, 300 MHz): 5 7.35 (s, 1H), 7.29 (m, 3H), 4.77 (s, 2H), 4.16 (m, 6H), 3.81 10 (m, 2H), 3.75 (s, 2H), 3.36 (s, 2H), 2.65 (m, 5H), 2.04 (m, 1H), 1,84 (m, 4H), 1.65 (m, 1 H), 1.24 (m, 3H). LCMS-ESI*: calc'd for C 25
HN
6 0 6 : 515.6 (M+H*); Found: 515.2 (M+H*). Example 17: Prepared using Method XII:
NH
2 H ONNN0 ON N 15 'H NMR (CD 3 0D, 300 MHz): 5 7.68 (s, 1H), 7.48 (s, 3H), 4.92 (s, 2H), 4.39 (m, 4H), 4.15 (s, 2H), 3.63-3.82 (m, 4H), 3.47 (m, 2H), 3.16 (m, 2H), 2.70 (m, 1H), 2.01-2.14 (m, 5H), 1.68 (m, 1H) - [HCI sat]. LCMS-ESI': calc'd for C 2
H
31 N60 3 : 439.5 (M+H'); Found: 439.3 (M+H*). 20 Compound Y: Prepared using Method X: 106 WO 2010/077613 PCT/US2009/067002
NH
2 O NN NO2 (MeO) 2 P-5xO N N CO 2 Et N -H NMR (CD 3 0D, 300 MHz): 8 7.37 (s, 1H), 7.31 (m, 3H), 4.79 (s, 2H), 4.44 (m, 2H), 4.18 (m, 4H), 3.83 (s, 2H), 3.75 (m, 3H), 3.35 (m, 3H), 2.74 (m, 4H), 2.31 (m, 2H), 1.88 (m, 4H), 1.26 (m, 3H). LCMS-ESI*: calc'd for C 24
H
3
N
6
O
8 P: 567.5 (M+H*); Found: 567.2 5 (M+H*). Example 18: Prepared using Method XII:
NH
2 H o N N 0 MeO' 0 N N OMe O e' - N H NMR (CD 3 0D, 300 MHz): 5 7.69 (s, 1H), 7.49 (s, 3H), 4.96 (s, 2H), 4.66 (m, 2H), 4.40 10 (s, 2H), 4.17 (s, 2H), 3.71 (d, 6 H, J= 11 Hz), 3.48 (m, 2H), 3.16 (m, 2H), 2.42 (m, 2H), 2.16 (m, 2H), 2.03 (m, 2H) - [HCI salt]. LCMS-ESI*: caic'd for C22H 32
N
6
O
5 P: 491.5 (M+H t ); Found: 491.2 (M+H*). Compound Z: Prepared using Method X:
NH
2 N NN.N 0 2 \NN O NO CO 2 Et 15 N 'H NMR (CD 3 OD, 300 MHz): 5 7.66 (s, 1H), 7.32 (s, 1H), 7.27 (m, 3H), 7.16 (s, 1H), 6.96 (s, IH), 4.77 (s, 2H), 4.47 (m, 2H), 4.32 (m, 2H), 4.18 (m, 4H), 3.72 (s, 2H), 2.61 (m, 2H), 1.82 (m, 2H), 1.24 (m, 3H). LCMS-ESI*: calc'd for C 2 5 HaaN 8
O
5 : 525.6 (M+H*); Found: 525.2 (M+H 4 ). 20 Example 19: Prepared using Method XII: 107 WO 2010/077613 PCTIUS2009/067002
NH
2 H NO N O I NN 'H NMR (CD 3 0D, 300 MHz): 5 9.17 (s, broad, 1H), 7.63-7.80 (m, 3H), 7.49 (m, 3H), 4.93 (s, 2H); 4.73 (s, broad, 2H), 4.39 (m, broad, 4H), 4.15 (s, 2H), 3.47 (m, 2H), 3.18 (m, 2H), 2.17 (m, 2H), 2.02 (m, 2H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 3
H
2 8
N
8 0 2 : 449.5 (M+H 4 ); 5 Found: 449.2 (M+H*). Compound AA: Prepared using Method X:
NH
2 MeO 2 S- O NO CO 2 Et N N N 'H NMR (CDaOD, 300 MHz): 6 7.40-7.47 (m, 4H), 4.81 (s, broad, 2H), 4.61 (s, 2H), 4.19 10 (m, broad, 6H), 3.50 (s, broad, 2H), 3.12 (m, 4H), 3.02 (s, 3H), 2.01 (m, 4H), 1.26 (m, 3H). LCMS-ESI*: calc'd for C 2 3
H
3 3 N80 7 S: 537.6 (M+H*); Found: 537.2 (M+H*). Example 20: Prepared using Method XII:
NH
2 H O N N 0 N N NN 15 'H NMR (CD 3 OD, 300 MHz): 5 7.74 (s, I H), 7.48 (s, 3H), 4.94 (s, 2H), 4.90 (s, 2H), 4.39 (s, 3H), 4.17 (s, 2H), 3.61 (m, broad, 2H), 3.48 (m, 2H), 3.14 (m, 2H), 3.06 (s, 3H), 2.13 (m, 2H), 2.01 (m, 2H) - [HCI salt]. LCMS-ESV*: calc'd for C 2 1
H
29
N
6 0 4 S: 461.6 (M+H*); Found: 461.2 (M+H*). 20 Compound AB: Prepared using Method X: 108 WO 2010/077613 PCT/US2009/067002
NH
2
NO
2 O N N CO 2 Et S N 'H NMR (CD 3 0D, 300 MHz): 6 7.23-7.34 (m, 4H). 5.20 (m, 1H), 4.77 (s, 2H), 4.19 (q, J 7 Hz, 2H), 4.16 (s, 2H), 3.68 (s, 2H), 2.58 (m, 4H), 1.73-1.87 (m, 10H), 1.60 (m, 2H), 1.26 (t, J = 7 Hz, 3H). LCMS-ESl*: calc'd for C 2 5
H
35 N,0 5 : 499.6 (M+H*); Found: 499,2 (M+H*). 5 Example 21: Prepared using Method XII:
NH
2 H O1 NO N 'H NMR (CD 3 0D, 400 MHz): 6 7.60 (s, 1H), 7.47 (m, 3H), 5.40 (m, 1H), 4.93 (s, 2H), 4.32 (s, 2H), 4.03 (s, 2H), 3.45 (m, 2H), 3.16 (m, 2H), 2.15 (m, 2H), 2.00 (m, 3H), 1.86 (m, 4H), 10 1.62-1.75 (m, 3H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 3
H
3 1
N
6 0 2 : 423.5 (M+H*); Found: 423.2 (M+H*). Compound AC: Prepared using Method X:
NH
2 0 N CO 2 Et "~ N 15 'H NMR (CD30D, 300 MHz): 5 7.40 (s, 2H), 7.33 (m, 3H), 4.79 (s, 2H), 4.36 (t, J = 5 Hz, 2H), 4.21 (m, 4H), 3.89 (s, 2H), 3.54 (m, 4H), 2.81 (m, 4H), 2.36 (t, J = 8 Hz, 2H), 2.02 (m, 2H), 1.90 (m, 4H), 1.26 (t, J = 7 Hz, 3H). LCMS-ESI*: cac'd for C 26
H.
3 NT0 6 : 542.6 (M+H*); Found: 542.2 (M+H*). 20 Example 22: Prepared using Method XII: 109 WO 2010/077613 PCT/US2009/067002
NH
2 H 0 NpO N~~NoN, N N 'H NMR (CD 3 0D, 400 MHz): 8 7.64 (s, 1H), 7.47 (s, 3H), 4.94 (s, 2H), 4.55 (m, 2H), 4.36 (s, 2H), 4.14 (s, 2H), 3.61 (m, 2H), 3.54 (t, 2 H, J = 5 Hz), 3.45 (m, 2H), 3.15 (m, 2H), 2.37 (t, J = 6 Hz, 2H) 2.13 (m, 2H), 2.02 (m, 4H) - [HCI salt]. LCMS-ESI t : calc'd for 5 C 2 4
H
31
N
7 0 3 : 466.6 (M+H*); Found: 466.1 (M+H"). Compound AD: Prepared using Method X:
NH
2 N NO 2 OI ON N CO 2 Et '~N 1 H NMR (CD 3 0D, 300 MHz): 8 7.47 (s, 1H), 7.37 (m, 3H), 7.27 (t, 2 H, J = 8 Hz), 6.92 (m, 10 3H), 4.80 (s, 2H), 4.54 (t, J= 5 Hz, 2H), 4.12-4.22 (m, 8H), 3.07 (m, 4H), 1.99 (m, 4H), 1.25 (t, J = 7 Hz, 3H). LCMS-ESr*: calc'd for C 2 8
H
35
N
6 0 6 : 551.6 (M+H*); Found: 551.2 (M+H*). Example 23: Prepared using Method Xil:
NH
2 H N N O N>N O~s.-- ,, NN 15N 'H NMR (CD30D, 300 MHz): 8 7.63 (s, 1H), 7.46 (s, 3H), 7.24 (t, 2 H, J = 6 Hz), 6.92 (t, J = 6 Hz, 1H), 6.86 (d, J = 6 Hz, 2H), 4.91 (s, 2H), 4.76 (s, broad, 2H), 4.33 (s, 2H), 4.26 (m, 2H), 4.14 (s, 2H), 3.43 (m, 2H), 3.12 (m, 2H), 2.11 (m, 2H), 1.98 (m, 2H) - [HCI salt). LCMS-ESI*: calc'd for C2 6
H
30
N
8 0 3 : 475.6 (M+H*); Found: 475.2 (M+H*), 20 Compound AE: Prepared using Method X: 110 WO 2010/077613 PCT/US2009/067002
NH
2 O N N CO 2 Et 'H NMR (CD 3 0D, 300 MHz): 6 7.26-7.37 (m, 4H), 4.99 (m, 1H), 4.78 (s, 2H), 4.20 (m, 4H), 3.77 (s, 2H), 2.68 (m, 4H), 1.85 (m, 4H), 1.50-1.62 (m, 2H), 1.29 (m, 2H), 1,25 (m, 6H), 0.90 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C2 5
H
3 7
N
6 0 5 : 501.6 (M+H*); Found: 5 501.2 (M+H*). Example 24: Prepared using Method XII:
NH
2 H H NO O N N b- N H NMR (CD 3 0D, 300 MHz): 8 7.64 (s, IH), 7.49 (m, 3H), 5.16 (m, 1H), 4.94 (s, 2H), 4.38 10 (s, 2H), 4.18 (s, 2H), 3.47 (m, 2H), 3.16 (m, 2H), 2.16 (m, 2H), 2.03 (m, 2H), 1.55-1.72 (m, 2H), 1.32 (m, 5H), 0.87 (t, J = 7 Hz, 3H) - [HCI salt]. LCMS-ESI: calc'd for
C
23
H
3 3
N
6 0 2 : 425,5 (M+H*); Found: 425.2 (M+H*). Compound AF: Prepared using Method X:
NH
2 OIN N CO 2 Et S N 15 H NMR (CD 3 0D, 300 MHz): 8 7.29-7.37 (m, 4H), 4.83 (m, 1H), 4.78 (s, 2H), 4.19 (m, 4H), 3.77 (s, 2H), 2.67 (m, 4H), 1.85 (m, 4H), 1.62 (m, 4H), 1.27 (t, J = 7 Hz 3H), 0.88 (t, 6 H, J = 7 Hz). LCMS-ESl*: calc'd for C 25
H
37
N
6 0 5 : 501.6 (M+H*); Found: 501.2 (M+H*). 20 Example 25: Prepared using Method XII: 111 WO 2010/077613 PCT/US2009/067002
NH
2 H ON N NN H NMR (CD 3 0D, 300 MHz): 5 7.60 (s, broad, IH), 7.49 (m, 3H), 4.94 (s, 2H), 4.39 (s, broad, 2H), 4.20 (s, 2H), 3.48 (m, 2H), 3.17 (m, 2H), 2.17 (m, 2H) 2.04 (m, 2H), 1.70 (m, 4H), 0.89 (m, broad, 6H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 3
H
3 N0 2 : 425.5 (M+H*); 5 Found: 425.2 (M+H'). Compound AG: Prepared using Method X (variation noted):
NH
2 S NO CO 2 Et > N Reaction was performed in CH 2
CI
2 without TFA at 40 0 C in sealed vial. 1 H NMR (CD 3 OD, 10 300 MHz): 6 7.20-7.32 (m, 4H), 4.78 (s, 2H), 4.20 (q, J = 7 Hz, 2H), 4.15 (s, 2H), 3.64 (s, 2H), 2.96 (t, 2 H, J =7 Hz), 2.54 (m, 4H), 1.80 (m, 4H), 1.60 (m, 2H), 1.42 (m, 2H), 1.26 (t, J = 7 Hz, 3H), 0.90 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 24
H
35
N
6 0 4 S: 503.6 (M+H*); Found: 503.2 (M+H*). 15 Example 26: Prepared using Method XII:
NH
2 H SIN N 'H NMR (CD 3 OD, 300 MHz): 8 7.61 (s, I H), 7.49 (m, 3H), 5.01 (s, 2H), 4.39 (s, 2H), 4.19 (s, 2H), 3.47 (m, 2H), 3.11 (m, 4H), 2.16 (m, 2H), 2.03 (m, 2H), 1.61 (m, 2H), 1.30 (m, 2H), 0,78 (t, J = 7 Hz, 3H). - [HCI salt]. LCMS-ESI*: calc'd for C 22
H
3
ON
6 0S: 427.6 (M+H 4 ); 20 Found: 427.2 (M+H*). Compound AH: Prepared using Method X: 112 WO 2010/077613 PCT/US2009/067002
NH
2 HON O N CO 2 Et N 'H NMR (CD 3 0D, 300 MHz): 5 7.29-7.36 (m, 4H), 4.77 (s, 2H), 4.16-4.25 (m, 6H), 3.77 (s, 2H), 3.57 (m, 2H), 2.68 (m, 4H), 1.85 (m, 4H), 1.75 (m, 2H), 1.58 (m, 2H), 1.26 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 24
H
35
N
6 0 6 : 503.6 (M+H*); Found: 503.2 (M+H*). 5 Example 27: Prepared using Method XI:
NH
2 H N N NN 1 H NMR (CD 3 0D, 300 MHz): 8 7.45-7.60 (m, broad, 4H), 4.96 (s, broad, 2H), 4.44 (m, broad, 2H), 4.19 (s, broad, 2H), 3.55 (s, 2H), 3,48 (m, 2H), 3.31 (s, broad, 2H), 3.18 (m, 10 broad, 2H), 2.15 (m, 2H), 2.03 (m, 2H), 1.81 (m, 2H), 1.58 (m, 2H) - [HCI salt]. LCMS ESI*: calc'd for C 2 2
H
31
N
6 0 3 : 427.5 (M+H*); Found: 427.2 (M+H*). Compound At: Prepared using Method X:
NH
2 N '- N0 2 HOO HO 0 N N CO 2 Et N N 15 1 H NMR (CD 3 OD, 300 MHz): 8 7.27-7.34 (n, 4H), 4.78 (s, 2H), 4.35 (t, J = 7 Hz, 2H), 4.20 (q, J = 7 Hz, 2H), 4.16 (s, 2H), 3.69 (s, 2H), 2.59 (m, 4H), 1.82-1.89 (m, 6H), 1.26 (t, J = 7 Hz, 3H), 1.22 (s, 6H). LCMS-ES*: calc'd for C 2 5
H
37
N
6 0 6 : 517.6 (M+H*); Found: 517.2 (M+H*). 20 Example 28: Prepared using Method XII: 113 WO 2010/077613 PCT/US2009/067002
NH
2 H NO HON ' O'Ni N N N 'H NMR (CD 3 OD, 300 MHz): 3 7.47-7.64 (m, broad, 4H), 4.94 (s, broad, 2H), 4.57 (m, broad, 2H), 4.41 (m, 2H), 4.19 (s, broad, 2H), 3.48 (m, 2H), 3.18 (m, 2H), 2.16 (m, 2H), 2.03 (m, 2H), 1.93 (m, 2H), 1.19 (s, 6H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 H 33NO 3 : 5 441.5 (M+H*); Found: 441.2 (M+H*). Compound AJ: Prepared using Method X:
NH
2 O N CO 2 Et ~- N 'H NMR (CD 3 OD, 300 MHz): 5 7.26-7.36 (m, 4H), 4.77 (s, 2H), 4.13-4.23 (m, 5H), 3.73 10 3.95 (m, 4H), 3.51 (m, 2H), 2.68 (m, 4H), 1.81-2.02 (m, 6H), 1.64 (m, 2H), 1.27 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 25
H
35
N
5 O: 515.6 (M+H*); Found: 515.2 (M+H1). Example 29: Prepared using Method XII:
NH
2 H N 0 N N 15 1 H NMR (CD 3 0D, 300 MHz): 5 7.66 (s, 1H), 7.49 (m, 3H), 4.96 (s, 2H). 4.37-4.47 (m. 4H), 4.18 (m, 1H), 4.16 (s, 2H), 3.80 (m, 2H), 3.48 (m, 2H), 3.17 (m, 2H), 2.16 (m, 2H), 2.01 (m, 2H), 1.92 (m, 2H), 1.70 (m, 2H) - {HCI salt], LCMS-ESI*: calo'd for C 2 3
H
31
N
6 0 3 : 439.6 (M+H*); Found: 439.2 (M+H*). 20 Compound AK: Prepared using Method X: 114 WO 2010/077613 PCT/US2009/067002
NH
2 tAN 0 N N CO 2 Et 'H NMR (CD30D, 300 MHz): 5 7.24-7.34 (m, 4H), 4.77 (s, 2H), 4.19 (q, J = 7 Hz, 2H), 4.16 (s, 2H), 4.05 (d, J = 7 Hz, 2H), 3.94 (m, 2H), 3.71 (s, 2H), 3.39 (m, 2H), 2.61 (m, 4H), 1.95 (m, 1H), 1.83 (m, 4H), 1.65 (m, 2H), 1.24-1.36 (m, 5H). LCMS-ESit: calc'd for 5 C 2 6
H
37
N
6 0 6 : 529.6 (M+H 4 ); Found: 529.2 (M+H*). Example 30: Prepared using Method XII:
NH
2 H N1 N N O N N N N 1 H NMR (CD 3 0D, 300 MHz): 6 7.67 (s, 1H), 7.49 (m, 3H), 4.96 (s, 2H), 4.40 (s, broad, 10 2H), 4.29 (d, J = 6 Hz, 2H), 4.16 (s, 2H), 3.95 (m, 2H), 3.48 (m, 2H), 3.40 (m, 2H), 3.17 (m, 2H), 2.16 (m, 2H), 1.98-2.07 (m, 3H), 1.65 (m, 2H), 1.34 (m, 2H) - [HCI salt]. LCMS ESI*: calc'd for C 24
H
3 3
N
6 0 3 : 453.6 (M+H*); Found: 453.2 (M+H*). Compound AL: Prepared using Method X:
NH
2 -NA2 O N N CO 2 Et 15 N 1 H NMR (CD30D, 300 MHz): 5 7.23-7.33 (m, 4H), 4.77 (s, 2H), 4.19 (q, 2 H, J= 7 Hz), 4.16 (s, 2H), 4.11 (d, J = 6 Hz, 2H), 3.66 (s, 2H), 2.56 (m, 4H), 1.80 (m, 4H), 1.58 (m, 1H), 1.41 (m, 4H), 1.28 (t, J = 7 Hz, 3H), 0.90 (t, J = 7 Hz, 6H). LCMS-ESI*: calc'd for
C
26
N
38 N,0 5 : 515.6 (M+H*); Found: 515.2 (M+H*). 20 Example 31: Prepared using Method XII: 115 WO 2010/077613 PCT/US2009/067002
NH
2 H ON N N 'H NMR (CD 3 OD, 300 MHz): 6 7.66 (s, 1H), 7.49 (m, 3H), 4.96 (s, 2H), 4.34-4,39 (m, 4H), 4.16 (s, 2H), 3.48 (m, 2H), 3.16 (m, 2H), 2.16 (m, 2H), 2.03 (m, 2H), 1.63 (m, 1H), 1.42 (m, 4H), 0.90 (t, J = 7 Hz, 6H) - [HCI salt]. LCMS-ESi*: calc'd for C 2 4
H
3 4
N
6 0 2 : 439.6 5 (M+H'); Found: 439.2 (M+H*). Compound AM: Prepared using Method XIII:
NH
2 N N0 2 -"NN NO CO 2 Et H N 'H NMR (CD 3 OD, 300 MHz): 6 7.34-7.20 (m, 4H), 4.74 (s, 2H), 4,17 (q, J = 7.0 Hz, 2H), 10 4.05-3.98 (m, broad, 2 lines, 2H), 3.63 (s, 2H), 3.23 (t, J = 6.7 Hz, 2H), 2.54 (m, 4H), 1.79 (m, 4H), 1.56-1.34 (m, 4H), 1.24 (t, J = 7.0 Hz, 3H), 0.89 (t, J = 7.4 Hz, 3H). LCMS-ESI*: calc'd for C 2 4
H
38
N
7 0 4 : 486.3 (M+H*); Found: 486.2 (M+H*), 243.7 ((M+2H*)/2). Example 32: Prepared using Method XIV:
NH
2 H wN 0 N N, N 15 N 'H NMR (CD 3 0D, 400 MHz): 6 7.56 (s, 1H), 7.46 (m, 3H), 4.90 (s, 1H), 4.37 (s, 1H), 4.08 (s, 1H), 3.46 (m, 2H), 3.32 (s, 1H) 3.29 (m, 2H), 3.16 (m, 2H), 2.14 (m, 2H), 2.01 (m, 2H), 1.51 (m, 2H), 1.32 (m, 2H), 0.86 (t, J = 7 Hz, 3H) - [HOI salt]. LCMS-ES*: calc'd for
C
2 2 H32N 7 O: 410.5 (M+H*); Found: 410.3 (M+H*). 20 Compound AN: Prepared using Method XIII: 116 WO 2010/077613 PCT/US2009/067002
NH
2 O N N CO 2 Et H H N 'H NMR (CD 3 0D, 300 MHz): 5 7.34-7.19 (m, 4H), 4.73 (s, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.10-3.95 (m, broad, 2 lines, 2H), 3.62 (s, 2H), 3.50 (m, 2H), 3.39 (m, 2H), 3.30 (s, 3H), 2.52 (m, 4H), 1.79 (m, 4H), 1.24 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for C 23
H
3 3
N
7 0 5 : 5 488.3 (M+H*); Found: 488.0 (M+H*), 244.6 ((M+2H)/2). Example 33: Prepared using Method XIV:
NH
2 H N N 0 O N N N H N 'H NMR (CD 3 0D, 400 MHz): 5 7.57 (s, 1H), 7.46 (m, 3H), 4.90 (s, 1H), 4.37 (s, 1H), 4.08 10 (s, 1H), 3.48 (m, 4H), 3.32 (s, IH), 3.30 (s, 3H), 3.16 (m, 2H), 2.14 (m, 2H), 2.00 (m, 2H) [HCI salt]. LCMS-ESi*: calc'd for C21H 30
N
7 0 2 : 412.5 (M+H*); Found: 412.2 (M+H*). Compound AO: Prepared using Method XIII:
NH
2 N N NO CO 2 Et '~ N 15 'H NMR (CD 3 OD, 300 MHz): 8 7.34-7.19 (m, 4H), 4.73 (s, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.15-3.96 (m, broad, 2 lines, 2H), 3.63 (s, 2H), 3.41-3.16 (m, broad, 2 lines, 2H), 2.53 (m, 4H), 1.79 (m, 4H), 1.25 (t, J = 7.0 Hz, 3H), 0.96-0.62 (m, 2 lines, broad, 9H). LCMS-ESl*: calc'd for C 2 5
H
3 8
N
7 0 4 : 500.3 (M+H*); Found: 500.1 (M+H*), 250.7 ((M+2H*)/2). 20 Example 34: Prepared using Method XIV: 117 WO 2010/077613 PCT/US2009/067002
NH
2 H N O N N N 'H NMR (CD 3 OD, 400 MHz): 6 7.56 (s, 1H), 7.46 (m, 3H), 4.90 (s, 1H), 4.36 (s, 1H), 4.08 (s, 1H), 3.43 (m, 2H), 3.32 (s, 1H), 3.17 (m, 2H), 3.16 (s, 2H), 2.16 (m, 2H), 2.01 (m, 2H), 0.87 (s, 9H) - [HCI salt]. LCMS-ESl*: calc'd for C2 3
H
34
N
7 0: 424.6 (M+H*); Found: 424.3 5 (M+H). Compound AP: Prepared using Method XIII:
NH
2 N
NO
2 N N N CO 2 Et H N 'H NMR (CD30D, 300 MHz): 8 7.36-7.20 (m, 4H), 4.75 (s, 2H), 4.17 (q, J = 7.0 Hz, 2H), 10 4.07 (app. s, broad, 2H), 3.62 (s, 2H), 2.67 (m, 1H), 2.53 (m, 4H), 1.79 (m, 4H), 1.23 (t, J = 7.0 Hz, 3H), 0.67 (m, 2H), 0.48 (m, 2H). LCMS-ESF: calc'd for C 23
H
32 Nr0 4 : 470.3 (M+H*); Found: 470.0 (M+H*), 235.6 ((M+2H t )/2). Example 35: Prepared using Method XIV:
NH
2 H N N N O NN H 15 N 'H NMR (CD 3 OD, 400 MHz): 8 7.60 (s, 1H), 7.46 (s, 3H), 4.89 (s, 1H), 4.37 (s, 1H), 4.06 (s, 1H), 3.46 (m, 2H), 3.29 (s, 1H), 3.16 (m, 2H), 2.63 (m, 1H), 2.14 (m, 2H), 2.01 (m, 2H), 0.87 (m, 2H), 0.64 (m, 2H) - {HCI salt]. LCMS-ES*: calc'd for C2 1
H
28
N
7 0: 394.5 (M+H*); Found: 394.2 (M+H*). 20 Compound AQ: Prepared using Method XIII: 118 WO 2010/077613 PCT/US2009/067002
NH
2 N ~NO 2 N N N CO 2 Et H N 'H NMR (CD 3 OD, 300 MHz): 8 7.34-7.20 (m, 4H), 4.73 (s, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.18-3.95 (m, broad, 2 lines, 2H), 3.61 (s, 2H), 2.51 (m, 5H), 1.83-1.53 (m, 6H), 1.79 (m, 4H), 1.39-1.09 (m, 7H). LCMS-ESI*: calc'd for C 2 6
H
38
N
7 0 4 : 512.3 (M+H*); Found: 512.1 5 (M+H*), 256.7 ((M+2H*)/2). Example 36: Prepared using Method XIV:
NH
2 H /J- H N N H N NN H NJMR (CD 3 0D, 400 MHz): 8 7.55 (s, 1H), 7.45 (m, 3H), 4.87 (s, 1H), 4.36 (s, 1H), 4.10 10 (s, 1H), 3.64 (m, IH), 3.44 (m, 2H), 3.32 (s, 1iH), 3.15 (m, 2H), 2.13 (m, 2H), 1.99 (m, 2H), 1.86 (m, 2H), 1.67 (m, 2H), 1.25 (m, 6H) - [HCI salt]. LCMS-ES*: calc'd for C2 4
H
34
N
7 0: 436.6 (M+H*); Found: 436.3 (M+H*). Compound AR: Prepared using Method XIII:
NH
2 0 NO2
I
NIN N"Y H~ 0 1.5 N 1 H NMR (CD 3 0D, 300 MHz): 5 7.38-7.21 (m, 4H), 4.73 (s, 2H), 4,17 (q, J = 7.0 Hz, 2H), 4.14-3.96 (m, broad, 2 lines, 2H), 3.65 (s, 2H), 3.40-3.25 (m, 3H), 3.29 (s, 3H), 2.55 (m, 4H), 1.80 (m, 4H), 1.24 (t, J = 7.0 Hz, 3H), 1.09 (d, J = 6.4 Hz, 3H). LCMS-ESI: calc'd for C 2 4
H
36
N
7 0 5 : 502.3 (M+H*); Found: 502.1 (M+H*), 251.6 ((M+2H*)/2). 20 119 WO 2010/077613 PCT/US2009/067002 Example 37: Prepared using Method XIV:
NH
2 H A N! N 0 N N N H N 'H NMR (CD 3 OD, 300 MHz): 5 7.55-7.40 (m, 4H), 4.91 (s, 1H), 4.37 (s, 1H), 4.08 (s, IH), 3.47 (m, 2H), 3.42-3.29 (m 1H), 3.37 (d, J = 4.9 Hz, 2H), 3.32 (s, 1H), 3.31 (s, 3H), 3.16 5 (m, 2H), 2.15 (m 2H), 2.01 (m, 2H), 1.16 (d, J = 6.8 Hz, 3H) - [HCI salt]. LCMS-ESI*: caic'd for C 22
H
3 2
N
7 0 2 : 426.3 (M+H'); Found: 426.2 (M+H'), 213.6 ((M+2H*)/2). Compound AS: Prepared using Method XIII:
NH
2 N NO 2 1 0 rN N N H0 No 10 'H NMR (CD 3 0D, 400 MHz): 8 7.60-7.36 (m, 4H), 6.49 (d, J = 2.2 Hz, 1H), 6.44 (d, J = 2.8 Hz, 1H), 6.40-6,26 (m, 1H), 4.80-4.73 (m, broad, 2 lines, 2H), 4.60-4.35 (m, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.16 (s, 2H), 4.16-4.08 (m, 2H), 3.06 (m, 4H), 1.98 (m, 4H), 1.25 (t, J = 7.0 Hz, 3H). LCMS-ESI: calc'd for C 25
H
32
N
7 0 5 : 510.2 (M+H*); Found: 510.1 (M+H'), 255.6 ((M+2H*)/2). 15 Example 38: Prepared using Method XIV:
NH
2 H NO N N N 0 NJ 'H NMR (CD 3 OD, 300 MHz): 8 7.60-7.40 (m, 4H), 6.40 (m, 1 H), 6.26 (app. d, J = 2,2 Hz, IH), 6.15 (app. d, J = 2.8 Hz, 1H), 4.91 (s, 1H), 4.49 (s, 1H), 4.36 (s, 1H), 4.34 (s, 1H), 20 4.07 (s, 1H), 3.56 (m, 2H), 3.32 (s, IH), 3.15 (m, 2H), 2.14 (m, 2H), 1.98 (m, 2H) - [HCI 120 WO 2010/077613 PCT/US2009/067002 salt]. LCMS-ESI*: calc'd for C 2 3
H
28
N
7 O2: 434.2 (M+H*); Found: 434.2 (M+H*), 217.5 ((M+2H 4 )/2). Compound AT: Prepared using Method XIII
NH
2 N N0 2 V-N N~ N 0-, H C 5~No 'H NMR (CD 3 0D, 400 MHz): 5 7.36-7.19 (m, 4HO, 4.71 (s, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.06-3.85 (m, broad, 2 lines, 2H), 3.61 (s, 2H), 3.20-3.00 (m, 2H), 2.51 (m, 4H), 1.79 (m, 4H), 0.90 (m, 1H), 0.40 (m, 2H), 0.13 (m, 2H). LCMS-ES*: calc'd for C 24
H
34
N
7 0 4 : 484.3 (M+H'); Found: 484.1 (M+H*), 242.7 ((M+2H*)/2). 10 Example 39: Prepared using Method XIV;
NH
2 H NI- N To N N N NINN 'H NMR (CD 3 0D, 300 MHz): 5 7.54-7.44 (m, 4H), 4.91 (s, 1H), 4.37 (s, 1H), 4.08 (S, 1H), 3.45 (m, 2H), 3.33 (s, 1H), 3.18 (d, J = 7.0 Hz, 2H), 3.16 (m, 2H), 2.15 (m, 2H), 1.99 (m, 15 2H), 1.06-0.97 (m, 1H), 0.48 (app. d, J = 7.6 Hz, 2H), 0.19 (app. d, J = 5.5 Hz, 2H) - [HCI salt). LCMS-ESI*: calc'd for C 22
H
30
N
7 0: 408.3 (M+H 4 ); Found: 408.2 (M+H*), 204.7 ((M+2H)/2). Compound AU: Prepared using Method XIII: 121 WO 2010/077613 PCT/US2009/067002
NH
2 , NO2 NN N H 0 O 'H NMR (CD 3 OD, 400 MHz): 8 7.34-7.19 (m, 4H), 4.71 (s, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.15-3.99 (m, broad, 2 lines, 2H), 3.62 (s, 2H), 3.50 (quintet, J = 6.4 Hz, 1H), 2.53 (m, 4H), 1.79 (m, 4H), 1.64 (m, 2H), 1.57 (m, 2H), 1.40 (m, 2H), 1.23 (t, J = 7.0 Hz, 3H). 5 LCMS-ESI: calc'd for C 24
H
3 4
N
7 0 4 : 484.3 (M+H*); Found: 484.2 (M+H*), 242.7 ((M+2H*)/2). Example 40: Prepared using Method XIV:
NH
2 H N N 0 N N N H N 10 'H NMR (CD 3 0D, 300 MHz): 5 7.50-7.40 (m, 4H), 4.80 (s, 1H), 4.34 (s, 1H), 4.22 (quintet, J = 8.4 Hz, 1H), 4.04 (s, IH), 3.44 (m, 2H), 3.30 (s, 1H), 3.14 (m, 2H), 2.24 (m, 2H), 2.13 (m, 2H), 2.03-1.88 (m, 4H), 1.68 (quintet, J = 8.9 Hz, 2H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 2
H
30
N
7 0: 408.3 (M+H*); Found: 408.2 (M+H*), 204.7 ((M+2H*)/2). 15 Compound AV: Prepared using Method XIII:
NH
2
-
NO
2 HN N N NIN-N ' No 122 WO 2010/077613 PCT/US2009/067002 'H NMR (CD 3 0D, 400 MHz): 8 7.34-7.19 (m, 4H), 4.71 (s, 2H), 4.20 (quintet, J = 5.6 Hz, IH), 4.17 (q, J = 7.0 Hz, 2H), 4.15-3.96 (m, broad, 2 lines, 2H), 3.75-3.62 (m, broad, 2 lines, 2H), 2.53 (m, 4H), 1.98-1.58 (m, 4H), 1.79 (m, 4H), 1.24 (m, 4H), 1.23 (t, J = 7.0 Hz, 3H). LCMS-ESl*: calc'd for C 2 5
H
3 6
N
7 0 4 : 498.3 (M+H*); Found: 498.2 (M+H*), 249.8 5 ((M+2H*)/2). Example 41: Prepared using Method XIV:
NH
2 H N N N N c H
-
NJ 'H NMR (CD 3 0D, 300 MHz): 6 7.51-7.40 (m, 4H), 4.92 (s, 1H), 4.37 (s, 1H), 4.08 (s, 1H), 10 3.48 (m, 2H), 3.30 (m, 1H), 3.19 (m, 2H), 2.17 (m, 2H), 2.08-1.86 (m, 4H), 1.79-1.63 (m, 2H), 1.63-1.45 (m, 4H) - [HCI salt]. LCMS-ESI*: calc'd for C 23
H
32
N
7 0: 422.2 (M+H t ); Found: 422.2 (M+H*), 211.7 ((M+2H*)/2). Compound AW: Prepared using Method XIII:
NH
2 N NO 15 N 'H NMR (CD 3 OD, 300 MHz): 5 7.40-7.20 (m, 4H), 4.76-4.71 (m, broad, 2 lines, 2H), 4.20 3.96 (m, 4H), 4.18 (q, J = 7.0 Hz, 2H), 4.01 (s, 2H), 3.73-3.65 (m, broad, 2 lines, 2H), 2.57 (m, 4H), 2.30 (quintet, J = 7.3 Hz, 2H), 1.81 (m, 4H), 1.25 (t, J = 7.0 Hz, 3H). LCMS-ESl*: calc'd for C 23
H
31
N
7 0 4 : 470.3 (M+H*); Found: 470.1 (M+H*), 235.6 ((M+2H*)/2). 20 Example 42: Prepared using Method XIV: 123 WO 2010/077613 PCT/US2009/067002
NH
2 H N 0 !N N 'H NMR (CD 3 0D, 300 MHz): 8 7.50-7.40 (m, 4H), 4.94 (s, 0.5H), 4.37 (s, 1H), 4.21 (app. t, J = 7.3 Hz, 2H), 4.09 (s, 0.5H), 4.05 (s, 1H), 3.60-3,48 (m, 3H), 3.32 (s, 1H), 3.20 (m, 2H), 2.45 (m, 1H), 2.17 (m, 2H), 1.98 (m, 2H) - [HCI salt]. LCMS-ESI*: calc'd for 5 C 21
H
28
N
7 0: 394.2 (M+H*); Found: 394.2 (M+H*), 197.7 ((M+2H*)/2), Compound AX: Prepared using Method XIII:
NH
2 N N02 N--N N N 'H NMR (CD 3 0D, 300 MHz): S 7.36-7.19 (m, 4H), 4.72 (s, 2H), 4.17 (q, J 7.0 Hz, 2H), 10 4.03 (s, 2H), 3.62 (s, 2H), 3.55-3.48 (m, 2H), 3.48-3.40 (m, 2H), 2.52 (m, 4H), 1.91 (m, 4H), 1.79 (m, 4H), 1.24 (t, J = 7.0 Hz, 3H). LCMS-ESI': calc'd for C 2 4
H
3 4
N
7 0 4 : 484.3 (M+H*); Found: 484.1 (M+H'), 242.7 ((M+2H)/2), Example 43: Prepared using Method XIV:
NH
2 K N N N N N T 15 A 1H NMR (CD 3 OD, 300 MHz): 8 7.58-7.43 (m, 4H), 4.99 (s, 0.5H), 4.89 (s, 0.5H), 4.35 (s, 1H), 4.05 (s, 1H), 3.62-3.45 (m, 4H), 3.44 (m, 2H), 3.14 (m, 2H). 3.31 (s, 1H), 3.14 (m, 2H), 2.17 (m, 2H), 2.15-1.80 (m, 6H) - [HCI salt}. LCMS-ESI': calc'd for C 22
H
3
ON
7 0: 408,3 (M+H*); Found: 408.2 (M+H*), 204.7 ((M+2H*)/2). 124 WO 2010/077613 PCT/US2009/067002 Compound AY: Prepared using Method XIII:
NH
2 N Q,, NO 2 N N N O HO N 'H NMR (CD 3 0D, 300 MHz): 5 7.36-7.19 (m, 4H), 4.80-4.70 (m, broad, 2 lines, 2H), 4.17 5 (q, J = 7.0 Hz, 2H), 4.14-3.95 (m, broad, 2 lines, 2H), 3,80-3.60 (m, 2H), 3.62 (s, broad, 2H), 3.44-3.16 (m, 2H), 3.02-2.86 (m, broad, 2 lines, 3H), 2.53 (m, 4H), 1.79 (m, 4H), 1.23 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for C 23
H
3 4
N
7 0 6 S: 536.2 (M+H*); Found: 536.1 (M+H*), 268.5 ((M+2H*)/2). 10 Example 44: Prepared using Method XIV:
NH
2 H N N 0 H N 'H NMR (CD 3 0D, 300 MHz): 5 7.60-7.40 (m, 4H), 4.92 (s, 1H), 4.36 (s, 1H), 4.12 (s, 1H), 3.81 (t, J = 7.3 Hz, 2H), 3.46 (m, 2H), 3.40-3.26 (m, 2H), 3.32 (s, 1H), 3.15 (m, 2H), 2.90 (s, 3H), 2.13 (m, 2H), 1.99 (m, 2H) - [HCI salt]. LCMS-ESF*: caic'd for C 21
H
30
N
7 0 3 S: 460.2 15 (M+H*); Found: 460.2 (M+H*), 230.7 ((M+2H*)/2). Compound AZ: Prepared using Method XiIl:
NH
2 0 N NO2 O N N tN H 0 N 125 WO 2010/077613 PCT/US2009/067002 'H NMR (CD 3 0D, 300 MHz): 5 7.36-7.19 (m, 4H), 4.80-4.68 (m, broad, 2 lines, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4,07 (s, 2H), 4.05 (q, J = 7.0 Hz, 4H), 3.62 (s, 2H), 3.52 (m, 2H), 2.52 (m, 4H), 2.20-1.93 (m, 2H), 1.79 (m, 4H), 1.26 (t, J = 7.0 Hz, 6H), 1.23 (t, J = 7.0 Hz, 3H). LCMS-ES I: calc'd for C 26
H
4 1
N
7 %-P: 594.3 (M+H); Found: 594.2 (M+H*), 297.6 5 ((M+2H*)/2). Example 45: Prepared using Method XIV:
NH
2 H o N N O N N N H 'H NMR (CD 3 0D, 300 MHz): 5 7.60-7.40 (m, 4H), 5.03 (s, 0.5H), 4.93 (s, 0.5H), 4.36 (s, 10 1H), 4.08 (s, 1H), 4.07-3.92 (m, 4H), 3.62-3.50 (m, 2H), 3.45 (m, 2H), 3.32 (s, 1H), 3.16 (m, 2H), 2.30-1.90 (m, 6H), 1.34-1.19 (m, 6H) - {HCI sat). LCMS-ESI*: calc'd for
C
24
H
37
N
7 0 4 P: 518.3 (M+H*); Found: 518.2 (M+H), 259.7 ((M+2H*)/2). Compound BA: Prepared using Method XIII:
NH
2 o N02 "N N" N H 0 No 'H NMR (CD 3 OD, 300 MHz): 5 7.38-7.21 (m, 4H), 4.74 (s, 2H), 4.33 (m, 1H), 4.17 (q, J = 7.0 Hz, 2H), 4.08-3.96 (m, broad, 2 lines, 2H), 3.93-3.80 (m, 2H), 3.80-3.70 (m, 2H), 3.62 (s, 2H), 3.54-3.48 (m, 1H), 2.53 (m, 4H), 2.22-2.06 (m, 1H), 1.79 (m, 4H), 1.24 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for C 24
H
34
N
7 0 5 : 500.3 (M+H*); Found: 500.1 (M+H*), 250.7 20 ((M+2H*)/2). Example 46: Prepared using Method XIV: 126 WO 2010/077613 PCT/US2009/067002
NH
2 H N N N N N H N 'H NMR (CD 3 OD, 300 MHz): S 7.6Q-7.40 (m, 4H), 4.95 (s, 0.5H), 4.37 (s, 1.5H), 4.10 (s, 1.0H), 3.91 (app. q, J = 7.3 Hz, 1H), 3.81-3.73 (m 2H), 3.65 (app. dd, J = 7.3 Hz, 2.2 Hz, 1 H), 3.46 (m, 2H), 3.33 (s, 1 H), 3.20-3.08 (m, 3H), 2.25-1.85 (m, 6H) - [HCI salt]. LCMS 5 ES1*: calc'd for C 22
H
30
N
7 0 2 : 424.2 (M+H*); Found: 424.2 (M+H*), 212.7 ((M+2H*)/2). Scheme 21
NH
2 Cl 1) NH 3 /MeOH, THF, 0 0 C N NO2
NO
2 I N N S N Cl 2) HN O N 00 BB BC 80 CN CN 10 Method XVIl: Dissolved BB (2.4 g, 10 mmol) in anhydrous THF (40 mL) and stirred under N 2 (g) in an ice bath. Added 7N NH 3 in MeOH solution (1.6 mL, 11 mmol) dropwise over 5-10 minutes. Reaction was stirred for 60 minutes. Dissolved BC (2.2 g, 10 mmol) in anhydrous THF (4mL) and added to the reaction in portions over 5-10 minutes. Added DIPEA (1.7 mL, 10 mmol) in portions over 5-10 minutes. Reaction mixture was then 15 stirred for 16 hours at room temperature. Diluted reaction with EtOAc and washed with saturated NaHC0 3 (aq) solution (2X) followed with saturated NaCI(aq). Dried organic extract over anhydrous Na 2
SO
4 and concentrated under reduced pressure. Re-dissolved resultant in small amount of EtOAc and added hexanes to give solid, which was collected and dried under high vacuum to give BD (3.7g, 9.2 mmol). 'H-NMR: 300 MHz. (DMSO-d 6 ) 20 d: 8.05 (s, broad, 2H), 7.78-7.52 (m, 4H), 4.73 (s, 2H), 4.17-4.08 (m, 4H), 2.28 (s, 3H), 1.17 (t, J = 6.9Hz, 3H). LCMS-ESI*: calc'd for C 1
H
18 N0 4 S: 403.1 (M+H*); Found: 403.0 (M+H*). Scheme 22 127 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2 N 1) peracetic acid N N 2 N N ONO OI 2) nBuOH, TFA BU BE CN CN Method XVIII: Dissolved BD (1 g, 2.5 mmol) in anhydrous acetonitrile (25 mL) and stirred under N 2 (g) in an ice bath. Added 32% peracetic acid solution (2.1 mL, 10 mmol) dropwise over 1Q minutes. Stirred for 2 hours. Added saturated Na 2
S
2 0 3 (aq) solution and 5 stirred for 5-10minutes. Extracted with EtOAc. Organic extract was then washed with saturated NaCl(aq), dried over anhydrous Na 2
SO
4 and concentrated under reduced pressure. Mixed the resultant with nBuOH (15 mL) and TFA (963 pL, 12.5 mmol) and then stirred at 100 C for 2-3 hours. Concentrated under reduced pressure. Dissolved in EtOAc and washed with saturated NaHCO 3 (aq) solution (2X) followed with saturated 10 NaCl(aq). Dried organic extract over anhydrous Na 2
SO
4 and concentrated under reduced pressure. Purified with Combiflash silica gel column (0-40% EtOAc in hexanes) to give BE (830mg, 1.95 mmol). 'H-NMR: 300 MHz, (CDC 3 ) d: 7.68-7.47 (m, 4H), 4.78 (s, 2H), 4.25-4.17 (m, 4H), 4.02 (s, 2H), 1.69 (m, 2H), 1.44 (m, 2H), 1.29 (t, J = 6.9Hz, 3H), 0.94 (t, J = 7.5Hz, 3H). LCMS-ESl*: calc'd for C 20
H
2 4 Ns0 5 : 429.2 (M+H*); Found: 429.0 15 (M+H*). 128 WO 2010/077613 PCT/US2009/067002 Scheme 23
NH
2
NH
2 Pd/C, H 2 -- O N 0 EtOH /ACN CN BE BF CN Method XIX: Dissolved BE (650 mg, 4.54 mmol) in EtOH and acetonitrile. Added 10% Pd/C and stirred under atmosphere H 2 (g) for 18 hours. Added 0.5M HCI(aq) (5 mL) and 5 filtered through Celite. Concentrated under reduced pressure to give BF (585 mg, 1.5 mmol). Purified with prep HPLC. 'H-NMR: 300 MHz, (DMSO-d 6 ) d: 9.70 (s, 1H), 7.78-7.54 (m, 4H), 6.23 (s, 2H), 4.68 (s, 2H), 4.04 (t, J = 6.6Hz, 2H), 3.89 (s, 2H), 1.54 (m, 2H), 1.31 (m, 2H), 0.85 (t, J = 7.5Hz, 3H). LCMS-ESI*: calc'd for C 18
H
2 0
N
6 0 2 : 353.2 (M+H*); Found: 353.1 (M+H*). 10 Scheme 24
NHI
2
NH
2 H N 0 formic acid N N 0 OIN N Raney-Ni O N N' FCN BG BF I Method XX: Dissolved BF (176 mg, 0.5 mmol) in formic acid (2 mL). Added Raney-Ni and stirred at 80*C for 90 minutes. Filtered through Celite and washed with formic acid. 15 Diluted filtrate with EtOAc and washed with water (2X), saturated NaHCO 3 (aq) solution (2X) followed with saturated NaCl(aq). Dried organic extract over anhydrous Na 2 SO, and concentrated under reduced pressure. Purified with Combiflash silica gel column (0-10% MeOH in DCM) to give BG (40mg, 0.11 mmol). 'H-NMR: 300 MHz, (DMSO-d 6 ) d: 9.99 (s, I H), 9.71 (s, I H), 7.84-7.57 (m, 4H), 6.23 (s, 2H), 4.74 (s, 2H), 4.07 (t, J = 6.6Hz, 2H), 20 3.87 (s, 2H), 1.56 (m, 2H), 1.32 (m, 2H), 0.85 (t, J = 7.5Hz, 3H). LCMS-ESI*: calc'd for
C
1
H
21
N
5 0 3 : 356.2 (M+H*); Found: 356.0 (M+H*). 129 WO 2010/077613 PCT/US2009/067002 Scheme 25 Example 47
NH
2 H
NH
2 H N " N 0 NaBH(OAc) 3 N N CO HOAc 0 N N CHON N BG C NH Example 47 0 Method XXI: Mixed BG (20 mg, 0.056 mmol) with anhydrous acetonitrile (500 pL). Added 5 morpholine (15 pL, 0.169 mmol) and HOAc (10 pL, 0.169 mmol) and stirred for 15 minutes. Added NaBH(OAc) 3 (36mg, 0.169 mmol) and stirred for 3 hours. Added more morpholine (15 pL, 0.169 mmol) and NaBH(OAc) 3 (36mg, 0.169 mmol) and stirred for 16 hours. Added MeOH and stirred for 5-10 minutes. Diluted with EtOAc and washed with saturated NaHCO 3 (aq) solution (2X) followed with saturated NaCI(aq). Dried organic 10 extract over anhydrous Na 2
SO
4 and concentrated under reduced pressure. Purified with Prep HPLC to give Example 47 (15mg, 0.035 mmol). 'H-NMR: 300 MHz, (Methanol-d 4 ) d: 7.72 (s, 1 H), 7.51 (m, 3H), 4.96 (s, 2H), 4.46 (t, J = 6.6Hz, 2H), 4.38 (s, 2H), 4.16 (s, 2H), 4.05-3.82 (m, 4H), 3.35-3.15 (m, 4H), 1.74 (m, 2H), 1.45 (m, 2H), 0.94 (t, J = 7.2Hz, 3H). LCMS-ESI*: calc'd for C 22
H
30
N
6 0 3 : 427.2 (M+H*); Found: 427.1 (M+H'). 15 Scheme 26 Example 48
NH
2 NH 2 H NN N O NaBH(OAc) 3 N NO ) N N OcN N L, CHO BG CH Example 48 Mixed BG (20 mg, 0.056 mmo) with anhydrous acetonitrile (5 mL). Added piperidine (55 20 pL, 0.56 mmol) and HOAc (16 pL, 0.28 mmol) and stirred for 15 minutes. Added NaBH(OAc)3 (59 mg, 0.28 mmol) and stirred for 3 hours. Added more piperidine (55 pL, 0.56 mmol) and NaBH(OAc) 3 ((59 mg, 0.28 mmol) and stirred for 48 hours. Added MeOH and 0.5M HCI(aq). Concentrated under reduced pressure. Purified with Prep HPLC to give Example 48 (13.8mg, 0.033 mmol). 1 H-NMR: 300 MHz, (Methanol-d 4 ) d: 7.51-7.45 25 (m, 4H), 4.82 (s, 2H), 4.24 (s, 2H), 4.18 (t, J = 6.3Hz, 2H), 3.95 (s, 2H), 3.14 (s, broad, 130 WO 2010/077613 PCT/US2009/067002 4H), 1.82-1.67 (m, 8H), 1.44 (m, 2H), 0.93 (t, J = 7.2Hz, 3H). LCMS-ESI*: calc'd for
C
2 3
H
32
N
8 0 2 : 425.3 (M+H*); Found: 425.2 (M+H*). Compound BH: Prepared using Method X:
NH
2 N NO 2 O N N CO 2 Et 5 N Ethyl-Na-[4-amino-2-n-butoxy-5-nitropyrimidin-6-yl],Nr-[3'-(pyrrolidin-1"-ylmethyl) benzyl]-glycinate: 'H NMR (CD 3 0D, 300 MHz): d 7.24-7.31 (m, 4H), 4.77 (s, 2H), 4.14 4.23 (m, 6H), 3.62 (m, 2H), 2.51 (m, 4H), 1.79 (m, 4H), 1.66 (m, 2H), 1.40 (m, 2H), 1.26 (t, J = 7 Hz, 3H), 0.94 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 2 4
H
35
N
6 0 5 : 487.6 (M+H'); 10 Found: 487.2 (M+H*). Example 49: Prepared using Method XII:
NH
2 O0 N N N 4-amino-2-n-butoxy-8-[3'-(pyrrolidin-1"-ylmethyl)-benzyl]-5,6,7,8-tetrahydropteridin 15 6-one: 1 H NMR (CD 3 0D, 300 MHz): d 7.65 (s, 1H), 7.50 (m, 3H), 4.96 (s, 2H), 4.44 (t, J = 7 Hz, 2H), 4.40 (s, 2H), 4.16 (s, 2H), 3.48 (m, 2H), 3.19 (m, 2H), 2.02-2.17 (m, 4H), 1.74 (m, 2H), 1.45 (m, 2H), 0.94 (t, J = 7 Hz, 3H) - [HCi salt]. LCMS-ESI*: calc'd for
C
2 2
H
31
N
8 0 2 : 411.5 (M+H*); Found: 411.3 (M+H*). 20 Scheme 27 N01. NaOBu, BuOK 0 N 2. distill off some solvent N N
NH
2 + AO 3. add hexane BI Method XXII: Cyanoacetylcyanamide, Monosodium Salt (Compound BI). In a 3.0 L round bottom 1 neck flask, a solution of cyanamide (50.0g, 1.19 mol), ethyl cyanoacetate 25 (126.4 mL, 1.19 mol), and anhydrous n-BuOH (1.00 L mL) was treated with 20% w/w 131 WO 2010/077613 PCT/US2009/067002 NaOBu/BuOH (571 mL, 1.19 mmol) at 23 0C. The reaction was stirred vigorously and became cloudy and thick. After 12-16 hrs, the reaction was fitted with a Distillation head. The sidearm of the distillation head was fitted with a long reflux condenser (water circulated). At the end of the condenser, a Claisen Vacuum adaptor was attached and led 5 into a receiver flask (2.0 L rb., cooled in an ice bath). All ground glass joints were greased and clamped. A vacuum of 10 mmHg or less was applied to the system at 23 *C (some mild bumping occurred. A dry-ice/acetone trap was employed in a dewar finger trap to catch uncondensed vapors) Once bumping had become minimal, the reaction was heated externally to 45-60 "C (oil or water bath), and solvent (1.1 L) was distilled off. 10 Vacuum was released, and while the system was still warm, hexanes (2.0 L) were added. System was allowed to cool to 23 *C, and a precipitate was observed. The slurry was filtered over coarse glass frits to capture the solid. The filter cake was washed with hexanes while suction was off (2 x 250 mL; each time stir the cake/hexanes, then turn suction back on). The cake was then dried in a vacuum oven at 40-45 *C overnight, 15 affording cyanoacetylcyanamide, monosodium salt (128.14 g, 82% yield) as a free flowing, slightly hygroscopic powder. The powder was immediately placed in a glass jar and stored in a dessicator. Scheme 28 N O N BuOH, HCI (gas) Ci NH 2 0 N 23 *C, 2 hrs only 0 N 20 Na H BJ Method XXIII: N-Cyanoacetyl-butylisouronium chloride (Compound BJ). A suspension of cyanoacetylcyanamide, monosodium salt BI (20.0 g, 153 mmol) in n-BuOH (300 mL) was treated with HCI (4.0 M in dioxane, 100 mL, 400 mmol). During addition the 25 suspension became more colloidal and there was a mild exotherm to an internal temperature of 35 *C, then the reaction transitioned to a thicker consistency. After 2 h, 10% w/v aq. NaHCO 3 (200 mL) was added cautiously (effervescence) until the pH of the aq. phase reached 7.5. The organic layer was collected, dried (Na 2
SO
4 ), and filtered over glass frits, then transferred into a 500 mL round bottom flask. Distillation of 330 mL of 30 solvent away from the dried organic phase was achieved using the procedure above (step 1, pressure -10 mmHg, 60 'C bath temp). The thick syrupy residue contains crude N-cyanoacetyl-butylisouronium chloride, BJ, which is unstable and immediately used in the next reaction. 132 WO 2010/077613 PCT/US2009/067002 Scheme 29 cl Na 2 00 3 H e NH 2 0 ~-N _O N H 2 ,90 C, 16 h O N OH BJ HB BK 5 Method XXIV: 4-Amino-2-butoxy-6-hydroxypyrimidine (Compound BK). An emulsion of all of the crude N-cyanoacetyl-butylisouronium chloride BJ (33.35 g, 153 mmol) in a mixture of dioxane and n-BuOH (-70 mL) was treated with 10% w/v aq. Na 2
CO
3 (200 mL) and was stirred vigorously at 90 *C for 16 h. The reaction was then allowed to cool to 23 *C over the next hour. A white semicrystalline precipitate formed. Then the system was 10 cooled to 0 oC for 3 h, and the white-brown precipitate was collected on coarse glass frits. The filter cake was washed with hexane (2 x 50 mL) and dried in a vacuum oven at 40 *C, giving desired product BK (14.1 g, 50% yield over 2 steps). The neutralized aqueous phase was then extracted with CH2CI 2 (3 x 50 mL). The extracts were combined, dried (MgSO 4 ), filtered, and concentrated to a brown oil, After standing at 23 *C overnight, the 15 oil solidified. The gooey solid was triturated with hexane (50 mL) and filtered. The collected solid proved to be additional pure product (1.17 g, 4% yield). 1 H NMR (DMSO d 6 , 400 MHz): 5 (ppm) 11.16 (s, broad, 1H), 6.29 (s, broad, 2H), 4.73 (s, 1H), 4.23 (t, J = 7 Hz, 2H), 1.70-1.60 (m, 2H), 1.43-1.33 (m, 2H), 0.92 (t, J = 7 Hz, 3H). 20 Scheme 30
NH
2 NH 2 N . Fuming HNO 3 N 0 NO 2 OH 0OC O N OH .HNO 3 BK BL Method XXV: 4-Amino-2-butoxy-5-nitro-6-hydroxypyrimidine, BL (nitrate salt and free base). A 50 mL flask containing fuming aqueous HNO 3 (18 mL) at 0 *C was treated 25 with 4-amino-2-butoxy-6-hydroxypyrimidine BK (8.00 g) via solid addition funnel under N 2 . The pyrimidine was added at a rate of ca. 266 mg every minute over a 30 min period. Reaction went from yellow to deep red. Once the addition was complete, the reaction was stirred at 0 *C for another 2 h. Then the reaction was added slowly to a mixture of CH 2
C
2 and H 2 0 (100 mL each) at 0 *C. After addition was complete, the diluted reaction was 133 WO 2010/077613 PCT/US20091067002 allowed to stir for 30 min. A pink precipitate formed and was collected via vacuum filtration. LCMS analysis and 'H NMR in DMSO (identical to values below) reveal that the compound is the mononitrate salt of the product (6.63 g, 52% yield). The organic layer was collected. The aq. layer was extracted exhaustively with CH 2 1C 2 (100 mL portions) 5 until the aqueous layer showed no traces of product. All organic phases were combined, dried (MgSO4), filtered, and concentrated. The residue was purified on silica get by flashing (Eluent: CH 2
C
2 :MeOH 100/0 to 80/20, linear gradient) giving the desired product BL as a free base (2.02 g, 20% yield)(yellow powder). 'H NMR (free base or nitrate salt, DMSO-d, 400 MHz): 5 (ppm) 12.07 (s, broad, 1H), 8.83 (s, broad, 1H), 8.77 (s, broad, 10 1H), 4.36 (t, J = 7 Hz, 2H), 1.73-1.63 (m, 2H), 1.44-1.34 (m, 2H), 0.94 (t, J = 7 Hz, 3H). Scheme 31
NH
2 TsCl
NH
2 N "1N NO 2 2,4,6-collidine N NO 2 O N OH *HNO 3
CH
3 CN, 60 "C O N OTs BL BM Method XXVI: 4-Amino-2-butoxy-5-nitro-6-(para-toluenesulfonyloxy)pyrimidine 15 (BM). A solution of 4-amino-2-butoxy-5-nitro-6-hydroxypyrimidine BL (nitrate salt form, 8.00 g, 27.5 mmol, 1.00 equiv, see note below) in acetonitrile (80.0 ml) was treated with 2,4,6-collidine (distilled under vacuum from NaH, 10.90 ml, 82.4 mmol, 3.00 equiv), followed by TsCl (26.21 g, 0.138 mol, 5.00 equiv). The reaction was stirred for 4 h at 60 0C. By this point, 95% conversion to the product was observed using LC-MS as the 20 analytical method (Water/Acetonitrile (with trace AcOH) 95:5-2:98 on a C-18 gemini column). The reaction was added dropwise to a 0 0C mixture of H 2 0 (400 mL) and CH 2 Cl2 (200 mL). After 10 min, the mixture was extracted (3 x 200 mL CH2C). All organic layers were combined, dried (Na 2
SO
4 ), filtered, and concentrated to a total volume of 50 mL. The crude solution of product was purified by directly loading onto a 330 g column of 25 silica gel, followed by chromatography (Eluent hexane/EtOAc 9:1 -) 0:100) giving semipure BM contaminated with 2,4,6-Collidine. The oily solid was taken up in hexane (50 mL) and agitated, then filtered over glass frits. The filter cake was washed with several 30 mL portions of hexane until no collidine was present, giving pure product BM (5.44 g, 52% yield). 'H NMR in CDCi was obtained, along with LCMS analysis. 'H NMR 30 (CDC1 3 , 400 MHz): 6 (ppm) 7.99 (d, J = 8.2 Hz, 2H), 7.95 (s, broad, 1H), 7.39 (d, J = 8.2 134 WO 2010/077613 PCT/US2009/067002 Hz, 2H), 6.19 (s, broad, 1H), 4.26 (t, J = 7.4 Hz, 2H), 2.48 (s, 3H), 1.73 (app. quintet, J = 7.4 Hz, 2H), 1.43 (app. sextet, J = 7.4 Hz, 2H), 0.96 (t, J = 7.4 Hz, 3H). Scheme 32
NH
2
NNH
2 NH2 NO 2 N NO 2
H
2 0 2 , HOAc iN
CH
3 S N NCCO 2 Et Na 2 WO4*2H 2 O MeO 2 S N NCCO 2 Et -N N D5N BN N Method XXVII: Ethyl-Na-14-amino-2-methafnesulfonyl-5-nitropyri midin-6-y],N,-[3' (pyrrolidin-1"-ylmethyl)-benzyl]-glycinate (BN). To a suspension of the sulfide D (100 mg, 0.217 mmol) in EtOH (2.0 mL) was added glacial AcOH (124 pL, 2.17 mmol) and 10 sodium tungstate dihydrate (21.5 mg, 65.1 pmot). The reaction was cooled to 0 *C, and 30% aq. hydrogen peroxide (245 pL, 2.17 mmol) was added dropwise over a 2 min period. After 9 h, the reaction was added to a 0 *C solution of 10% w/v aq. Na 2
S
2 0 3 (6 mL). After 5 min, the reaction was extracted with CH 2
CI
2 (7 x 10 mL). The combined organic layers were dried over Na 2
SO
4 , filtered, and concentrated under vacuum to a 15 yellow powder, containing the sulfone BN and the corresponding sulfoxide as a 1:1 mixture (45.5mg, 43% yield based on mass of sulfone). In all subsequent chemistry, both the sulfoxide and sulfone react similarly. 'H NMR (sulfone, CDC 3 , 300 MHz): 6 (ppm) 7.50-7.24 (m, 4H), 4.79 (s, 2H), 4.21 (q, J = 7.0 Hz, 2H), 4.16 (s, 2H), 3.97 (s, 2H), 3.17 (s, 3H), 3.01-2.85 (m, 4H), 2.02-1.91 (m, 4H), 1.28 (t, J = 7.0 Hz, 3H). LCMS-ESl*: calc'd 20 for C 21
H
29
N
6 0 6 S (sulfone): 493.2 (M+H*); Found: 493.1 (M+H*). Scheme 33 H3NO Cl E O NaBH(OAc) 3 0 1,2-dichloroethane ~ N AcOH, 23*C B0 25 Method XXVIII: Ethyl-Nr-[3-(pyrroIdin-I'-ylmethyI)-benzyl]-B-aIaninoate (BO). To a suspension of ethyl QG-alaninoate hydrochloride (890 mg, 6.39 mmol, 1.1 equiv), 3 (pyrrolidin-1'-ylmethyl)-benzaldehyde (1.10 g, 5.81 mmol, 1.0 equiv), NaBH(OAc) 3 (2.46 135 WO 2010/077613 PCT/US2009/067002 g, 11.6 mmol, (2.0 equiv), and 1,2-dichloroethane (7.0 mL) was added glacial AcOH (830 pL, 5.81 mmol, 1.0 equiv) at 23 *C. To aide fluidity, more 1,2-dichloroethane (500 pL) was added. After 75 min, the reaction was carefully quench with 0.1 M aq HC, adjusting the pH to -3. Then saturated aq Na 2
CO
3 was added until the pH was -8. The reaction was 5 extracted with CH 2 0 2 (3 x 150 mL). All organic layers were combined, dried (Na 2 SO4), filtered, and concentrated to a pale yellow oil BO (740 mg, 44% yield), 'H NMR (CDC 3 , 300 MHz): 8 (ppm) 7.30-7.21 (m, 4H), 4.16 (q, J = 7.0 Hz, 2H), 3.80 (s, 2H), 3.64 (s, 2H), 2.99 (s, broad, 1H), 2.91 (t, J = 6.4 Hz, 2H), 2.58-2.48 (m, 4H), 2.53 (t, J = 6.4 Hz, 2H), 1.85-1.76 (m, 4H), 1.26 (t, J 7.0 Hz, 3H). LOMS-ESl*: calc'd for C 17
H
2 7
N
2 0 2 : 291.2 10 (M+H*); Found: 291.1 (M+H*). Scheme 34 Cl NH2
TN
2
NH
3 , Et 3 N "CN 2 ' N A Ci THF, -78 -4 00 LS N 2 15 Method XXIX: 4-Amino-6-chloro-2-methylthio-5-nitropyrimidine (B). A solution of 4,6-dichloro-2-(methylthio)-5-nitropyrimidine (3.53 g, 14.7 mmol) in THF (15 mL) at - 78 *C was added Et 3 N (3.75 mL, 27.0 mmol), followed by NH 3 (7 N in MeOH, 1.80 mL, 12.86 mmol). The reaction was then warmed to 0 *C and stirred for 1 h. The crude solution of product B was immediately used in the next reaction (Scheme 35). 20 Scheme 35
NH
2
NH
2 S N C02 -7- 3 ~ N&NO2o B EtsN N, N OJ o + THF S N N O NN -78 23 *C p N H I BO Method XXX: Compound BP. A solution of 4-amino-6-chloro-2-(methylthio)-5 nitropyrimidine (from the previous reaction above) at - 78 *C was added Et 3 N (3.75 mL, 25 27.0 mmol) and ethyl-N-[3-(pyrroidin-1'-ylmethyl)-benzyl)-R-alaninoate (3.56 g, 12.3 136 WO 2010/077613 PCT/US2009/067002 mmol). The reaction was allowed to warm to 23 *C ovemight. The reaction was quenched with aq. saturated NH 4 CI (excess) and extracted with EtOAc (2x). All organic layers were combined, dried (Na 2 SO4), filtered, and concentrated. The residue was purified on silica gel using 20% MeOH/CH 2
C
2 (isocratic) as the eluent, giving product BP (6.5 g, yield not 5 determined because some solvent was present). 'H NMR (CDCi 3 , 300 MHz): 5 (ppm) 7.26-7.16 (m, 4H), 4.55 (s, 2H), 4.11 (q, J = 7.0 Hz, 2H), 3.74 (t, J = 7.0 Hz, 2H), 3.61 (s, 2H), 3.48 (s, 2H), 2.64 (t, J = 7.0 Hz, 2H), 2.54-2.45 (m, 4H). 2.43 (s, 3H), 1.83-1.74 (m, 4H), 1.22 (t, J = 7.0 Hz, 3H). LCMS-ESl*: calc'd for C 22
H
3 1 N70 4 S: 475.2 (M+H'); Found: 475.0 (M+H*). 10 Scheme 36 N NON 2 0 2 0 2 , HOAc N NH N0 2 0 S N O NNa 2
W
4 . 2H 2 0 0 N NO BP EtOH, 00 0CBQ B a--No BO Method XXXI: Compound BP. A solution of the sulfide BP (869 mg, 1.83 mmol), in 15 absolute EtOH (20 mL) at G *C was added sodium tungstate dihydrate (180 mg, 0.550 mmol), followed by glacial AcOH (590 pL, 18.3 rnmol). Finally, 30% w/v aq. H 2 0 2 (2.77 mL, 18.3 mmol) was added dropwise. Once the reaction was complete, it was added dropwise to a mixture of 10% w/v aq. Na 2
S
2 0 3 (excess relative to H 2 0 2 ) and CH 2 Cl 2 The mixture was then extracted repeatedly with CH 2
CI
2 . All organic extracts were combined, 20 dried (Na 2
SO
4 ), filtered, and concentrated to yellow solid (3.0 g, yield not found because some glacial AcOH and CH 2 Cl 2 are still present). The crude solid BQ was used in the next reaction without further purification. LCMS-ESI*: calc'd for C2H 3 1
N
6
O
6 S: 507.2 (M+H*); Found: 507.1 (M+H*). 25 Scheme 37
NH
2 NH2 N NO2 0 TFA N NO 2 0 S'i N N O BuOHO N N O N -100*C BR N Q 13 137 WO 2010/077613 PCT/US2009/067002 Method XXXII: Compound BR. A solution of the sulfone BQ (crude from above, 927 mg net mass) in n-butanol (15 mL) was treated with TFA (420 pL) and stirred at 95 *C. More TFA (280 pL) was added after 2.5 h, and the reaction was heated to 100 *C. 3 hours later, the reaction was quenched with saturated aq. NaHCO 3 . The mixture was 5 extracted with CH 2
C
2 (8x), and all organic layers were combined, dried (Na 2 SO4), filtered, and concentrated. The residue was purified on silica gel using 20% MeOH in CH 2 C1 2 (isocratic) as the eluent. Product-containing fractions, which were semipure, were combined and purified on a C-18 reversed-phase column (first eluent: H 2 0/CH 3 CN 100:0 -+ 0:100; second eluent CH 3 CN/MeOH 100:0 -+ 0:100) giving pure product BR (59 mg, 10 yield not determined). 1 H NMR (CDCl 3 , 300 MHz): 8 (ppm) 7.26-7.06 (m, 4H), 4.53 (s. 2H), 4.24 (t, J = 6.7 Hz, 2H), 4.11 (q, J = 7.0 Hz, 2H), 3.71 (t, J = 7.0 Hz, 2H), 3.58 (s. 2H), 3.48 (s, 2H), 2.64 (t, J = 6.7 Hz, 2H), 2.52-2.43 (m, 4H), 1.81-1.74 (m, 4H), 1.74-1.56 (m, 2H), 1.50-1.33 (m, 2H), 1.22 (t, J = 7.0, 3H), 0.93 (t, J = 7.3 Hz, 3H). LCMS-ESl*: calc'd for C 2 5
H
3 7
N
6 0 5 : 501.3 (M+H*); Found: 501.1 (M+H 4 ). 15 Scheme 38: Example 50 NH2H 0 NO2 e N O N N N N 0 N N AcOH, 60 *C BR N Example 50 Method XXXIII: Example 50. A suspension of the nitro compound BR (5.0 mg) and zinc powder (6.5 mg) in glacial AcOH (500 pL) was heated to 60 *C. After 1 h, more zinc 20 powder (6.5 mg) was added, and heating was continued. 2 hours later, the reaction was diluted with H 2 0 (500 pL) and directly purified on a 4.3 g C-18 reversed-phase sep-pak column (0.05% w/v aq. HCI/CH 3 CN 100:0 - 0:100) giving Example 50 (3.9 mg, 78% yield) as a di-HCI salt. 1 H NMR (CD 3 OD, 300 MHz): 6 (ppm) 7.57-7.39 (m, 4H), 5.00 (s, 2H), 4.38 (s, 2H), 4.28 (t, J = 6.5 Hz, 2H), 3.86-3.82 (m, 2H), 3.50-3.40 (m, 2H), 3.20-3.09 25 (m, 2H), 2.88-2.78 (m, 2H), 2.24-2.08 (m, 2H), 2.08-1.96 (m, 2H), 1.64 (app. Quintet, J = 6.5 Hz, 2H), 1.34 (app. Septet, J = 7.0 Hz, 2H), 0.87 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for C 2 3
H
3 3
N
6 0 2 : 425.3 (M+H*); Found: 425.3 (M+H*). Scheme 39 138 WO 2010/077613 PCT/US2009/067002 C NNH 2 N NO2 Et 3 N, NH 3 , THF N NO 2 Cl N" Cl -78 -* 0 "C LC N CI Method XXXIV, Part 1: 6-amino-2,4-dichloro-5-nitropyrimidine. A solution of 2,4,6 trichloro-5-nitropyrimidine (94 mg, 0.413 mmol) in THF (5 mL) was cooled to - 78 *C and treated with Et 3 N (110 pL, 0.757 mmol), followed by NH 3 (7 N in MeOH, 50 pL, 0.344 5 mmol). The reaction was warmed to 0 0 C. Once TLC indicated complete consumption of the starting material, the crude product solution was immediately used in the reaction below (Scheme 40). Scheme 40 0 "--O - -N . N H N BO NH 2 + N NO 2 O Et 3 N, NH 3 , THF N N [ NH 2 -7C0C 0 N C-~ NO2 78 - 0 *C B N -- NO 2 BSN
I
10 - Cl N CI Method XXXIV, Part 2: Compound BS. A crude solution of 6-amino-2,4-dichloro-5 nitropyrimidine (from reaction above) was cooled to - 78 *C and Et 3 N (110 pL, 0.757 mmol) was added, followed by a solution of Ethyl-Nar[3-(pyrroIdin-V-ylmethyl)-benzyl]-& alaninoate (100 mg, 0.344 mmol) in THF (1.0 mL). The reaction was warmed to 0 *C. 15 After 80 min, the reaction showed complete conversion to BS. An aliquot was analyzed via LCMS. The remainder of the solution was immediately used in the next reaction below. LCMS-ESF*: calc'd for C 21
H
28
CIN
6 0 4 : 463.2 (M+H*); Found: 463.1 (M+H* for 35 CI) and 465.1 (M+H' for a 7 C1). 20 Scheme 41 139 WO 2010/077613 PCT/US2009/067002
NH
2 j
NH
2 C- N NO O TBuNH 2 C NOO CKI NF N0 0C H!N(' K BS N BT Method -XXXIV, Part 3: Compound BT. A solution of the crude chloropyrimidine BS (from the reaction above) in THF was treated with n-butylamine (170 pL) and heated to 5 80 *C. After 2.5 h, H 2 0 (100 pL) was added to improve fluidity, and heating was continued. The completed reaction was loaded directly onto a C-18 reversed-phase column and chromatographed (eluent: 0.1% w/v aq. TFA/CH 3 CN 100:0 -+ 0:100), giving pure product BT (23.5 mg, 14% yield over 3 steps). 'H NMR (CD30D, 300 MHz): 5 (ppm) 7.32-7.14 (m, 4H), 4.64-4.61 (app. d, broad, J = 5.5 Hz, 2H), 4.07 (q, J = 7.0 Hz, 2H), 10 3.72-3.61 (m, 2H), 3.62 (s, 2H), 3.30 (s, 2H), 2.72-2.60 (m, 2H), 2.58-2.46 (m, 4H), 1.84 1.73 (m, 4H), 1.69-1.24 (m, 4H), 1.20 (t, J = 7.0 Hz, 3H). LCMS-ESI t : calc'd for C2 6
H
38
N
7 0 4 : 500.3 (M+H*); Found: 500.1 (M+H*). Scheme 42 S NH 2 N N o K 2
CO
3 H 0 N Br O N 15 ~ ACN 70 CO) Method XXXV: Compound BU. (2-Morpholinopyridin-4-yl)methylamine (900 mg, 4.657 mmol) was dissolved in acetonitrile and combined with solid potassium carbonate (2.52 g, 18.23 mmol) followed by heating to 70 *C. Ethyl-2-bromoacetate (566 pL, 5.114 mmol) was then added over 10-15 minutes and the mixture was continued to stir at 70 *C for 45 20 min wherein the consumption of SM was observed by HPLC analysis. The mixture was removed from heat source, allowed to cool to RT and was diluted with EtOAc (100 mL) and H 2 0. The reaction was washed with brine (3x) and dried with Na 2
SO
4 , filtered, and concentrated. Desired product BU was obtained in 84.4% yield and used without purification. 25 Scheme 43 140 WO 2010/077613 PCT/US2009/067002
NH
2 rNl N NO 2 H o C N 1. 7N NH 3 in MeOH, O*C NO O (~~ ~ N 0 N0 2
SNN
BU -S. Cl 2. Arnine, THF, rt A N BV 0 Method XXXVI: Compound BV. Dichloropyrimidine A (1.0715 g, 4.502 mmol) was dissolved in 25 mL THF and cooled to 0 *C. NH 3 was added (3.5 Equiv) and the mixture was allowed to stir cold for 1 h. Aminoester (1.22 g, 4.37 mmol) was then added 5 dropwise as a solution in 10 mL THF over 10-15 minutes, and the resulting mixture was allowed to warm to room temperature. After 3 h, the reaction was quenched with the addition of water, diluted with EtOAc and the pH was adjusted to = 8 using solid K 2 COS. The mixture was washed with water, washed with brine then dried with sodium sulfate and concentrated in vacuo. The crude product was then chromatographed on silica with 10 a CH 2
CI
2 and 20% MeOH/CH 2
C
2 gradient over 10-15 column volumes to give BV. Scheme 44 NC) .Boc NCNH Boc20, TEA N N THF BW BX Method XXXVII: Compound BX. Compound BW (500 mg, 3.16 mmol) was added to 15 THF (15 mL). To this was added triethylamine (659 pL, 4.74 mmol). A solution of Boc anhydride (759 mg, 3.48 mmol) in THF was added in portions. The mixture was stirred for 2 hours. After this, the reaction was diluted with EtOAc and washed with saturated NaHCO 3 (aq) (2X) followed with 5% citric acid(aq) and then saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 and concentrated under reduced 20 pressure. The product was purified with silica gel chromatography (0-20% EtOAc in hexanes) to give BX (751 mg, 2.9 mmol). 'H NMR: (CDC1 3 , 300 MHz): 5 7.44-7.25 (m, 3H), 4.60 (s, 2H), 3.67 (t, J = 5.7Hz, 2H), 2.89 (t, J = 6.0Hz, 211), 1.50 (s, 9H). Scheme 45 25 141 WO 2010/077613 PCT/US2009/067002 NC N'Boc Pd/C, H 2 , H 2 N N' Boc MeOH,HOAc BX BY Method XXXVIII: Compound BY. Compound BX (751 mg, 2.9 mmol) was dissolved in MeOH. To this was added HOAc (300 pL) and 10% Pd/C. The mixture was stirred under I atm H 2 for 6 hours. The mixture was filtered through Celite and the filtrate was 5 concentrated under reduced pressure. The residue was dissolved in EtOAc and washed with saturated NaHCO 3 (aq) (2X) followed with saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 and concentrated under reduced pressure to give BY (474 mg, 1.47 mmol). 'H NMR: (CDC13, 300 MHz): 8 7.13 (m, 3H), 4.56 (s, 2H), 3.87 (s, 2H), 3.63 (s, 2H), 2.80 (m, 2H), 1.49 (s, 9H). LCMS-ESI*: calc'd for C 11
H
1 6
N
2 0 2 : 206.1 (M 10 tBu+H*); Found: 206.8 (M-tBu+H*) Scheme 46 Boc K2CO, THF/DCM HBoc
H
2 N N O T H - BY K Br BZ Method XXXIX: Compound BZ. Compound BY (474 mg, 1.47 mmol) was added to 15 anhydrous THF (15 mL). To this was added potassium carbonate and the reaction was stirred under N 2 in an ice bath. A solution of ethyl bromoacetate in anhydrous THF was added dropwise. To this was added anhydrous CH 2
C
2 (5 mL) and the mixture was stirred for 48 hours. The reaction was diluted with EtOAc and washed with saturated NaHCO 3 (aq) (2X) followed with saturated NaCI(aq), The organic extract was dried over 20 anhydrous Na 2
SO
4 and concentrated under reduced pressure. The product was purified with Prep HPLC to give BZ (180 mg, 0.52 mmol), "H NMR: (CDC1 3 , 300 MHz): 6 7.12 (m, 3H), 4.57 (s, 2H), 4.22 (m, 2H), 3.77 (s, 2H), 3.64 (m, 2H), 3.41 (s, 2H), 2.82 (m, 2H), 1.50 (s, 9H), 1.29 (t, J = 7.2Hz, 3H). LCMS-ESl*: calcd for C 1
H
2 8
N
2 0 4 : 349.2 (M+H*); Found: 348.9 (M+H*) 25 Scheme 47: Example 51 142 WO 2010/077613 PCT/US2009/067002
NH
2 NH 2 H NO2 1) Fe, HOAc ON ,N AN B OIN N
K
0 O NN 2) HCI -'0NNH 0 CA Example 51 Method XL: Example 51. Compound CA was dissolved in HOAc (6 mL). To this was added iron powder and the reaction was stirred at 60 *C for 3 hours. The mixture was filtered and washed with HOAc. The mixture was concentrated under reduced pressure. 5 The Boc protected lactam intermediate was purified with silica gel chromatography (0-5% MeOH in CH 2 C1 2 ). The material was then dissolved in MeOH to this was added 4N HCI in dioxane. The mixture was stirred for 30-60 minutes, concentrated under reduced pressure, and then purified with Prep HPLC Phenomenex Gemini 5u C18 column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to give Example 10 51 (109 mg, 0.28 mmol). 'H NMR: (CD 3 0D, 300 MHz): 6 7.30-7.22 (m, 3H), 4.88 (s, 2H), 4.45 (t, J = 6.3Hz, 2H), 4.37 (s, 2H), 4.09 (s, 2H), 3.51 (t, J = 6.3Hz, 2H), 3.12 (m, 2H), 1.76 (m, 2H), 1.47 (m, 2H), 0.96 (t, J = 7.5Hz, 3H). LCMS-ESI*: calc'd for C20H 2 7 NO,: 383.2 (M+H*); Found: 383.0 (M+H*). 15 Scheme 48: Example 52
NH
2 H NH 2 N ~ NpO lodoethane N NpO O N N DMF, DIPEA O N N N NH N Example 51 Example 52 Method XLI: Example 52. Example 51 (20 mg, 0.0417 mmol) was dissolved in anhydrous DMF (1 mL). To this was added lodoethane 3.7 pL, 0.0459 mmol) and DIPEA (16 pL, 0.0917 mmol). The mixture was stirred for 14 hours. The product was purified 20 with Prep HPLC Phenomenex Gemini 5u C 1 8 column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to give Example 52 (6.4 mg, 0.0156 mmol). 'H NMR: (CD 3 OD, 300 MHz): 8 7.32-7.25 (m, 3H), 4.65 (m, 1H), 4.46 (t, J = 6.9Hz, 2H), 4.35 (m, 1H), 4.10 (s, 2H), 3.80 (m, 1H), 3.39-3.19 (m, 8H), 1.75 (m, 2H), 1.46 (m, 5H), 0.97 (t, J = 7.5Hz, 3H). LCMS-ESF*: calc'd for C 22
H
31
N
6 0 2 : 411.2 (M+H); Found: 411.1 (M+H-). 25 Scheme 49 143 WO 2010/077613 PCT/US2009/067002
NH
2 CI N NO 2 N NO 2 1) NH 3 /EtOH, Cs 2
CO
3 ,THF, 0 OC I N Cl N N O CI N~ ~ 2) xa C N Cl NC OEt O CB Method XLII: Compound CB. To a solution of 2,4,6-trichloro-5-nitropyrimidine (200 mg, 0.88 mmol) in THF (3 ml) at 0 0C was added Cs 2
CO
3 (286 mg, 0.88 mmol) and NH 3 in EtOH (2 M, 540 pL, 1.08 mmol) dropwise. The reaction mixture was stirred for 30 min. 5 After 2,4,6-trichloro-5-nitropyrimidine was consumed, a solution of 3-((2-ethoxy-2 oxoethylamino)methyl)benzonitrile (190 mg, 0.88 mmol) in THF (2ml) was added to the reaction mixture at 0 00. Then the reaction mixture was allowed to rise to room temperature and stirred for 2 h. The reaction mixture was washed with saturated NaHCO 3 (aq) and extracted with CH 2 0 2 (x3). The organic phase was combined, dried 10 over Na 2
SO
4 , filtered and concentrated. The residue was purified by silica gel column (0 50% EtOAc in hexanes) to give CB. 1 H NMR: (CDC1 3 , 300 MHz): 6 7.65-7.43 (m, 4H), 4.75 (s, 2H), 4.23-4.19 (m, 2H), 4.03 (s, 2H), 1.28 (t, J = 6.9 Hz, 3H). LCMS-ESI*: calc'd for C 18
H
1 6
CIN
6 0 4 : 391.8 (M+H*); Found: 391.0 (M+H*). 15 Scheme 50
NH
2 NH 2 N N 2 NO, CI1NO 0 Pd(PPh 3
)
4 , K 2
CO
3 , Toluene N N C NON N CB CN CC CN Method XLIII: Compound CC. To a solution of CB in toluene was added pent-1 enylboronic acid (420 mg, 3.04 mmol), K 2
CO
3 (350 mg, 3.07 mmol) and tetrakis(triphenylphosphine)palladium (353 mg, 0.30 mmol). The reaction mixture was 20 reacted at 100 0C for 4 h. The reaction was cooled down, washed with saturated NaHCO 3 (aq) and extracted with CH 2 0 2 (x3). The organic phase was combined, dried over Na 2
SO
4 and filtered. The filtrate was concentrated down and purified by silica gel column (0-50% EtOAc in hexanes) to give CC. 'H NMR: (CDC13, 300 MHz): 6 7.70-7.44 (m, 4H), 7.14-6.99 (m, 1H), 6.18 (d, J = 15.3 Hz, 1H), 4.78 (s 2H), 4.27-4.19 (m, 2H), 4.05 (s, 2H), 144 WO 2010/077613 PCT/US2009/067002 2.28-2.15 (m, 2H), 1.59-1.14 (m, 2H), 1.28 (1, J = 7.5 Hz, 3H), 0.98-0.91 (m, 31-). LCMS ESI*: calc'd for C 21
H
26
N
6 0 4 : 425.5 (M+H*); Found: 425.1 (M+H*). Scheme 51
NH
2
NH
2 N ~-NO 2 N ~-NO 2 ~'N NO O Pd/C, EtOH, H 2 NO N N N ' N ,,, O CC CD 5 CN CN Method XLIV: Compound CD. To a solution of CC (200 mg, 0.47 mmol) in EtOH (5ml) was added Pd/C (1 00mg). The reaction vessel was flushed with H 2 and then stirred under an H 2 atmosphere for 20 min. Then more Pd/C (30 mg) was added and stirred for another 10 min. The reaction mixture was filtered over Celite and was concentrated to 10 give CD, which was used without purification. LCMS-ESI*: calc'd for C 21
H
27
N
6 0 4 : 427.5 (M+H*); Found: 427.2 (M+H*). Scheme 52
NH
2 NH 2 N NO 2 N- NO NO O Zn, Glacial HOAc, 601C NN N N N o CCE CN CN 15 Method XLV: Compound CE. To a solution of CD (120 mg, 0.28 mmol) in glacial acetic acid (3ml) was added zinc powder (370 mg, 5.7 mmol). The reaction mixture was stirred at 60 *C for 3 h. The solvent was removed to dryness under reduced pressure. The residue was washed with saturated NaHCO 3 (aq) solution and extracted with CH 2
C
2 (x3). The organic phase was combined, dried over Na 2
SO
4 and filtered. The filtrate was 20 concentrated down and purified by silca gel column (0-100% EtOAc in hexanes) to give CE. 'H NMR (CD 3 0D, 300 MHz): 6 7.80-7.52 (m, 4H), 4.79 (s, 2H), 3.98 (s, 2H), 3.35 (s, 2H), 1.69-1.29 (m, 6H), 0.90-0.86 (m, 3H). LCMS-ESI*: calc'd for C 19
H
23
N
6 0: 351.4 (M+H*); Found: 351.2 (M+H*). 145 WO 2010/077613 PCT/US2009/067002 Scheme 53: Example 53
NH
2 H
NH
2 N O N N O N N 1) DIBAL-H, CH 2
C
2 N N "'N N 2) pyrrolidine, NaBH(OAc) 3 ,
CH
2
CI
2 CE Example 53 N Method XLVI: Example 53. To a solution of CE (50 mg, 0.14 mmol) in CH 2
C
2 (2ml) at 0 *C was added DIBAL-H (IM in toluene, 710 pL, 0.71 mmol) dropwise. The reaction 5 mixture was stirred at 0 'C for 15 min. The reaction was quenched by water. The mixture was extracted with CH 2 C1 2 (x3). The organic phase was combined, dried over Na 2
SO
4 and filtered. The filtrate was concentrated down. The residue was dissolved in
CH
2
CI
2 /MeOH(1:1, 2ml) and to this was added pyrrolidine (60 pL, 0.72 mmol), sodium triacetoxyborohydride (75 mg, 0.35 mmol) at 0 "C. The reaction mixture was stirred at 10 room temperature for 1 h. The reaction was quenched by adding drops of 1 N HCI, filtered and purified by reverse phase HPLC (5-100% Acetonitrile in H20) to give Example 53. 'H-NMR (300 MHz, methanol-d4): 6 7.49-7.47 (m, 4H), 4.82 (s, 2H), 4.99 (s, 2H), 4.38 (s, 2H), 4.14 (s, 2H), 3.47-3.42 (m, 2H), 3.22-3.18 (m, 2H), 2.72 (t, J = 7.2 Hz, 2H), 2.20-2.16 (m, 2H), 2.03-2.00 (m, 2H), 1.36-1.34 (m, 4H), 0.90 (t, J = 6.6 Hz, 3H). LCMS-ESF~: calc'd 15 for C 23
H
33
N
6 0: 409.5 (M+H*); Found: 409.1 (M+H'). Scheme 54: Example 54
NH
2 methyl piperidine-4-carboxylate NH 2 H N N Na(OAc) 3 BH, MeOH/DCM N N TOO N N0 N N NCHO N a-NO' BG IC Example 54 0 Method XLVII: Example 54. To a solution of the aldehyde BG (20 mg, 0.056 mmol) in 20 MeOH/CH 2 Cl 2 (1:1, 3m) was added methyl piperidine-4-carboxylate (40 mg, 0.28 mmol) and sodium triacetoxyborohydride (30 mg, 0.14 mmol) at 0 *C. The reaction mixture was stirred at room temperature for 2 days. The reaction was quenched by adding drops of 1N HCt, filtered and purified by reverse phase HPLC (5-100% acetonitrile in H 2 0) to give 146 WO 20101077613 PCT/US20091067002 Example 54. 'H NMR (CD 3 OD, 300 MHz): 5 7.53-7.48 (m, 4H), 4.92 (s, 2H), 4.39-4.33 (m, 4H), 4.09 (s, 2H), 3.70 (s, 3H), 3.55-3.51 (m, 2H), 3.08-2.99 (m, 2H), 2.70-2.66 (m, 1 H), 2.25-2.20 (m, 2H), 1.87-1.82 (m, 2H), 1.75-1.67 (m, 2H), 1.48-1.40 (m, 2H), 0.94 (t, J = 7.8 Hz, 3H). LCMS-ESI*: calc'd for C 2 5
H
35
N
6 0 4 : 483.6 (M+H 4 ); Found: 483.3 (M+H*). 5 Compound CF, Prepared using Method XI:
NH
2 NN NO 2 N 'H NMR (CD 3 OD, 300 MHz): 5 7.52-7.36 (m, 4H), 4.78 (s, 1H), 4.39 (t, J 6.3 Hz, 2H), 4.20 (s, 1H), 4.17 (q, J = 7.0 Hz, 2H), 4.08 (s, IH), 3.36 (s, 1H), 3.06 (m, 4H), 2.60 (qt, JFH 10 = 8.5 Hz, JHH = 6.3 Hz, 2H), 1.98 (m, 4H), 1.25 (t, J = 7.0 Hz, 3H). 1 9 F NMR (CD 3 OD, 282 MHz): . -66.8 (t, JFH = 8.5 Hz, 3F). LCMS-ESI*: calc'd for C 2 3
H
3 0
F
3
N
5 0 5 : 527.2 (M+H*); Found: 527.2 (M+H*). Example 55, Prepared using Method XII:
NH
2 H F3C,' NN- N 0o N N N 15 'H NMR (CD30D, 300 MHz): 8 7.40-7.20 (m, 4H), 4.77 (s, 1H), 4.40 (t, J = 6.3 Hz, 2H), 4.39 (s, 1H), 3.92 (s, 1H), 3.31 (s, 1H), 2.50 (m, 4H), 2.11-1.95 (m, 2H), 1.78 (m, 4H) [free base]. "F NMR (CD 3 0D, 282 MHz): 5 -66.8 (m, 3F). LCMS-ESl*: calc'd for
C
21
H
2 8
F
3
N
6 0 2 : 451.2 (M+H*); Found: 451.2 (M+H*). 20 Compound BI, Prepared using Method XI: 147 WO 2010/077613 PCT/US2009/067002
NH
2 N NO 2 N N N No 'H NMR (CD 3 OD, 300 MHz): 5 7.40-7.25 (m, 4H), 4.76 (s, 1H), 4.26 (t, J = 6.3 Hz, 2H), 4.17 (q, J = 7.0 Hz, 2H), 4.16 (s, IH), 3.72 (s, 1H), 3.32 (s, 1H), 2.63 (m, 4H), 2.28 (qt, JFs = 11.4 Hz, JHH = 6.3 Hz, 2H), 1.95-1.75 (m, 2H), 1.83 (m, 4H), 1.25 (t, J = 7.0 Hz, 3H). 5 ' 9 F NMR (CD 3 0D, 282 MHz): . -68.5 (t, JFH = 11.4 Hz, 3F). LCMS-ESI*: calc'd for
C
24
H
3 2
F
3 NO0 5 : 541.2 (M+H*); Found: 541.2 (M+H*). Example 56, Prepared using Method XII:
NH
2 H N N F3C O N O N~ 10 1 H NMR (CD 3 OD, 300 MHz): 6 7.40-7.20 (m, 4H), 4.79 (s, 1H), 4.27 (t, J = 6.3 Hz, 2H), 4.27 (s, 1H), 3.91 (s, 1H), 3.34 (s, 1H), 2.69 (m, 4H), 2.34-2.18 (m, 2H), 1.96-1.82 (m, 2H), 1.85 (m, 4H) [free base]. 1 9 F NMR (CD 3 OD, 282 MHz): 5 -68.5 (m, 3F). LCMS-ESIl: calo'd for C21H 28
F
3
N
6 0 2 : 465.2 (M+H*); Found: 465.2 (M+H*). 15 Compound CG, Prepared using Method XV parts I and 2:
NH
2 N
NO
2 MeS N O 2 Et N 'H NMR (CD30D, 300 MHz): 5 7.25-7.37 (m, 2H), 4.75 (s, 2H), 4.12 (m, 4H), 3.52 (s, 2H), 2.38 (s, 3H), 2.35 (m, 4H), 1.73 (m, 4H), 1.20 (t, J = 7 Hz, 3H). LCMS-ESF: calc'd for
C
2 1
H
2 9
N
6 0 4 S: 461.6 (M+H*); Found: 461.2 (M+H). 20 148 WO 2010/077613 PCT/US2009/067002 Compound CH, Prepared using Method VIII:
NH
2 N~ NO 2 MeO 2 S N N CO 2 Et (N 1 Ethyl-N-[4-amino-2-methanesulfonyl-5-nitropyrimidin-6-y],N-[2'-(pyrrolidin-1" ylmethyl)-benzyl]-glycinate: LCMS-ESI*: calc'd for C 21
H
2
N
6 0 6 S: 493.6 (M+H*); Found: 5 493.2 (M+H). Compound CI, Prepared using Method X:
NH
2 N '
NO
2 O N CO 2 Et N 0 'H NMR,(CD 3 0D, 300 MHz): 5 7.26-7.34 (m, 4H), 4.77 (s, 2H), 4.07-4.23 (m, 6H), 3.53 (s, 10 2H), 2.36 (m, 4H), 1.73 (m, 4H), 1.64 (m, 2H), 1.41 (m, 2H), 1.22 (t, J = 7 Hz, 3H), 0.94 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 24
H
35
N
6 0 5 : 487.6 (M+H*); Found: 487.2 (M+H*). Example 57, Prepared using Method XII:
NH
2
-
N TO ,---OIN- N N 15 1 H NMR (CD 3 0D, 300 MHz): 5 7.37-7.67 (m, 4H), 5.20 (s, 2H), 4.58 (s, 2H), 4.39 (t, J = 7 Hz, 2H), 4.16 (s, 2H), 3.61 (m, 2H), 3.31 (m, 2H), 2.21 (m, 2H), 2.09 (m, 2H), 1.67 (m, 2H), 1.42 (m, 2H), 0.90 (t, J = 7 Hz) - tHCI salt]. LCMS-ESI: calc'd for C 22
H
31
N
6 0 2 : 411.5 (M+H*); Found: 411.2 (M+H). 20 Compound CJ, Prepared using Method XI: 149 WO 2010/077613 PCT/US2009/067002
NH
2 Me N NO 2 0 N N CO 2 Et N 'H NMR (CD 3 0D, 300 MHz): 8 7.26-7.37 (m, 4H), 4.99 (m, 1H), 4.78 (s, 2H), 4.20 (m, 4H), 3.77 (s, 2H), 2.68 (m, 4H), 1.85 (m, 4H), 1.50-1.62 (m, 2H), 1.29 (m, 2H), 1.25 (m, 6H), 0.90 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 25
H
37
N
6 0 5 : 501.6 (M+H*); Found: 5 501.2 (M+H*). Example 58, Prepared using Method XII:
NH
2 H Me N N O O'N N N 'H NMR (CD 3 OD, 300 MHz): 6 7.64 (s, 1H), 7.49 (m, 3H), 5.16 (m, 1H), 4.94 (s, 2H), 4.38 10 (s, 2H), 4.18 (s, 2H), 3.47 (m, 2H), 3.16 (m, 2H), 2.16 (m, 2H), 2.03 (m, 2H), 1.55-1.72 (m, 2H), 1.32 (m, 5H), 0.87 (t, J = 7 Hz, 3H) - [HCI salt]. LCMS-ESF*: calc'd for
C
23
H
33
N
6 0 2 : 425.5 (M+H*); Found: 425.2 (M+H*). Compound CK, Prepared using Method Xl:
NH
2 Me N NO 2 OIN N CO 2 Et 15 N 'H NMR (CD 3 OD, 300 MHz): 8 7.26-7.37 (m, 4H), 4.99 (m, 1H), 4.78 (s, 2H), 4.20 (m, 4H), 3.77 (s, 2H), 2.68 (m, 4H), 1.85 (m, 4H), 1.50-1.62 (m, 2H), 1.29 (m, 2H), 1.25 (m, 6H), 0.90 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 2 5
H
37
N
6 0 5 : 501.6 (M+H*); Found: 501.2 (M+H*). 20 Example 59, Prepared using Method XII: 150 WO 2010/077613 PCT/US2009/067002
NH
2 H Me N N O N N N 'H NMR (CD 3 OD, 300 MHz): 5 7.64 (s, 1H), 7.49 (m, 3H), 5.16 (m, 1H), 4.94 (s, 2H), 4.38 (s, 2H), 4.18 (s, 2H), 3.47 (m, 2H), 3.16 (m, 2H), 2.16 (m, 2H), 2.03 (m, 2H), 1.55-1.72 (m, 2H), 1.32 (m, 5H), 0.87 (t, J = 7 Hz, 3H) - [HCI salt]. LCMS-ESIl: calc'd for 5 C 23
H
33
N
6 0 2 : 425.5 (M+H'); Found: 425.2 (M+H*). Compound CL, Prepared using Method Xl:
NH
2 Me N NO 2 O N N CO 2 Et NN 'H NMR (CD 3 OD, 300 MHz): 5 7.31 (m, 4H), 5.00 (m, 1H), 4.76 (s, 2H), 4.19 (q, J = 7 Hz, 10 2H), 4.13 (s, 2H), 3.64 (s, 2H), 2.56 (m, 4H), 1.82 (m, 4H), 1.62 (m, 2H), 1.40 (m, 2H), 1.25 (m, 6H), 0.90 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 25
H
37
N
6 0 5 : 501.6 (M+H*); Found: 501.2 (M+H*). Example 60, Prepared using Method XII:
NH
2 H Me NO 15 N 'H NMR (CD 3 0D, 300 MHz): 5 7.47-7.58 (m, 4H), 5.12 (m, 1H), 4.94 (s, 2H), 4.39 (s, 2H), 4.14 (s, 2H), 3,47 (m, 2H), 3.19 (m, 2H), 2.12 (m, 2H), 2.03 (m, 2H), 1.55-1.72 (m, 2H), 1.36 (m, 5H), 0.87 (t, J = 7 Hz, 3H) - [HCI salt]. LCMS-ESIl: calc'd for C 23
H
33
N
6 0 2 : 425.5 (M+H*); Found: 425.2 (M+H*). 20 Compound CM, Prepared using Method XI: 151 WO 2010/077613 PCT/US2009/067002
NH
2 Me N 0 NO 2 N N CO 2 Et N 'H NMR (CD 3 OD, 300 MHz): 6 7.31 (m, 4H), 5.00 (m, 1H), 4.76 (s, 2H), 4.19 (q, J = 7 Hz, 2H), 4.13 (s, 2H), 3.64 (s, 2H), 2.56 (m, 4H), 1.82 (m, 4H), 1.62 (m, 2H), 1.40 (m, 2H), 1.25 (m, 6H), 0.90 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C2 5
H
3 7 N0, 5 : 501.6 (M+H*); 5 Found: 501.2 (M+H*). Example 61, Prepared using Method XII:
NH
2 H Me N N o ,-- OIN N N 'H NMR (CD 3 0D, 300 MHz): 6 7.47-7.58 (m, 4H), 5.12 (m, 1H), 4.94 (s, 2H), 4.39 (s, 2H), 10 4.14 (s, 2H), 3.47 (m, 2H), 3.19 (m, 2H), 2.12 (m, 2H), 2.03 (m, 2H), 1.55-1.72 (m, 2H), 1.36 (m, 5H), 0.87 (t, J = 7 Hz, 3H) - [HCI salt]. LCMS-ESI*: calc'd for C 2 3
H
33
N
6 0 2 : 425.5 (M+H*); Found: 425.2 (M+H*). Compound CN, Prepared using Method X:
NH
2 N O N NO XOA N CO 2 Et 15 N 'H NMR (CD 5 OD, 300 MHz): 6 7.22-7.32 (m, 4H), 4.76 (s, 2H), 4.14-4.29 (m, 6H), 3.63 (s, 2H), 2.53 (m, 4H), 1.80 (m, 4H), 1.28 (m, 6H). LCMS-ESI*: calc'd for C 22
H
31
N
6 0 5 : 459.5 (M+H*); Found: 459.2 (M+H*). 20 Example 62, Prepared using Method XII: 152 WO 2010/077613 PCT/US2009/067002
NH
2 H NO N 0 NN 'H NMR (CD 3 0D, 300 MHz): 6 7.68 (s, 1H), 7.49 (m, 3H), 4.96 (s, 2H), 4.48 (q, J = 7 Hz, 2H), 4.41 (s, 2H), 4.15 (s, 2H), 3.47 (m, 2H), 3.18 (m, 2H), 2.17 (m, 2H), 2.03 (m, 2H), 1.37 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 20
H
27
N
6 0 2 : 383.5 (M+H*); Found: 383.1 5 (M+H*). Compound CM, Prepared using Method X:
NH
2 N NO 2 HO N N CO 2 Et N 1 H NMR (CD 3 0D, 300 MHz): 8 7.42-7.56 (m, 4H), 4.81 (s, 2H), 4.40 (s, 2H), 4.21 (q, J = 7 10 Hz, 2H), 4.12 (s, 2H), 3.50 (m, 2H), 3.17 (m, 2H), 2.17 (m, 2H), 2.00 (m, 2H), 1.25 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 2 0
H
27
N
6 0 5 431.5 (M+H*); Found: 431.2 (M+H'). Example 63, Prepared using Method XII: NH2 H HO NT HON NN 15 'H NMR (CD 3 OD, 300 MHz): 6 7.64 (s, 1H), 7.45-7.53 (m, 3H), 4.85 (s, 2H), 4.40 (s, 2H), 4.08 (s, 2H), 3.48 (m, 2H), 3.18 (m, 2H), 2.14 (m, 2H), 2.01 (m, 2H). LCMS-ESIl: caic'd for C 18
H
2 3
N
6 0 2 : 355.4 (M+H*); Found: 355.1 (M+H*). Compound CN, Prepared using Method IV and Method VI parts I and 2: 153 WO 2010/077613 PCT/US2009/067002 HN
CO
2 Et > N LCMS-ESI*: calc'd for C 1 2
H
27
N
2 0 2 : 291 4 (M+H*); Found: 291.2 (M+H).
NH
2 N NO 2 MeS N N CO 2 Et I N 'H NMR (CD30D, 300 MHz): 8 7.27 (s, 1H), 7.20 (m, 3H), 4.78 (d, J = 16 Hz, 1H), 4.63 (q, 5 J = 7 Hz, 1H), 4.55 (d, J = 16 Hz, 1 H), 4.20 (m, 2H), 3.56 (m, 2H), 2.44 (m, 2H), 2.36 (s, 3H), 1.76 (m, 4H), 1.63 (d, J = 7 Hz, 3H), 1.25 (t, J = 7 Hz, 3H). LCMS-ESIl: calc'd for
C
22
H
31
N
6 0 4 S: 475.6 (M+H*); Found: 475.2 (M+H). Compound CO, Prepared using Method VIII:
NH
2 N NO 2 MeO 2 S N N CO 2 Et 10 N LCMS-ESI*: calc'd for C2H 31
N
6
O
6 S: 507.6 (M+H 4 ); Found: 507.2 (M+H). Compound CP, Prepared using Method X:
NH
2 O N N CO 2 Et N 15 1 H NMR (CD30D, 300 MHz): 8 7.30 (s, 1H), 7.22 (m, 3H), 4.80 (d, J = 16 Hz, 1H), 4.57 (m, 2H), 4.12-4.25 (m, 4H), 3.58 (m, 2H), 2.46 (m, 4H), 1.76 (m, 4H), 1.62 (m, 5H), 1.44 (m, 2H), 1.24 (t, J = 7 Hz, 3H), 0.96 (t, J = 7 Hz). LCMS-ESI*: calc'd for C 2 5
H
37
N
6 0S: 501.6 (M+H*); Found: 501.2 (M+H). 154 WO 2010/077613 PCT/US2009/067002 Example 64, Prepared using Method XII:
NH
2 H N N 'H NMR (CD 3 0D, 300 MHz): 8 7.66 (s, 1H), 7.49 (m, 3H), 5.34 (d, J= 16 Hz, 1H), 4.64 (d, J = 16 Hz, 1H), 4.40 (m, 4H), 4.22 (q, J = 7 Hz, 1H), 3,46 (m, 2H), 3.18 (m, 2H), 2.17 (m, 5 2H), 2.03 (m, 2H), 1.70 (m, 2H), 1.44 (m, 5H), 0.93 (t, J = 7 Hz, 3H). LCMS-ESF: calc'd for C 23
H
33
N
6 0 2 : 425.5 (M+H*); Found: 425.2 (M+H). Compound CQ: Prepared via Method IV: HN CO 2 Et xb N 10 LCMS-ESr*: calc'd for C 12
H
27
N
2 0 2 : 291.4 (M+H*); Found: 291.1 (M+H). Compound CR, Prepared using Method Vil parts I and 2:
NH
2 HN C0 2 Et N N MeS N N CO 2 Et CQ CR 'H NMR (CD 3 OD, 300 MHz): 5 7.21-7.30 (m, 4H), 4.76 (d, J = 16 Hz, 1H), 4.57 (m, 2H), 15 4.20 (m, 2H), 3.58 (s, 2H), 2.50 (m, 4H), 2.36 (s, 3H), 1.78 (m, 4H), 1.62 (d, J = 7 Hz, 3H), 1.25 (t, J = 7 Hz, 3H). LCMS-ESIl: calc'd for C 22
H
31
N
6 O4S: 475.6 (M+H*); Found: 475.2 (M+H). Compound CS, Prepared using Method Vill: 155 WO 2010/077613 PCT/US2009/067002
NH
2 N NO 2 MeO 2 S N CO 2 Et N LCMS-ESI*: calc'd for C 22
H
31
N
6 0 6 S: 507.6 (M+H*); Found: 507.2 (M+H). Compound CT, Prepared using Method X:
NH
2 N NO 2 N CO 2 Et 5 N 'H NMR (CD 3 OD, 300 MHz): 6 7.23-7.31 (m, 4H), 4.78 (d, J = 16 Hz, 1H), 4.54 (m, 2H), 4.11-4.22 (m, 4H), 3.59 (m, 2H), 2.51 (m, 4H), 1.79 (m, 4H), 1.62 (m, 5H), 1.43 (m, 2H), 1.25 (t, J = 7 Hz, 3H), 0.95 (t, J = 7 Hz). LCMS-ESI*: calc'd for C 2 eHsyNsO 5 : 501.6 (M+H*); Found: 501.2 (M+H). 10 Example 65, Prepared using Method XII:
NH
2 H N 0 N NN 'CLNO 'H NMR (CD 3 0D, 300 MHz): 6 7.61 (d, J = 8 Hz, 2H), 7.49 (d, J = 8 Hz, 2H), 5.32 (d, J = 16 Hz, 1H), 4.65 (d, J = 16 Hz, 1H), 4.40 (m, 4H), 4.22 (q, J = 7 Hz, 1H), 3.48 (m, 2H), 15 3.19 (m, 2H), 2.17 (m, 2H), 2.03 (m, 2H), 1.70 (m, 2H), 1.45 (m, 5H), 0.94 (t, J = 7 Hz, 3H). LCMS-ESl*: calc'd for C 23
H
33
N
6 0 2 : 425.5 (M+H*); Found: 425.2 (M+H). Scheme 55: Example 66, Method Vill followed by Method X followed by Method X11 156 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2
NO
2 N 0NN 2 MethodVlt NO N 00 0 N.- N. N CU N CV
H
2
NNH
2 NO 0 Method X0 S"N N~' NO 2 ' N O V N C W NH-2 NH2 H O~ NeMethod X11 O N N 0V N O CW N Example 66 0-' 0 Compound CU, which was made from BU following the same procedure to make D, was 5 converted to CV using Method Vill, then the butoxy group was installed following Method X to give CW. Finally, the final product Example 66 was produced by following Method XIl. 'H NMR (DMSO-de, 300 MHz): 5 9.70 (s, 1 H), 8.05 (d, J = 5.1 Hz, 1 H), 6.73 (s, 1 H), 6.58 (d, J = 5.1 Hz, 1H), 6.22 (s, broad, 2H), 4.56 (s, 2H), 4.06-4.02 (m, 2H), 3.86 (s, 2H), 3.67-3.66 (m, 4H), 3.41-3.37 (m, 4H), 1.57-1.50 (m, 2H), 1.35-1.17 (m, 2H), 0.88 10 0.83 (m, 3H). LCMS--ESl*: calc'd for C20H21N703: 413.47 (M+H*); Found: 414.1 (M+H*). Example 67, Method X followed by Method XII 157 WO 2010/077613 PCT/US2009/067002
NH
2 NN 01 O N N N N 0 From the corresponding sulfone/sulfoxide, this compound was made following Method X using tetrahydrofurfurol as the alcohol. Method XII was then employed to achieve the final product. 'H NMR (DMSO-d, 300 MHz): 5 9.71 (s, broad, 1H), 8.05 (d, J = 5.1 Hz, 5 1H), 6.73 (s, IH), 6.54 (d, J = 4.8 Hz, 1H), 6.23 (s, broad, 2H), 4.56 (s, 2H), 4.01 (s, 2H), 3.87 (s, 2H), 3.71-3.58 (m, 7H), 3.46-3.39 (m, 4H), 1,93-1.75 (m, 4H). LCMS-ESr*: calc'd for C 21
H
28
N
7 0 4 : 441.48 (M+H*); Found: 442.1 (M+H*). Scheme 56: Prepared Via Method X1
NH
2 NH 2 01 N 1, N0 -1-"0 If N N Method X1 O N N O 0 BN CX N N\ 10 Compound CX was made following Method XI using the corresponding sulfone BN (125 mg) and (1S, 3R, 5R)-bicyclo [3.1.0]hexan-3-ol (440 mg) with 2.5 mL of DMF as cosolvent and 4 drops of TFA at 102 *C over 2 h. The mixture was quenched with water, diluted with EtOAc, and the pH was adjusted to = 8 using solid K 2 CO The mixture was 15 partitioned into EtOAc, and the organic layer was dried with Na 2
SO
4 , filtered, and concentrated in vacuo. Chromatography on silica gel using CH 2
CI
2 and MeOH/CH 2
C
2 as eluent afforded 23 mg of desired compound CX. LCMS-ESI*: calc'd for C 2 6
H
35
N
6 O: 510.59 (M+H*); Found: 511.1 (M+H*). 20 Scheme 57: Example 68, Method X11 158 WO 2010/077613 PCT/US2009/067002
NH
2 O N O Method XU O $ O O N01N CX Example 68 No No The unpurified CX from before was carried forward following: Method X11 in MeOH and stirred 3 h until starting material consumed by HPLC/LCMS. The mixture was diluted with
CH
2
C
2 , filteed through short plug of Celite, and the Celite was washed with copious 5 methano:CH 2
C
2 (50-50), and the filtrate was concentrated. The residue was re dissolved in acetonitrile, and filtered through a 0.2 micron filter to remove any residual Celite. Water was added, the mixture was frozen and lyophilized. 4.7 mg of Example 68 was obtained, 1 H NMR (DMSO-d, 300 MHz): 8 11.37 (s, broad, IH), 10.23-10.17 (m, 1 H), 7.54-7.39 (m, 4H), 5.35-5.25 (m, 1 H), 4.76 (m, 2H), 4.29-4.28 (m, 2H), 4.05 (m, 3H). 10 3.28 (s, broad, 2H), 2.98 (s, broad, 2H), 2.14-1.46 (m, 9H), 1.38-1.16 (m, 3H). LCMS ESl*: calc'd for C 24
H
1
N
6 0 2 : 434.53 (M+H*); Found: 435.1 (M+H*). Scheme 58: Example 69, Method X followed by Method X11
NH
2 NH2 NO2
N
2 O N2 ON N O - N N NO OHNA CV N N N oOH 0 N Method X K) 159 WO 2010/077613 PCT/US2009/067002
NH
2 NH 2 H N N0 2 N~ N 0O NO N Method XII O N O NAN N N CY Example 69 CN QN 0CO Starting from CV, Method X was employed to install the cyclopentoxy functionality on the pyrimidine ring and give CY. This intermediate was then advanced into Method XII to give rise to Example 69. 'H NMR: (DMSO-d 6 , 300 MHz): 5 9.70 (s, broad, 1H), 8.04 (s, 5 1H), 6.77 (s, 1H), 6.58 (s, 1H), 6.19 (s, broad, 2H), 5.08 (s, broad, 2H), 4.55 (s, broad, 2H), 3.85 (s, broad, 1H), 3.66 (s, broad, 4H), 3.38 (s, broad, 4H), 1.78-1.22 (m, broad, 8H). LCMS-ESI: calc'd for C 21
H
28
N
7 0 3 : 425.48 (M+H*); Found: 426.1 (M+H'). Scheme 59: Prepared Via Method XVII:
NH
2 C NO 2 1) NHi/MeOH, THF, 0*C N N2 Nt .-- N 0oc CI 2) DIPEA S N N A N O CZ 10 0 BZ Compound A (224 mg, 0.844 mmol) was dissolved in anhydrous THF (10 mL) and the mixture was stirred under N 2 (g) in an ice bath. A 7 N NH 3 in MeOH solution (131 pL, 0.92 mmol) in THF (1 mL) was added dropwise over 3 minutes. The reaction was stirred for 30 minutes, after which more 7 N NH 3 in MeOH solution (40 pL, 0.36 mmol) was added, 15 and the mixture was stirred for 30 more minutes. A solution of BZ (267 mg, 0.767 mmol) in anhydrous THF (2 mL) was added to the reaction, followed by DIPEA (267 pL, 1.53 mmol). The reaction mixture was then stirred for 2 hours at room temperature, diluted reaction with EtOAc and washed with saturated NaHCO 3 (aq) solution (2X) followed with saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 , filtered, and 20 concentrated under reduced pressure. Purification with silica gel chromatography (0-30% EtOAc in hexanes) gave CZ (353 mg, 0.663 mmol). 'H NMR (CDCl 3 , 300 MHz): 6 7.11 160 WO 2010/077613 PCT/US2009/067002 7.04 (m, 3H), 4.66 (s, 2H), 4.55 (s, 2H), 4.21 (m, 2H), 4.05 (s, 2H), 3.64 (m, 2H), 2.82 (m, 2H), 2.42 (s, 3H), 1.50 (s, 9H), 1.27 (t, J = 7.2Hz, 3H). Scheme 60; Compound CA Prepared Via Method XVIII:
NH
2 NH 2 N ~ NO 2 1) peracetic acid N NO 2 SN N NBO 30. C OI W S NN B 2) nBuOH, TFA 0 N Boc 5 0 CZ 0 CA Compound CZ (353 mg, 0.663 mmol) was dissolved in anhydrous acetonitrile (13 mL) and stirred under N 2 (g) in an ice bath. A 32% peracetic acid solution (700 pL, 3.22 mmol) was added and the mixture was stirred for 4-5 hours. To this was added saturated Na 2
S
2
O
3 (aq) solution and EtOAc and the mixture was stirred for 5 minutes. The organic 10 extract was then washed with NaHCO 3 (aq) solution followed with saturated NaCI(aq), dried over anhydrous Na 2
SO
4 , filtered, and concentrated under reduced pressure. The intermediate was added to n-BuOH (10 mL) and TFA (204 pL, 2.65 mmol) and then stirred at 100 *C for 7 hours. The mixture was concentrated under reduced pressure to give Compound CA that was used without purification. 15 Scheme 61: Example 70, Method XLVIII
NH
2 H
NH
2 H N - N 0 cyclopropylmethyl-Br N N 0 S N' N' DMF, DIPEA O N N Example 51 NH Example 70 N Example 51 (20 mg, 0.0417 mmor) was dissolved in anhydrous DMF (1 mL. To this was added bromomethylcyclopropane (4.5 pL, 0.0459 mmol) and DIPEA (16 pL, 0.0917 20 mmol), and the mixture was stirred for 14 hours. Purification with Prep HPLC Phenomenex Gemini 5u C18 column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to gave Example 70 (8.2mg, 0.0188 mmol). 1 H NMR
(CD
3 0D, 300 MHz): 5 7.32-7.26 (m, 3H), 4.73 (m, 1H), 4.42 (m, 3H), 4.11 (s, 2H), 3.87 (m, 1H), 3.43-3.19 (m, 8H), 1.77 (m, 2H), 1.48 (m, 2H), 1.26 (m, 1H), 0.96 (t, J = 7.5Hz, 25 3H), 0.83 (d, J = 7.2Hz, 2H), 0.52 (d, J = 4.5Hz, 2H). LCMS-ES*: calc'd for C22H 3
N
6
O
2 : 437.3 (M+H*); Found: 437.2 (M+H*). 161 WO 2010/077613 PCT/US2009/067002 Scheme 62: Example 71, Method XLVIII:
NH
2 H
NH
2 H N N O 2-iodopropane N 'IN O N N DMF, DIPEA 0 N N Example NH Example 7 N Example 51 (20 mg, 0.0417 mmol) was dissolved in anhydrous DMF (1 mL). To this was 5 added 2-iodopropane (4.6 pL, 0.0459 mmol) and DIPEA (16 pL, 0.0917 mmol), and the mixture was stirred for 14 hours. Purification with Prep HPLC Phenomenex Gemini 5u Ci column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to gave Example 71 (5.5 mg, 0.0130 mmol). 1 H NMR (CD 3 0D, 300 MHz): 5 7.30-7.28 (m, 3H), 5.52 (m, 1H), 4.68 (m, 1H), 4.45 (m, 4H), 3.78 (m, 2H), 3.38-3.15 (m, 6H), 1.75 10 (m, 2H), 1.47 (m, 8H), 0.97 (t, J = 7.5Hz, 3H). LCMS-ESI*: calc'd for C2 3
H
33
N
6 0 2 : 425.3 (M+H*); Found: 425.2 (M+H*). Scheme 63: Example 72, Method XLVIII:
NH
2 H
NH
2 H N N Bromomethylbutane N N O N DMF, DIPEA 0 N N lC N H N 0 N 2 Example 51 Example 72 15 Example 51 (20 mg, 0.0417 mmol) was dissolved in anhydrous DMF (1 mL). To this was added bromomethylbutane (5.2 pL, 0.0459 mmol) and DIPEA (16 pL, 0.0917 mmol), and the mixture was stirred for 14 hours. Purification with Prep HPLC Phenomenex Gemini 5u C1 column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to gave Example 72 (8.4 mg, 0.0186 mmol). 'H NMR (CD 3 0D, 300 MHz): 5 7.35 20 7.20 (m, 3H), 5.43 (m, 1H), 4.41 (m, 4H), 3.70 (m, 1H), 3.32-3.22 (m, 7H), 3.13 (m, 1H), 2.89 (m, 1H), 2.22 (m, 2H), 1.99 (m, 4H), 1.73 (m, 2H), 1.45 (m, 2H), 0.94 (t, J = 7.5Hz, 3H). LCMS-ESIl: calc'd for C 2 5
H
35
N
6 0 2 : 451.3 (M+H*); Found: 451.2 (M+H*). Compound DC, Method Vil followed by Method Vill followed by Method X 162 WO 2010/077613 PCT/US2009/067002
NH
2 C N NO 2 N NO 2 Method VIll -J N N O" Method Vill SIN CI O A NC DA
NH
2
NH
2 NN NO 2 N N0N 2 O - NO 0 Method X NS N N "-r_ O--N N- 0 \10 0o NC NC O DB DC Prepared by using Method Vil Compound DA: LCMS-ESI*: calc'd for C, 8
H
20
N
6 0 4 S: 417.4 (M+H'); Found: 417.0 (M+H*). After Method Vill: Compound DB: LCMS-ESI*: 5 calc'd for C 18
H
2 0
N
6 0 6 S: 449.4 (M+H'); Found: 448.8 (M+H). After Method X: Compound DC: 1 H NMR (CDCl 3 , 300 MHz): 3 7.68-7.48 (m, 4H), 5.10-4.90 (m, IH), 4.22-4.09 (m, 4H), 3.91 (d, J = 4.8 Hz, 2H), 1.72-1.65 (m, 2H), 1.52-1.40 (m, 2H), 1.29-1.19 (m, 6H), 0.95 (t, J = 7.5 Hz, 3H). LCMS-ESI 4 : calc'd for C 2 1
H
27
N
6 0 6 : 443.5 (M+H*); Found: 443.1 (M+H*). 10 Scheme 64: Compound DD Prepared Via Method XXXIll:
NH
2 NH 2 NINO2 HOAc, Zn NNNNO N OnO N N N 60 C DC NC DD Compound DD was made by a similar method to that used to make compound CE. LCMS-ESI*: calc'd for C 19
H
23
N
6 0 2 : 367.4 (M+H*); Found: 367.1 (M+H*). 15 Scheme 65: Example 73, Method XLIX: 163 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2 H NH N N 0 O , N 1) DIBAL, DCM O N 2) pyrrolidine, NaBH(OAc)3, DCM DD CN Example 73 N 'H NMR (CD30D, 300 MHz): 5 7.60-7.50 (m, 4H), 4.22-4.17 (m, 1H), 4.50-4.41 (m, 4H), 4.13 (d, J = 16.8 Hz, 1H), 3.60 (d, J = 17.1 Hz, 1H), 3.49-3.42 (m, 2H), 3.20-3.17 (m, 2H), 2.20-2.16 (m, 2H), 2.03-2.00 (m, 2H), 1.80-1.68 (m, 5H), 1.52-1.42 (m, 2H), 0.98 (t, J = 5 7.5 Hz, 3H). LCMS-ESI*: calc'd for C 23
H
33
N
6 0 2 : 425.5 (M+H*); Found: 425.3 (M+H*). Example 74, Method XXXIII followed by Method XLIX:
NH
2 H N NN N Nz N 'H NMR (CD30D, 300 MHz): 6 7.58-7.48 (m, 4H), 6.22-6.18 (m, IH), 4.45-4.35 (m, 4H), 10 4.12 (d, J = 17.1 Hz, 1H), 3.58 (d, J = 16.8 Hz, 1H), 3.49-3.42 (m, 2H), 3.22-3.18 (m, 2H), 2.20-2.16 (m, 2H), 2.03-2.00 (m, 2H), 1.80-1.45 (m, 7H), 0.98 (t, J = 7.5 Hz, 3H). LCMS
ESI
t : calc'd for C 23 H33N 6 0 2 : 425.5 (M+H*); Found: 425.2 (M+H*). Scheme 66: Example 75, Method L:
NH
2 piperidine-4-carboxylic acid NH 2 H N H Na(OAc) 3 BH, MeOH/DCM N N O N~ NO< NaBH 3 CN, DMF , xO N 01,N N CH N N BG CHO Example 75 N OH 15 0 To a solution of BG (20 mg, 0.056 mmol) in MeOH/CH 2
C
2 (1:1, 3 mL) was added piperidine-4-carboxylic acid (33 mg, 0.25 mmol) and sodium triacetoxyborohydride (30 mg, 0.14 mmol) at 0 *C. The reaction mixture was stirred at room temperature for 2 days. Then the solvent was removed and the residue was redissolved in DMF (2 mL). To the 20 mixture was added sodium cyanoborohydride (15mg, 0.24 mmol). The reaction mixture 164 WO 2010/077613 PCT/US2009/067002 was stirred at room temperature for 1 day, The reaction was quenched with 1N HCI, the mixture was diluted by MeOH, filtered and purified by reverse phase HPLC (5-100% acetonitrile in H 2 0) to give Example 75. 'H NMR (CD 3 0D, 300 MHz): 5 7.53-7.49 (m, 4H), 4.93 (s, 2H), 4.39-4.33 (m, 4H), 4.10 (s, 2H), 3.55-3.51 (m, 2H), 3.08-2.99 (m, 2H), 5 2.63-2.60 (m, 1H), 2.26-2.21 (m, 2H), 1.87-1.83 (m, 2H), 1.73-1.68 (m, 2H), 1.50-1.38 (m, 2H), 0.94 (t, J = 7.5 Hz, 3H). LCMS-ESl*: calc'd for C 24 H1 3
N
8 0 4 : 469.5 (M+H*); Found: 469.2 (M+H*). Scheme 67: Example 76, Prepared using Method XIV:
NH
2 NH2H
NO
2 O Raney Ni N N
H
2 I N N O HN N N H MeOH / H 2 H N 0 BT N Example 76 10A Example 76. A flask containing a solution of BT (23.0 mg) in MeOH (4.0 mL) was treated with a slurry of 50% w/v aq. Raney Nickel (1 mL). The system was purged/backfilled with
H
2 /vacuum several times, then stirred vigorously under a balloon of H 2 at 23 CC for 4 days. The reaction was filtered over Celite with the aide of MeOH/CH 2
C
2 . The filtrate 15 was concentrated, giving Example 76 as a yellow solid (20 mg, 99% yield). IH NMR
(CD
3 0D, 300 MHz): 5 (ppm) 7.31-7.17 (m, 4H), 4.77 (s, 2H), 3.65-3.58 (m, 2H), 3.61 (s, 2H), 3.17 (t, J = 7.0 Hz, 2H), 2.63-2.56 (m, 2H), 2.54-2.47 (m, 4H), 1.83-1.74 (m, 4H), 1.47-1.38 (m, 2H), 1.38-1.18 (m, 2H), 0.83 (t, J = 7.0 Hz, 3H). LCMS-ESF*: calc'd for
C
2 3 H3 4 N70: 424.3 (M+H*); Found: 424.2 (M+H*). 20 Scheme 68: Compound DE Prepared Via Method XIll
NH
2 NH 2 NO 0 N N2O BN DE N N 0 0 N The sulfone BN, (74.3 mg) was dissolved in 1.5 mL THF, and 300 pL of tetrahydrofurfuryl amine was added. The mixture was heated to 60 *C for one hour, then quenched by the 165 WO 2010/077613 PCT/US2009/067002 addition of water, and diluted with EtOAc. After washing organic layer with water, then brine, the organic extracts were dried with sodium sulfate, filtered, and concentrated in vacuo. The product DE was purified with silica gel chromatography, eluting with MeOH/CH 2
C
2 to give 35.3 mg. LCMS-ESI*: calc'd for C 25
H
35
N
7 0 5 : 513.59 (M+H*); 5 Found: 514.0 (M+H*), 257.6 (M+2H*/2). Scheme 69: Example 77, Method XII
NH
2 N N N0N 2 N 2 H N O O N NN 0 §NN N H O Method X11 N N N DE N Example 77 N Compound DE was advanced by Method X11 to give rise to give Example 77. 1 H NMR 10 (DMSO-d 6 , 300 MHz): 8 9.52 (s, broad, 1H), 7.27-7.21 (m 4H), 5.85 (s, broad, 2H), 4.67 (s, 2H), 3.96-3.86 (m, 1H), 3.70 (m, 3H), 3.64-3.45 (m, 3H), 3.35-3.08 (m, 3H), 2.49 (s, broad, 4H), 1.89-1.64 (m, 6H), 1.58-1.41 (m, 2H). LCMS-ES*: calc'd for C 23
H
32
N
7 0 2 : 437.54 (M+H*); Found: 438.2 (M+H*). 15 Scheme 70: Compound DF Prepared Via Method XIII
NH
2 NH 2 N - N 0N 2 N N 0N 2 N NNH2 II H N Method XIII N CV DF CN CN 0 0 Starting from CV, Method XIll was employed with butylamine. After purification on silica gel eluting with CH 2
CI
2 and a 20% MeOH/CH 2
C
2 gradient, Compound DF was obtained. LCMS-ES*: calc'd for C 23
H
32
N
7 0 2 : 488.54 (M+H*); Found: 489.1 (M+H*) , 245.0 ((M + 20 2H*)/2). 166 WO 2010/077613 PCT/US2009/067002 Scheme 71: Example 78, Method Xli
NH
2
NH
2 H
NO
2 N N;O N Method XII N N H NV O N DF Example 78 N0 Compound DF was advanced using Method Xi1 to give rise to Example 78. 1 H NMR (DMSO-d 6 , 300 MHz): 5 10.05 (s, 1H), 7.80 (s, broad, 1H), 7.51 (d, broad, J = 5.7Hz, 1H), 5 7.39 (s, broad, 2H), 7.03 (s, 1H), 6.81 (s, 1H), 4.71 (s, 2H), 4.10 (s, 2H), 3.72 (s, broad, 4H), 3.58 (s, broad, 4H), 3.16-3.14 (m, 2H), 1.38-1.16 (m, 4H), 0.78 (t, J 7 Hz, 3H). LCMS-ESI*: calc'd for C 20
H
29
N
8 0 2 : 412.49 (M+H*); Found: 413.2 (M+H*). Scheme 72: Example 79, Made Using Method XXI:
NH
2 H
NH
2 H N N O NaBH(OAc) 3 N I NO 1- O'N: NO N N CHO HOAc 1 -N NH Example 79N 10 BG xl 90 Compound BG (23 mg, 0.066 mmol) was added to anhydrous NMP (1 mL). To this was added methyl piperazine (73 pL, 0.66 mmol) and HOAc (19 pL, 0.33 mmol) and the mixture was stirred for 5 minutes. To this was added NaBH(OAc) 3 (140 mg, 0.66 mmol) 15 and the mixture was stirred for 16 hours. The mixture was diluted with MeOH and purified with Prep HPLC Phenomenex Gemini 5u C 1 8 column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to give Example 79 (16 mg, 0.036 mmol). 1 H NMR (CD 3 0D, 300 MHz): 6 7.48-7.45 (m, 4H), 4.44 (m, 2H), 4.19 (s, 2H), 4.11 (s, 2H), 3.52 (bs, 4H), 3.32 (bs, 3H), 1.75 (m, 2H), 1.46 (m, 2H), 0.95 (t, J = 7.2Hz, 3H). 20 LCMS-ESi*: calc'd for C 23
H
34
N
7 02: 440.3 (M+H*); Found: 440.2 (M+H*). Scheme 73: Example 80, Made Using Method XXI: 167 WO 2010/077613 PCT/US2009/067002
NH
2 H
NH
2 H N N O NaBH(OAc) 3 N N O OHN NO NN N BG / PN Example 80 H
NH
2 Compound BG (23 mg, 0.066 mmol) was added to anhydrous NMP (1 mL). To this was added 2-amino pyridine (62mg, 0.66 mmol) and HOAc (19 pL, 0.33 mmol) and the 5 mixture was stirred for 5 minutes. To this was then added NaBH(OAC) 3 (140 mg, 0.66 mmol) and the mixure was stirred for 16 hours. The mixture was diluted with MeOH and purified with Prep HPLC Phenomenex Gemini 5u Cjs column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to give Example 80 (6 mg, 0.014 mmol). 'H NMR (CD 3 OD, 300 MHz): 5 7.93 (m, 2H), 7.43-7.37 (m, 4H), 7.09 (d, J = 10 8.7Hz, 1H), 6.93 (m, 1H), 4.62 (s, 2H), 4.39 (t, J = 6.3Hz, 2H), 4.07 (s, 2H), 1.74 (m, 2H), 1.44 (m, 2H), 0.94 (t, J = 7.2Hz, 3H). LCMS-ESI': calc'd for C 23
H
28
N
7 0 2 : 434.2 (M+H); Found: 434,1 (M+H 4 ). Scheme 74 O 2-Methoxyethanol, CI N N N HCI (gas) e O N N BI 23 *C, 2 hrs only HND 15 Na I H DG Method LI: N-Cyanoacetyl-(2-methoxyethoxyl)-isouronium chloride (Compound DG). A suspension of cyanoacetylcyanamide, monosodium salt BI (20.0 g, 153 mmol) in 2-Mwthoxyethanol (100 mL) was treated with HCI (4.0 M in dioxane, 100 mL, 400 mmol). 20 During addition the suspension became more colloidal and there was a mild exotherm to an internal temperature of 52 "C. After 3 h, 10% w/v aq. NaHCO 3 (140 mL) was added cautiously (effervescence) until the pH of the aq. phase reached 8.0. The organic layer was collected, and the aqueous phase was extracted (2 x 100 mL EtOAc). All organic layers were combined, dried (Na 2
SO
4 ), and filtered over glass frits, and concentrated to a 25 volume of - 10 mL. The thick syrupy residue contains crude N-cyanoacetyl-(2 methoxyethoxyl)-isouronium chloride, DG, which is unstable and immediately used in the next reaction. LCMS-ESI*: calc'd for C 7
H
12
N
3 0 3 : 186.1 (M+H 4 ); Found: 186.0 (M+H'). Scheme 75 168 WO 2010/077613 PCT/US2009/067002
NH
2 CI NH 2 O Na 2 CO3 O N H 2 0, 90 C 16h O OH DG HD DH Method 11: 4-Amino-2-(2'-Methoxyethoxyl)-6-hydroxypyrimidine (Compound DH). An emulsion of all of the crude N-cyanoacetyl-butylisouronium chloride DG (28.4 g, 153 5 mmol) in a mixture of dioxane and 2-methoxyethanol (-10 mL) was treated with 10% w/v aq. Na 2
CO
3 (120 mL) and was stirred vigorously at 90 *C for 18 h. The reaction was then allowed to cool to 23 0C over the next hour- The reaction was extracted with several portions of EtOAc. The aqueous layer was neutralized to pH = 7.0 with conc. aq. HCI and concentrated to a semisolid. The organic layers and aqueous-derived semisolid were 10 combined, and triturated with hot MeOH/EtOAc. The system was cooled to 23 *C and filtered. The filtrate was concentrated and the residue purified via flash chromatography on silica gel (Eluent: DCM/MeOH 100:0 -- 80:20), giving semipure product Compound DH as an oily solid. The solid was was triturated with DCM, and the white crystals of pure Compound DH were obtained via filtration (584 mg, 2% yield over 2 steps). 'H NMR 15 (DMSO-d 6 , 300 MHz): 5 (ppm) 11.22 (s, broad, 1H), 10.43 (s, broad, 1H), 7.40 (s, broad, 1H), 6.39 (s, 1H), 4.36 (t, J = 4.6 Hz, 2H), 3.61 (t, J = 4.6 Hz, 2H), 3.30 (s, 3H). LCMS ESI*: calc'd for C 7
H
12
N
3 0 3 : 186.1 (M+H*); Found: 186.0 (M+H*). Scheme 76
NH
2 NH 2 N N Fuming HNO 3 N NO 2 - OIN OH 0 N OH DH DI
HNO
3 +
H
2
SO
4 HN'NO2 0 -+ 230C 0 N OH DJ 20 169 WO 2010/077613 PCT/US2009/067002 Method Lill: 4-Amino-2-(2'-methoxyethoxyl)-5-nitro-6-hydroxypyrimidine, DJ. A flask containing fuming aqueous HNO 3 (1.0 mL) at 0 *C was treated with 4-amino-2-(2 methoxyethoxy)-6-hydroxypyrimidine DH (500 mg) in portions over a 10 min period. The maroon reaction was treated with additional fuming HND 3 (200 pL). After 2 h, the reaction 5 was added dropwise to H 2 0 (10 mL) at 0 *C. pH was adjusted to 11.0 via portionwise addition of solid Na 2
CO
3 at 0 *C. Then 1.0 M aq HCI was added dropwise until the pH reached 3.0. The pink solid that precipitated was removed via filtration, and the filtrate was allowed to stand open to the air overnight. The solution went from purple to yellow. The filtrate was then directly loaded onto a C18 Teledyne Isco 'gold' 50 gram column and 10 flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -. 0:100) giving a mixture of DI and DJ. This mixture was dissolved in a minimum of DMSO and directly loaded onto a Teledyne Isco 'gold' 15 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -+ 0:100), giving separated products DI (higher polarity product)(175 mg, 28% yield) and DJ (lower polarity product)(44.2 mg, 7% yield). Data for Di (high polarity product): 1 H NMR 15 (DMSO-d 6 , 300 MHz): 5 (ppm) 12.15 (s, 1H), 8.83 (s, 1H), 8.79 (s, 1H), 4.50 (t, J = 4.6 Hz, 2H), 3.66 (t, J = 4.6 Hz, 2H), 3.31 (s, 3H). LCMS-ESI*: calc'd for C 7
H
11
N
4 0 5 : 231.1
(M+H
4 ); Found: 230.9 (M+H*). Data for DJ (high polarity product): 'H NMR (DMSO-Cd 6 , 300 MHz): 5 (ppm) 12.40 (s, broad, 1H), 6.38 (s, 1H), 4.43 (t, J = 4.6 Hz, 2H), 3.66 (t, J = 4.6 Hz, 2H), 3.31 (s, 3H). LCMS-ESl*: calc'd for C 7
H
11
N
4 0 5 : 231.1 (M+H*); Found: 230.8 20 (M+H*). An analytically pure sample of DI (36.3 mg) was treated with fuming HNO 3 (500 pL) at 0 OC. Then conc. aq, H 2
SO
4 (500 pL) was introduced dropwise over a 3 min period. After 5 min, the reaction was added to an ice-cold suspension of NaHCO 3 (2.52 9) in H 2 0 (10 mL) in a dropwise fashion. The reaction was allowed to warm to 23 OC. The 25 homogeneous solution was directly loaded onto a Teledyne Isco 'gold' 15 gram column and flashed (Eluent: 0.05% wlv aq. HCIICH 3 CN 95:5 -, 0:100), giving DJ (16.2 mg, 45% yield) having analytical data as detailed above. Scheme 77
H
2 N Br K2CO3 23 -+ 50 *C DK 30 * HCI 170 WO 2010/077613 PCT/US2009/067002 Method LIV: Ethyl N 8 -(4'-lodobenzyl)-glycinate, hydrochloride, compound DK. A suspension of ethyl glycinate hydrochloride (944 mg) in DMF (6.0 mL) was stirred for 5 min. p-lodobenzyl bromide (2.00 g) was added. The heterogeneous system was warmed to 50 0C and stirred for 5 min, during which time, most solids dissolved. K 2
CO
3 (2.80 g, 5 granular) was added steadily over 5 min. After 2 h, the reaction was cooled to 23 *C. Conc. aq. HCI (3.3 mL) was added, followed by H20 (7.0 mL). The heterogeneous mix was stirred for 15 min and filtered (the cake was washed with CH 3 CN (4 x 5 mL)). The net filtrate was concentrated until no CH3CN remained. The crude product solution was filtered through a 0.45 micron Teflon filter and loaded directly onto a Teledyne Isco 'gold' 10 100 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -* 0:100), giving DK (688 mg, 29% yield) as an HCI salt. 'H NMR (DMSO-d 6 , 300 MHz): 5 (ppm) 9.78 (s, 2H), 7.84 (d, J = 7.8 Hz, 2H), 7.36 (d, J = 7.8 Hz, 2H), 4.23 (q, J = 7.0 Hz, 2H), 4.15 (s, 2H), 3.95 (s, 2H), 1.25 (t, J = 7.0 Hz, 3H). LCMS-ESlt: calc'd for CH 15 1N0 2 : 320.0 (M+H); Found: 319.9 (M+H*). 15 Scheme 78 H 0 KOAc O K 2 CO3 O N PdC 2 (dppf) 2 HCI
N
0 -B N N*HG PhMe/EtOH/
H
2 0 DK 80 *C N DL Method LV: Compound DL. A suspension of ethyl N 2 -(4'-Iodobenzyl)-glycinate, 20 hydrochloride (DK)(200 mg), 3-(pyrrolidin-1'-ylmethyl) benzeneboronic acid pinacolate diester (162 mg), KOAc (166 mg), H 2 0 (1.0 mL), absolute EtOH (1.0 mL), and PhMe (2.0 mL) was degassed with argon via needle for 5 min. PdCl 2 (dppf) (12 mg) was added and the reaction was heated to 80 0C. After 12 h, no conversion was achieved, so K 2 C0 3 (233 mg) was added, followed after 2h, by additional PdC 2 (dppf) (12 mg). After the reaction 25 was complete, it was cooled to 23 0C and partitioned between 10% Na 2
CO
3 and EtOAc. The organic phase was collected, dried (Na 2
SO
4 ), filtered, and concentrated. The residue was treated with 1.0 M aq. HCI and CH 3 CN (minimum to achieve solution) and directly loaded onto a Teledyne Isco 'gold' 50 gram column and flashed (Eluent: 0.05% w/v aq. 171 WO 2010/077613 PCT/US2009/067002
HCI/CH
3 CN 95:5 - 0:100), giving DL (185.2 mg, 77% yield) as a white solid (in the dihydrochloride form). 'H NMR (CD 3 0D, 300 MHz): 6 (ppm) 7.96 (s, 1H), 7.85 (d, J = 8.3 Hz, 2H), 7.85-7.76 (m, 1H), 7.65 (d, J = 8.3 Hz, 2H), 7.64-7.58 (m, 2H), 4.49 (s, 2H), 4,35 (s, 2H), 4.33 (q, J = 7.0 hz, 2H), 4.03 (s, 2H), 3.60-3.48 (m, 2H), 3.31-3.27 (m, 2H), 2.23 5 2.13 (m, 2H), 2.12-2.00 (m, 2H), 1.33 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for
C
22
H
29
N
2 0 2 : 353.2 (M+H*); Found: 353.1 (M+H*). Scheme 79 H 0 0 B
K
2 00 3 B PdC1 2 (dPPf) NA +_____ * 2 HCI PhMe/EtOH/
H
2 0 DK 80 "C N DM 10 Method LVI: Compound DM. A suspension of ethyl N,-(4'-lodobenzyl)-glycinate, hydrochloride (DK)(200 mg), 4-(pyrrolidin-1'-ylmethyl) benzeneboronic acid pinacolate diester (162 mg), PdCl 2 (dppf) (24 mg) and K 2 C0 3 (233 mg) in PhMe (2.0 mL), absolute EtOH (1.0 mL), and H 2 0 (1.0 mL) was degassed with argon from a needle for 2 min. 15 Then the reaction was heated to 80 *C for 16 h. The reaction was cooled to 23 0 C, and the pH was adjusted to 1.0 using 1.0 M aq HCI (-4.0 mL). The reaction was concentrated to remove PhMe and EtOH, and H 2 0 was added along with CH 3 CN (minimum needed for salvation). The solution was loaded onto a Teledyne Isco 'gold' 50 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -- 0:100), giving DM (187 mg, 78% 20 yield) as a white solid (in the dihydrochloride form). 1 H NMR (CD 3 0D, 300 MHz): S (ppm) 7.891 (d, J = 7.6 Hz, 2H), 7.890 (d, J = 7.6 Hz, 2H), 7.67 (d, J = 7.6 Hz, 2H), 7.62 (d, J = 7.6 Hz, 2H), 4.44 (s, 2H), 4.33 (s, 2H), 4.32 (q, J = 7.0 Hz, 2H), 4.02 (s, 2H), 3.58-3.48 (m, 2H), 3.30-3.18 (m, 2H), 2.24-2.11 (m, 2H), 2.10-1.96 (m, 2H), 1.32 (t, J = 7.0 Hz, 3H). LCMS-ESI: calc'd for C 22
H
29
N
2 0 2 : 353.2 (M+H*); Found: 353.0 (M+H*). 25 Scheme 80 172 WO 20101077613 PCT/US20091067002 CI NH 2 N-, -Naq.NH 4 0H N' N CI NMP, 60 C ' H N CI 0 0 DN Method LVII: Compound DN. A solution of 2-carboxy-4,6-dichloropyrimidine (1.00 g) in NMP (10 mL) at 23 *C was treated dropwise with conc. aq. NH 4 0H (2.0 mL). Once 5 effervescence ceased, the reaction was slowly warmed to 60 OC, and held at this temperature for 4 h. The reaction was cooled to 23 *C, and H 2 0 (10 mL) was added, giving a milky suspension. Conc. aq. HCI (2.0 mL) was added dropwise. After 30 min, the suspension was filtered, and the filter cake was dried in a vacuum oven at 45 *C, giving DN (537 mg, 61%) as a white solid. 'H NMR (DMSO-d 6 , 300 MHz): 6 (ppm) 13.40 (s, 10 broad, 1H), 7.58 (app. s, broad, 2H), 6,58 (s, 1H). LCMS-ESI: compound does not ionize. Scheme 81:
NH
2
NH
2 N-Me-Propylamine N' N NMM, HATU HO N CI DMF, 60C N CI 00 DN DO 15 Method LVIII: Compound DO: A suspension of 4-amino-2-carboxy-6-chloropyrimidine (535 mg), DMF (3.0 mL), and N-Methyl Morpholine (1.72 mL) was heated to 60 *C. N Methyl-Propylamine (642 pL) was added, along with more DMF (1.0 mL, to aide fluidity). Then HATU (1.19 g) was introduced. After the reaction was complete, it was concentrated at 60 0C to remove volatile amines. The reaction was cooled to 23 0 C, and 20 1.0 M aq HCI (2.0 mL) was added. The solution was directly loaded onto a Teledyne Isco 'gold' 50 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -+ 0:100), giving DO (618 mg, 87%) as an orange oil, which solidified upon standing. 'H NMR (DMSO-ds, 300 MHz)(compound exists as a mixture of two amide rotamers at 23 OC with some associated protons having distinct resonances): 5 (ppm) 7.50 (app. s, broad, 2H), 25 6.49 (s, 1H), 3.36 (t, J = 7.6 Hz, 1.5 H, one rotamer), 3.06 (t, J = 7.6 Hz, 1.5 H, one rotamer), 2.93 (s, 1.5 H, one rotamer), 2.80 (s, 1.5H, one rotamer), 1.56 (app. qt, J = 7.6 Hz, 7.6 Hz, 2H, both rotamers), 0.91 (t, J = 7.6 Hz, 1.5H, one rotamer), 0.76 (t, J = 7.6 173 WO 2010/077613 PCTIUS2009/067002 Hz, 1.5H, one rotamer). LCMS-ESI*: calc'd for C 9
H
14 C1N 4 0 229.1 (M+H*) and 231.1 (M+2+H t ); Found: 229.1 (M+H*) and 231.1 (M+2+H*). Scheme 82
NH
2
HNO
3
NH
2 N-
H
2 SO4 NJINO2 N CI 0-+23*C N N C1 0 0 5 DO DIP Method LIX: Compound DP. A flask containing the pyrimidine DO (538 mg) was cooled to 0 0C. Fuming HNO 3 (1.0 mL) was added, After the initial exotherm had subsided, conc. aq. H 2
SO
4 (1.0 mL) was introduced over a 3 min period. The reaction was then allowed to 10 warm to 23 *C. After 45 h, the reaction was added dropwise to an ice-cold suspension of NaHCO 3 (5.0 g) in H 2 0 (20 mL). A yellow precipitate formed. The quenched reaction was then treated with CH 3 CN (4.5 mL) and DMF (1.5 mL). The now homogeneous solution was directly loaded onto a Teledyne Isco 'gold' 50 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -- 0:100), giving DP (180.4 mg, 28% yield) as a colorless 15 oil. 1 H NMR (CDCIs, 300 MHz)(compound exists as a mixture of two amide rotamers at 23 *C with some associated protons having distinct resonances): 6 (ppm) 7.91 (app. s, broad, 2H), 3.50 (t, J = 7.6 Hz, 1H, single rotamer), 3.17 (t, J = 7.6 Hz, 1H, single amide rotamer), 3.10 (s, 1.5H, single rotamer), 2.98 (s, 1.5H, single rotamer), 1.68 (app. qt, J = 7.6 Hz, 7.6 Hz, 2H, both rotamers), 0.97 (t, J = 7.6, 1.5H, single rotamer), 0.85 (t, J = 7.6 20 Hz, 1.5H, single rotamer). LCMS-ESIl: calc'd for CH 1 3
CIN
5 0 3 : 274.1 (M+H*) and 276.1 (M+2+H*); Found: 274.0 (M+H*) and 276.0 (M+2+H*). Scheme 83:
NH
2 N
NO
2
NH
2 HN I I
NO
2 NEt 3 N N N N 0 + I - 0 N CI DMF O 23 "C sFHCl N DIR E~No E DQ 174 WO 2010/077613 PCT/US2009/067002 Method LX: Compound DQ. A solution of E (30 mg) in DMF (500 pL) was added to a vial containing the pyrimidine DP (30 mg). Finally, Et 3 N (31 pL) was added at 23 oC. After 2 h, the reaction was complete. 1.0 M aq. HCI (300 pL) and CH 3 CN (50 pL). The reaction 5 was loaded directly onto a Teledyne Isco 'gold' 5.5 gram column and flashed (Eluent 0.05% w/v aq. HCI/CH 3 CN 95:5 --+ 0:100), giving DQ (16.4 mg, 27% yield) as a monohydrochloride salt. 1 H NMR (CDC 3 , 300 MHz)(compound exists as a mixture of two amide rotamers at 23 oC with some associated protons having distinct resonances): 6 (ppm) 12.65 (s, broad, 1H), 7.71 (app. s, broad, 2H), 7.44-7.26 (m, 4H), 4.83 (s, 2H), 10 4.30-4.02 (m, 4H), 3.63-3.57 (m, 2H), 3.43 (t, J = 7.6 Hz, 1H, single rotamer), 3.17 (t, J = 7.6 Hz, IH, single rotamer), 3.02 (s, 1.5H, single rotamer), 3.01-2.79 (m, 4H), 2.92 (s, 1.5H, single rotamer), 2.30-2.20 (m, 2H), 2.20-2.10 (m, 2H), 1.61 (app. qt, J = 7.6 Hz, 7.6Hz, 2H, both rotamers), 1.27 (t, J = 6.8 Hz, 3H), 0.93 (t, J = 7.6 Hz, 1.5H, single rotamer), 0.85 (t, J = 7.6 Hz, 1.5H, single rotamer). LCMS-ESr*: calc'd for C 25
H
36
N
7 0 5 : 15 514.3 (M+H*); Found: 514.2 (M+H). Scheme 84: Example 81
NH
2
NH
2 N NO 2 N N O N N N AcOH, 23*C N N *HCI 000 No No DO Example 81 20 Method LXI: Example 81. A solution of the amide DQ (16.4 mg) in glacial AcOH (1.64 mL) was treated with zinc powder (48 mg) at 23 0C. After the reaction was complete (3 h), it was diluted with H 2 0 (300 pL) and loaded onto a Teledyne Isco 'gold' 5.5 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 - 0:100), giving Example 81 (1.8 mg, 14% yield) as a white solid in monohydrochloride form. 'H NMR (CD 3 OD, 300 25 MHz)(compound exists as a mixture of two amide rotamers at 23 OC with some associated protons having distinct resonances): 5 (ppm) 7.60-7.42 (m, 4H), 5.50 (s, 2H), 4.94 (s, 2H), 4.38 (s, 2H), 4.18 (app. s, 1H, single rotamer), 4.16 (app. s, 1H, single rotamer), 3.55-3.41 (m, 2H), 3.40-3.25 (m, 2H), 3.14 (s, 1.5H, single rotamer), 3.07 (s, 175 WO 2010/077613 PCT/US2009/067002 1.5H, single rotamer), 2.22-2.08 (m, 2H), 2.08-1.99 (m, 2H), 1.68-1.64 (m, 2H, both rotamers), 0.97 (t, J = 7.6 Hz, 1.5H, single rotamer), 0.75 (t, J = 7.6 Hz, 1.5H), single rotamer). LCMS-ESI*: calc'd for C 23
H
32
N
7 0 2 : 438.3 (M+H*); Found: 438.2 (M+H*) and 219.7 ((M+2H*)/2). 5 Scheme 85
NH
2
NH
2
NO
2 OH 0 NA NO O''s 'ThN TA N N" ,- N 6--o F 0 100 *C .HCI NoN BN DR Method LXII: Compound ZZ. A suspension of the sulfone (BN)(15.8 mg), (R)-1 10 methoxy-2-propanol (300 pL), and TFA (10 pL) was heated to 100 0C for 17.5 h. The reaction was cooled to 23 OC, diluted with H 2 0 (600 pL) and loaded directly onto a Teledyne Isco 'gold' 5.5 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN .95:5 -+ 0:100), giving DR (13 mg, 76% yield) as a monohydrochloride salt. 1 H NMR (CDCl 3 , 300 MHz): S (ppm) 12.64 (s, 1H), 9.68 (s, 1H), 8.36 (s, 1H), 7.93 (s, IH), 7.49 15 7.20 (m, 4H), 5.27 (s, broad, 2H), 4.87 (s, 2H), 4.40-4.08 (m, 5H), 3,67-3.30 (m, 4H), 3.34 (s, 3H), 2.85-2.70 (m, 2H), 2.30-2.20 (m, 2H), 2.20-2.10 (m, 2H), 1.35-1.18 (m, 6H). LCMS-ESI*: calc'd for C 24
H
35 N30 6 : 503,3 (M+H*); Found: 503.2 (M+H t ). Scheme 86 NH
NH
2
NH
2 HATU N NO 2 + NN NO 2 Et 3 N N N . 2HC0 O O N OH DMF 0 DI 23 *C N N H HC\ DL 20 DS 176 WO 2010/077613 PCT/US2009/067002 Method LXIll: Compound DS. A suspension of nitropyrimidine (DI)(1 5.3 mg), amino acid ester (DL)(31.4 mg), and DMF (589 pL) was treated with Et 3 N (37 pL). HATU (33 mg) was introduced, followed by more DMF (589 pL) to aide fluidity. After 1h, the completed reaction was treated with 1.0 M aq. HOI (300 pL) followed by CH3CN (100 pL). The 5 reaction was directly loaded onto a Teledyne Isco 'gold' 15 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -- + 0:100), giving DS (31.1 mg, 78% yield) as a monohydrochloride salt. 1 H NMR (CDC1 3 , 300 MHz): 6 (ppm) 12.74 (s, broad, 1H), 8.96 (s, broad, 1H), 8.24 (s, broad, 1H), 8.07 (s, 1H), 7.72-7.40 (m, 5H), 7.35 (d, J = 7.0 Hz, 2H), 4.82 (s, 2H), 4.47 (s, 2H), 4.30-4.10 (m, 6H), 3.62-3.51 (m, 4H), 3.35 (s, 3H), 2.94 10 2.70 (m, 2H), 2.29-2.12 (m, 2H), 2.11-2.00 (m, 2H), 1.27 (t, J = 7.0 Hz, 3H). LCMS-ES*: calc'd for C 29
H
7
N
6 0 6 : 565.3 (M+H*); Found: 565.3 (M+H*). Scheme 87
NH
2 H NH2 H H2 H N N O DM 0 OH:N N N O MnO 2 N 1,NO 1N _N 9 DMSO - --OAN NXOH N >'O
H
2 0, 80 C Ir - - HC I HCI Example 49 Example 82 Example 83 (free base form) 15 Method LXIV: Examples 82 and 83: A solution of Example 49 (free base, 10.2 mg) in DMSO (800 pL) and H 2 0 (200 pL) was heated to 80 OC and treated with MnO 2 (85%, activated, from Sigma-Aldrich, 21 mg). After 45 min, the reaction was quickly cooled to 23 'C and filtered through a 0.45 micron Teflon filter. The filtrate was directly loaded onto a 20 Teledyne Isco 'gold' 5.5 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 -+ 0:100), giving Example 82 (1.0 mg, 8.7% yield, higher-polarity product) as a monohydrochloride salt. 1 H NMR (CD 3 0D, 300 MHz): 6 (ppm) 7.60-7.39 (m, 4H), 5.48 (app. s, 1H), 5.38 (app. d, J = 15.2 Hz, 1H), 5.05 (s, 1H), 4.36 (s, 2H), 4.36-4.34 (m, 2H), 3.60-3.40 (m, 2H), 3.32-3.10 (m, 2H), 2.20-2.05 (m, 4H), 1.69 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 25 1.41 (qt, 7.6 Hz, 7.6 Hz, 2H), 0.93 (t, J = 7.0 Hz, 3H). LCMS-ES*: calc'd for C 22
H
31
N
6 0 3 : 427.2 (M+H*) and calc'd for C 22
H
29
N
6 0 2 : 409.2 (M- OH); Found: 409.1 (M- OH)*. In addition, Example 83 (5.7 mg, 50% yield, lower-polarity product) was obtained as a monohydrochloride salt. 'H NMR (CD 3 OD, 300 MHz): 5 (ppm) 7.60-7.39 (m, 4H), 5.50 (s, 2H), 4.34 (q, J = 7.0 Hz, 2H), 4.33 (s, 2H), 3.48-3.39 (m, 2H), 3.20-3.04 (m, 2H), 2.20 177 WO 2010/077613 PCT/US2009/067002 2.05 (m, 2H), 2.05-1.90 (m, 2H), 1.70 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1,42 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.93 (t, J = 7.6 Hz, 3H). LCMS-ESl*: calc'd for C22H 29
NO
3 : 425.2 (M+H*); Found: 425.2 (M+H*). 5 Scheme 88:
NH-
2
NH
2 H NO MnO 2 O SN ' N ~ DM80 01~o N. N 0
H
2 0, 23 "C Example 4 Example 84 (free base form) Method LXV: Example 84 A solution of Example 4 (free base form, 9.9 mg) in DMSO (2.4 mL) was treated with H 2 0 (600 pL) followed by MnO 2 (85%, activated, from Sigma 10 Aldrich, 104 mg) at 23 OC. Once the reaction was complete, it was filtered through a 0.45 micron Teflon filter. The filtrate was directly loaded onto a Teledyne Isco 'gold' 5.5 gram column and flashed (Eluent: 0.05% w/v aq. HCIICH 3 CN 95:5 -- + 0:100), giving Example 84 (3.0 mg, 27% yield) as a monohydrochloride salt. 'H NMR (CD 3 0D, 300 MHz): 6 (ppm) 7.53 (d, J = 7.8 Hz, 2H), 7.46 (d, J = 7.8 Hz, 2H), 5.50 (s, 2H), 4.34 (s, 2H), 4.32 (t, J = 15 7.6 Hz, 2H), 3.50-3.38 (m, 2H), 3.21-3.09 (m, 2H), 2.25-2.18 (m, 2H), 2.17-1.99 (m, 2H), 1.70 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.45 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.94 (t, J = 7.6 Hz, 3H). LCMS-ESI*: calc'd for C 2 2
H
2 9
N
6 0 3 : 425.2 (M+H*); Found: 425.1 (M+H*). Scheme 89: 20
NH
2
NH
2 N N NO 2 TEA, THF N NO 2 O N OTs EtO 2 C^N I .OI N N CO 2 Et BM oc DT NBoc 178 WO 2010/077613 PCT/US2009/067002 Method LXVI: Compound DT: To a solution of compound BM (220 mg, 0.57 mmol) in THF, was added triethyl amine (160 pL, 1.14 mmol), tert-butyl 6-((2-ethoxy-2 oxoethylamino)methyl)-3,4-dihydroisoquinoline-2(1 H)-carboxylate (200 mg, 0.57 mmol). The reaction mixture was stirred at room temperature for 2h. After reaction finished, the 5 reaction mixture was diluted with EtOAc, treated with saturated aq. NaHCO 3 , and extracted by EtOAc (3 x). The organic layer was combined, dried over MgSO 4 , filtered, concentrated, and purified on a silica gel column. (Eluent: 0 -- 100% EtOAc in Hexanes), giving Compound DT. 'H NMR (CDC1 3 , 300 MHz): d (ppm) 7.30-7.06 (m, 3H), 4.66 (s, 2H), 4.54 (s, 2H), 4.21-4.10 (m, 4H), 4.03 (s, 2H), 3.62-3.34 (m, 2H), 2.81-2.79 (m, 2H), 10 1.69-1.65 (m, 2H), 1.50 (s, 9H), 1.48-1.43 (m, 2H), 1.28-1.22 (m, 3H), 0.96-0.89 (m, 3H). Compound DU: Prepared by Method I: 0 HO OH 0 HN CO 2 Et 2* N ZZ Compound DU was prepared according to Method 1: (Free base form of DU was 15 converted to dioxalic acid salt by slurrying with 2.0 equiv. of oxalic acid in warm absolute EtOH. Precipitate was dried in a vacuum oven after filtration). 'H NMR (D 2 0, 300 MHz): d 7.46 (s, 4H), 4.29 (s, 2H), 4.25 (s, 2H), 4.16 (q, J = 7.0 Hz, 2H), 3.90 (s, 2H), 3.39 (m, 2H), 3.06 (m, 2H), 2.04 (m, 2H), 1.84 (m, 2H), 1.15 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for C 1
H
2 5
N
2 0 2 : 277.4 (M+H*); Found: 277.1 (M+H*). 20 Compound DV, Method LX
NH
2 N NO 2 NN N 0 0 .HCI DV 179 WO 2010/077613 PCT/US2009/067002 Compound DV was prepared from Compound DU and Compound DP according to Method LX: 11% yield; compound is a monohydrochloride salt. 1 H NMR (CDC1 3 , 300 MHz)(compound exists as a mixture of two amide rotamers at 23 *C with some associated protons having distinct resonances): 3 (ppm) 12.75 (s, 1H), 7.66 (app. s, 5 broad, 2H), 7.38 (app. s, broad, 2H), 4.76 (s, 2H), 4.33-4.27 (m, 4H), 3.62 (s, 2H), 3.16 (t, J = 7.6 Hz, 1H, single rotamer), 3.02 (t, J = 7.6 Hz, 1H, single rotamer), 2.91 (s, 1.5H, single rotamer), 2.90-2.80 (m, 2H), 2.84 (s, 1.5H, single rotamer), 2.80-2.65 (m, 2H), 2.30-2.18 (m, 2H), 2.18-2.06 (m, 2H), 1.64 (app. qt, J = 7.6 Hz, 7.6 Hz, 2H, both rotamers), 1.24 (t, J = 6.8 Hz, 3H), 0.97 (t, J = 7.6 Hz, 1.5H, single rotamer), 0.87 (t, J = 10 7.6 Hz, 1.5H, single rotamer). LCMS-ESI': calc'd for C 2 5
H
36
N
7 0 5 : 514.3 (M+H'); Found: 514.2 (M+H'). Example 85: Prepared by Method LXI:
NH
2 H N - N 0 N ; N N 0 .HCI N Example 85 15 Example 85 was obtained in 20% yield as a white solid in the form of a monohydrochloride salt. 'H NMR (CD 3 0D, 300 MHz)(compound exists as a mixture of two amide rotamers at 23 *C with some associated protons having distinct resonances): S (ppm) 7.62-7.53 (m, 2H), 7.50-7.45 (m, 2H), 5.50 (s, 2H), 4.97 (s, 2H), 4.40 (s, 2H), 4.19 20 (app. s, 1H, single rotamer), 4.15 (app. s, IH, single rotamer), 3.55-3.40 (m. 2H), 3.40 3.25 (m, 2H), 3.20 (s, 1.5H, single rotamer), 3.09 (s, 1.5H, single rotamer), 2.30-1.95 (m, 4H), 1.69-1.65 (m, 2H, both rotamers), 0.96 (t, J = 7.6 Hz, 1.5 H, single rotamer), 0.76 (t, J = 7.6 Hz, 1.5K, single rotamer). LCMS-ESI*: calc'd for C 2 3
H
32
N
7 0 2 : 438.3 (M+H*); Found: 438.2 (M+H') and 219.7 ((M+2H*)/2). 25 Compound 86: Prepared by Method LXII: 180 WO 2010/077613 PCT/US2009/067002 N2 O --IOIN N O-, 0 *HCI No DW Compound DW was prepared in 38% yield as a monohydrochloride salt. 1 H NMR (CDCl 3 , 300 MHz): 5 (ppm) 12.63 (s, 1H), 7.75-7.30 (m, 4H), 5.24-5.06 (m, 2H), 4.79 (s. 5 2H), 4.32-4.16 (m, 5H), 3.66-3.35 (m, 4H), 3.34 (s, 3H), 2.85-2.70 (m, 2H), 2.30-2.20 (m, 2H), 2.20-2.10 (m, 2H), 1.34-1.20 (m, 6H). LCMS-ESI*: calc'd for C 2 4
H
3 5
N
6 0 6 : 503.3 (M+H*); Found: 503.2 (M+H*). Example 87: Prepared by Method LXI:
NH
2 N N TO O% O NN~' .2HCl N 10 Example 87 Example 87 was obtained in 43% yield as a dihydrochloride salt. 'H NMR (CD 3 0D, 300 MHz): 8 (ppm) 7.56 (s, 1H), 7.54-7.50 (m, 3H), 5.38-5.30 (m, 1H), 4.94 (s, 2H), 4.39 (s, 2H), 4.17 (s, 2H), 3.60-3.48 (m, 4H), 3.34 (s, 3H), 3.26-3.17 (m, 2H), 2.22-2.12 (m, 2H), 15 2.11-1.99 (m, 2H), 1.32 (d, J = 6.4 Hz, 3H). LCMS-ESI': calc'd for C22H 31
N
6
O
3 : 427.2
(M+H
4 ); Found: 427.2 (M+H), 214.2 ((M+2H*)12). Example 88: Prepared by Method LXI: 181 WO 2010/077613 PCT/US2009/067002
NH
2 O O . 2HCl N Example 88 Example 88 was obtained in 18% yield as a dihydrochloride salt. 1 H NMR (CD 3 0D, 300 MHz): 8 (ppm) 7.54 (s, 1H), 7.53-7.50 (m, 3H), 5.37-5.29 (m, 1H), 4.94 (s, 2H), 4.39 (s, 5 2H), 4.14 (s, 2H), 3.58-3.45 (m, 4H), 3.34 (s, 3H), 3.22-3.18 (m, 2H), 2.27-1.96 (m, 4H), 1.31 (d, J = 6.4 Hz, 3H). LCMS-ESI*: calc'd for C 22
H
3 1
N
6 0 3 : 427.2 (M+H*); Found: 427.2 (M+H'), 214.2 ((M+2H 4 )12). Compound DX: Prepared by Method LXIII:
NH
2 N NNO 2 OIN N O eHCI 10DX Compound DX was prepared in 54% yield as a monohydrochloride salt. 'H NMR
(CD
3 0D, 300 MHz): 6 (ppm) 7.76 (d, J = 7.6 Hz, 2H), 7.66 (d, J = 7.6 Hz, 2H), 7.63 (d, J = 7.6 Hz, 2H), 7.48 (d, J = 7.6 Hz, 2H), 4.91 (s, 2H), 4.48 (t, J = 4,4 Hz, 2H), 4.44 (s, 2H), 15 4.30 (s, 2H), 4.23 (q, J = 7.0 Hz, 2H), 3.65 (t, J = 4.4 Hz, 2H), 3.60-3.48 (m, 2H), 3.35 (s, 3H), 3.30-3.17 (m, 2H), 2.25-2.15 (m, 2H), 2.10-1.99 (m, 2H), 1.27 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for C 2 9
H
37
N
6 0 6 : 565.3 (M+H*); Found: 565.1 (M+H*). Compound DY: Prepared by Method LXIII: 182 WO 2010/077613 PCT/US2009/067002
NH
2 N NO 2 N0 N NN N HCI DY Compound DY was prepared in 75% yield as a monohydrochloride salt. 'H NMR (CDC13, 300 MHz): 5 (ppm) 12.76 (s, broad, 1H), 8.85 (s, broad, IH), 8.21 (s, broad, 1H), 8.07 (s, 5 1H), 7.72-7.40 (m, 5H), 7.40-7.33 (m, 2H), 4.80 (s, 2H), 4.37-4.10 (m, 6H), 3.73-3.59 (m, 2H), 2.94-2.79 (m, 2H), 2.30-2.15 (m, 2H), 2,14-1.96 (m, 2H), 1.75-1.62 (m, 2H), 1.43 1.30 (m, 2H), 1.27 (t, J = 7.0 Hz, 3H), 0.91 (t, J = 7.3 Hz, 3H). LCMS-ESI*: calc'd for
C
30
H
3 9
N
6 0 5 : 563.3 (M+H t ); Found: 563.3 (M+H*). 10 Compound DZ: Prepared by Method LXIII:
NH
2 N '--NO 2 O N N -~ 0 N HCI DZ Compound DZ was prepared in 54% yield as a monohydrochloride salt. 1 H NMR
(CD
3 0D, 300 MHz): S (ppm) 7.75 (d, J = 7.9 Hz, 2H), 7.66 (d, J = 7.9 Hz, 2H), 7.63 (d, J = 15 7.9 Hz, 2H), 7.47 (d, J = 8.3 Hz, 2H), 4.94 (s, 2H), 4.43 (s, 2H), 4.39 (t, J = 6.7 Hz, 2H), 4.35 (s, 2H), 4.22 (q, J = 7.0 Hz, 2H), 3.58-3.48 (m, 2H), 3.30-3.16 (m, 2H), 2.25-2.10 (m, 2H), 2.10-1.96 (m, 2H), 1.71 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.45 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 1.27 (t, J = 7.0 Hz, 3H), 0.93 (t, J = 7.6 Hz, 3H). LCMS-ESI*: calc'd for C 3 0
H
3 9 N,0 5 : 563.3 (M+H*); Found: 563.2 (M+H*). 20 Example 89: Prepared by Method LXV: 183 WO 2010/077613 PCT/US2009/067002
NH
2 H O N 0 0N~ N t O HCI N,,r Example 89 Example 89 was obtained in 35% yield as a monohydrochloride salt. 'H NMR (CD 3 0D, 300 MHz): 5 (ppm) 7.55-7.38 (m, 4H), 5.58 (s, 2H), 4.73 (s, 2H), 4.31 (t, J = 7.6 Hz, 2H), 5 3.72-3.59 (m, 2H), 3.42-3.30 (m, 2H), 2.32-2.20 (m, 2H), 2.20-2.02 (m, 2H), 1.71 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.42 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.94 (t, J = 7.6 Hz, 3H). LCMS-ESV*: calc'd for C22H2gNO 3 : 425.2 (M+H*); Found: 425.2 (M+H*). Example 90: Prepared by Method LXV:
NH
2 H I N O H N N N N e HCI 10 Example 90 Example 90 was obtained in 14% yield as a monohydrochloride salt. 'H NMR (CD 3 0D, 300 MHz): 8 (ppm) 7.70-7.40 (m, 4H), 4.36 (q, J = 7.6, 2H), 3.60-3.20 (m, 4H), 2.25-1.95 (m, 4H), 1.60-1.20 (m, 4H), 0.94 (t, J = 7.6 Hz, 2H); other resonances were too broad or 15 poorly resolved to be labeled definitively. LCMS-ESF: calc'd for C 22
H
30
N
7 0 2 : 424.2 (M+H'); Found: 424.2 (M+H'). Example 91: Prepared by Method LXV: 184 WO 2010/077613 PCT/US2009/067002
NH
2 N 0 N O N N O N *HCI Example 91 Example 91 was obtained in 80% yield as a monohydrochioride salt. 'H NMR (CD 3 0D, 300 MHz): 8 (ppm) 7.60-7.35 (m, 4H), 5.52 (s, 2H), 4.40-4.36 (m, 2H), 4.34 (s, 2H), 3.69 5 3.65 (m, 2H), 3.60-3.23 (m, 4H), 3.38 (s, 3H), 2.30-2.20 (m, 2H), 2.20-2.10 (m, 2H). LCMS-ESl*: calc'd for C 21
H
27
N
6 0 4 : 427.2 (M+H*); Found: 427,2 (M+H*). Example 92: Prepared by Method LXV
NH
2 H N~ N O 0 .HCl N Example 92 10 Example 92 was obtained in 9% yield as a monohydrochloride salt. To reach complete conversion, an extra 100 equiv of MnO 2 was implemented. 'H NMR (CD 3 0D, 300 MHz)( compound exists as a mixture of two amide rotamers at 23 0C with some associated protons having distinct resonances): 6 (ppm) 7.60-7.40 (m, 4H), 5.52 (s, 2H), 4.38 (s, 2H), 15 3.80-3.25 (m, 6H), 3.08 (s, 1.5H, single rotamer), 2.93 (s, 1.5H, single rotamer), 2.25-2.10 (m, 2H), 2.10-1.95 (m, 2H), 1.47 (app. t, J = 8.4 Hz, 1H, single rotamer), 1.05 (app. t, J = 8.4 Hz, 1H, single rotamer), 0.98-0.86 (m, 1.5H, single rotamer), 0.85-0.78 (m, 1.5H, single rotamer). LCMS-ES*: calc'd for C 23
H
30
N
7 0 3 : 452.2 (M+H*); Found: 452.2 (M+H*). 20 Example 93: Prepared by Method LXV 185 WO 2010/077613 PCT/US2009/067002
NH
2 H N 0 N Example 93 Example 93 was obtained in 16% yield as a monohydrochloride salt. To reach complete conversion, an extra 100 equiv of MnO 2 was implemented. 'H NMR (CD 3 OD, 300 MHz)( 5 compound exists as a mixture of two amide rotamers at 23 *C with some associated protons having distinct resonances): S (ppm) 7.60-7.40 (m, 4H), 5.52 (s, 2H), 4.34 (s, 2H), 3.80-3.25 (m, 6H), 3.05 (s, 1.5H, single rotamer), 2.88 (s, 1.5H, single rotamer), 2.21-2.10 (m, 2H), 2.10-1.96 (m, 2H), 1.47 (app. t, J = 8.4 Hz, IH, single rotamer), 0.95 (app. t, J = 8.4 Hz, 1H, single rotamer), 0.92-0.86 (m, 1.5H, single rotamer), 0.82-0.70 (m, 1.5H, 10 single rotamer). LCMS-ESI*: caic'd for C 23
H
3 aN 7 0 3 : 452.2 (M+H*); Found: 452.2 (M+H*). Example 94: Prepared by Method LXI
NH
2 H ON 0 NN . 2HCI Example 94 15 Example 94 was obtained in 87% yield as a dihydrochloride salt. 1 H NMR (CD 3 OD, 300 MHz): 8 (ppm) 7.89 (s, 1H), 7.79-7.70 (m, 3H), 7.61-7.43 (m, 4H), 4.96 (s, 2H), 4.61 (t, J = 4.7, 2H), 4.47 (s, 2H), 4.16 (s, 2H), 3.73 (t, J = 4.7 Hz, 2H), 3.60-3.43 (m, 2H), 3.38 (s, 3H), 3.30-3.18 (m, 2H), 2.25-2.13 (m, 2H), 2.11-1.96 (m, 2H). LCMS-ESI*: calc'd for
C
27
H
33
N
6 0 3 : 489.3 (M+H*); Found: 489.2 (M+H*), 245.2 ((M+2H*)/2). 20 Example 95: Prepared by Method LXV 186 WO 2010/077613 PCT/US2009/067002
NH
2 H N 0 N j 'HCJ Example 95 Example 95 was obtained in 97% yield as a monohydrochloride salt. 'H NMR (CD 3 OD, 300 MHz): 5 (ppm) 7.80-7.46 (m, 8H), 5.53 (s, 2H), 4.46 (t, J = 4.5 Hz, 2H), 4.45 (s, 2H), 5 3.68 (t, J = 4.5 Hz, 2H), 3.58-3.42 (m, 2H), 3.36 (s, 3H), 3.35-3.21 (m, 2H), 2.28-2.10 (m, 2H), 2.10-1.99 (m, 2H). LCMS-ESi*: calc'd for C 27
H
31
N
6 0 4 : 503.2 (M+H*); Found: 503.2 (M+H*). Example 96: Prepared by Method LXI
NH-
2 H NF1 N N O N O N N 2HCI 10 Example 96 Example 96 was obtained in 87% yield as a dihydrochloride salt. 'H NMR (CD 3 0D, 300 MHz): 5 (ppm) 7.89 (s, 1H), 7.76-7.70 (m, 3H), 7.61-7.44 (m, 4H), 4.97 (s, 2H), 4.49 (t, J = 7.6 Hz, 2H), 4.47 (s, 2H), 4.17 (s, 2H), 3.58-3.51 (m, 2H), 3.31-3.19 (m, 2H), 2.23-2.11 15 (m, 2H), 2.10-1.99 (m, 2H), 1.77 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.48 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.95 (t, J = 7.6 Hz, 3H). LCMS-ESI*: calc'd for C2eH 5
N
6 0 2 : 487.3 (M+H*); Found: 487.2 (M+H*) and 244.2 ((M+2H*)/2). Example 97: Prepared by Method LXV 187 WO 2010/077613 PCT/US2009/067002
NH
2 H N N O N N:C NN HCI Example 97 Example 97 was obtained in 21% yield as a monohydrochloride salt. 1 H NMR (CD 3 OD, 300 MHz): 5 (ppm) 7.80-7.43 (m, 8H), 5.54 (s, 2H), 4.45 (s, 2H), 4.32 (t, J = 7.6 Hz, 2H), 5 3.58-3.47 (m, 2H), 3.45-3.38 (m, 2H), 2.21-1.87 (m, 4H), 1.76 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.47 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.95 (t, J = 7.6 Hz, 3H). LCMS-ESI*: calc'd for
C
2 8
H
33
N
6 0 3 : 501.3 (M+H*); Found: 501.2 (M+H*). Example 98: Prepared by Method LXI
NH
2 H N O OOXN N N . 21HCI 10 Example 98 Example 98 was obtained in quantitative yield as a dihydrochloride salt. 'H NMR
(CD
3 0D, 300 MHz): 5 (ppm) 7.77 (d, J = 7.8 Hz, 2H), 7.71 (d, J = 7.8 Hz, 2H), 7.64 (d, J = 7.8 Hz, 2H), 7.50 (d, J = 7.8 Hz, 2H), 4.97 (s, 2H), 4.62 (t, J = 4.4 Hz, 2H), 4.45 (s, 2H), 15 4.18 (s, 2H), 3.72 (t, J 4.4 Hz, 2H), 3.58-3.49 (m, 2H), 3,38 (s, 3H), 3.30-3.17 (m, 2H), 2.26-2.12 (m, 2H), 2.11-1.99 (m, 2H). LCMS-ESI 4 : calc'd for C 27
H
33
N
6 0 3 : 489.3 (M+H*); Found: 489.1 (M+H*) and 245.2 ((M+2H*)/2). Example 99: Prepared by Method LXV 188 WO 2010/077613 PCT/US2009/067002
NH
2 H N 0 .HCI Example 99 Example 99 was obtained in 20% yield as a monohydrochloride salt. 'H NMR (CD 3 0D, 300 MHz): S (ppm) 7.74 (d, J = 7.8 Hz, 2H), 7.62-7.50 (m, 6H), 5.53 (s, 2H), 4.43 (t, J = 5 4.4 Hz, 2H), 4.42 (s, 2H), 3.66 (t, J = 4.4 Hz, 2H), 3.58-3.44 (m, 2H), 3.42-3.30 (m, 2H), 2.25-2.10 (m, 2H), 2.10-1.99 (m, 2H). LCMS-ESI*: calc'd for C 27
H
3 1
N
6 0 4 : 503.2 (M+H*); Found: 503.1 (M+H t ). Example 100: Prepared by Method LXI
NH
2 H N N0 N N * 2HCI 10 Example 100 Example 100 was obtained in 86% yield as a dihydrochioride salt. 1 H NMR (CD 3 0D, 300 MHz): 6 (ppm) 7.77 (d, J = 7.8 Hz, 2H), 7.70 (d, J = 7.8 Hz, 2H), 7.64 (d, J = 7.8 Hz, 2H), 7.49 (d, J = 7.8 Hz, 2H), 4.96 (s, 2H), 4.49 (t, J = 7.6 Hz, 2H), 4.44 (s, 2H), 4.18 (s, 2H), 15 3.60-3.50 (m, 2H), 3.27-3.19 (m, 2H), 2.22-2.10 (m, 2H), 2.09-1.96 (m, 2H), 1.76 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.46 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.95 (t, J = 7.6 Hz, 3H). LCMS-ESI*: calc'd for C 28
H
35
N
6 0 2 : 487.3 (M+H*); Found: 487.1 (M+H*) and 244.2 ((M+2H*)i2). Example 101: Prepared by Method LXV 189 WO 2010/077613 PCT/US2009/067002
NH
2 N N 0 N - N t O N * HCI Example 101 Example 101 was obtained in 23% yield as a monohydrochloride salt. 1 H NMR (CD 3 0D, 300 MHz): S (ppm) 7.74 (d, J = 7.8 Hz, 2H), 7.62-7.50 (m, 6H), 5.54 (s, 2H), 4.42 (s, 2H), 5 4.29 (t, J = 7.6 Hz, 2H), 3.56-3.41 (m, 2H), 3.38-3.26 (m, 2H), 2.27-2.10 (im, 2H), 2.09 1.96 (m, 2H), 1.69 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.45 (qt, J = 7.6 Hz, 7.6 Hz, 2H), 0.96 (t, J = 7.6 Hz, 3H). LCMS-ESI': calc'd for C 28
H
33 NeO 3 : 501.3 (M+H*); Found: 503.1 (M+H*). Compound EA: Prepared by Method I 10 CN EA Compound EA was made using THF at 23 IC with a 2h reaction time. Reaction was quenched with water and chromatoraphed on an ISCO silica column (Eluent: 0 - 40%B 15 A=DCM B= MeOH/DCM 1:4). Product EA was obtained as a free base. 'H NMR (DMSO d, 300 MHz): d (ppm) 7.74-7.73- (d, J=5.1 Hz, 1H), 7.69-7.65 (m, 2H), 7.53-7.48 (m, 1H), 3.81-3.55 (m, 2H), 2.96-2.88 (m, 1H), 2.59-2.56 (m, 1H), 1.99-1.89 (m, 1H), 1.82-1.73, (m, 1H), 1.35-1.26 (m, 2H), 0.92-0.90 (d, J = 14,4 Hz, 6H). LCMS-ESIl: calc'd for
C
14 H 19
N
2 : 215.3 (M+H*); Found: 215.1 (M+H*). 20 Compound EB: Prepared by Method il 190 WO 2010/077613 PCT/US2009/067002
NH
2 N EB Compound EB was made synthesized in THF over a 100h reaction timeframe. Crude material was carried forward without further purification, and was obtained as a free base. 5 LCMS-ES*: calc'd for C 14
H
23
N
2 : 219.3 (M+H*); Found: 219.2 (M+H*). Compound EC: Prepared by Method IV >N H 0 N EC 10 Compound EC was synthesized over a 3h reaction timeframe and quenched with water. After chromatography on an ISCO silica column (Eluent: 0 - 40% B over 15 min; A=DCM, B= MeOH/DCM 1:4), EC was obtained as a free base. 'H NMR (DMSO-d", 300 MHz): d (ppm) 7.26-7,12 (m, 4H), 4.12-4.05 (m, 2H), 3.78-3.74 (d, J = 20.0 Hz, 1H), 3.68 (s, 2H), 3.62 (s, broad, 1H), 3.47-3.42 (d, J = 14.0 Hz, 1H), 3.27-3.26 (d, J = 3.6 Hz, 2H), 15 2.96-2.90 (m, 1H), 1.98-1.89 (m, 2H), 1.79-1.72 (m, 1H), 1.34-1.24 (m, 2H), 1.20-1,16 (t, J = 7.0 Hz, 3H), 0.94-0.90 (m, 6H). LCMS-ESl*: calc'd for C, 8
H
2 9
N
2 0 2 : 305.4 (M+H*); Found: 305.2 (M+H*). Compound ED: Prepared by Method LXVI
NH
2 ''
NO
2 N N0 N' 20 ED 191 WO 2010/077613 PCT/US2009/067002 Compound ED was prepared using a 3,5h reaction timeframe. The product was chromatographed on an 12 gram ISCO silica column (Eluent: 0 -+ 30% B ramp over 5 min. A= DCM B= MeOH/DCM 1:4). ED was obtained as a free base. 'H NMR (DMSO-d4 300 MHz): d (ppm) 7.97 (s, broad, 2H), 7.26-7.09 (m, 4H), 4.67 (s, 2H), 4.10-4.06 (m, 5 6H), 3.76-3.71 (d, J = 14.1 Hz, 1H), 3.61 (s, 1H), 3.44-3.39 (d, J=14.1 Hz 1H), 2.87 (s, broad, 1H), 1.94-1.88 (m, 1H), 1.70 (s, broad, 1H), 1.6-1.51 (m, 2H), 1.37-1.14 (m, 7H), 0.90-0.84 (m, 9H). LCMS-ESI*: calc'd for C 2 6
H
3 gN 6 0: 514.6 (M+H'); Found: 515.3 (M+H*). 10 Example 102: Prepared by Method XIV
NH
2 ,-- t -ON N> O N3 Example 102 Example 102 was synthesized over a 2h reaction timeframe. Example 102 was obtained as a free base. 1 H NMR (DMSO d 300 MHz): d (ppm) 11.06 (s, broad, 1H), 10.60 (s, 15 broad, 1H), 10.29 (s, broad, 1H), 7.76-7.71 (m, 4H), 4.79 (s, 2H), 4.31-4.17 (m, 4H), 4.07 4.04 (d, J = 8.7 Hz, 2H), 3.72 (m, 1H), 3.61-3.50 (m, 1H), 2.28-2.00 (m, broad, 3H), 1.71 1.53 (m, 4H), 1.36-1.16 (m, 7H), 1.13-1.04 (m, 2H), 0.85-0.80 (t, J =7.6 Hz, 3H). LCMS ESI*: calc'd for C 24
H
35
N
6 0 2 : 438.6 (M+H*); Found: 439.3 (M+H). 20 Compound EE: Prepared by Method XXXVII NC NC N-.Boc EE H NMR (CDC1 3 , 300MHz) 5 (ppm) 7.48-7.45 (m, 2H), 7.21 (d, IH, J = 8.1 Hz), 4,62 (s, 2H), 3.67 (t, J = 5.8 Hz, 2H), 2.87 (t, J = 5.5 Hz, 2H), 1.50 (s, 9H). 25 Compound EF: Prepared by Method XXXVIII 192 WO 2010/077613 PCT/US2009/067002
H
2 N EF 'H NMR (CD3OD, 300MHz) S (ppm) 7.14-7.03 (m, 3H), 4.74 (s, 2H), 3.71 (s, 2H), 3.57 (t, J = 5.7 Hz, 2H), 2.78 (t, J = 5.8 Hz, 2H), 1.48 (s, 9H). LCMS-ESI*: calc'd for C, 5
H
23
N
2 0 2 : 5 263.3 (M+H*); Found: 262.9 (M+H*). Compound EG: Prepared by Method XXXIX EtO 2 C NN EG 10 'H NMR (CDCl 3 , 300MHz) 5 (ppm) 7.18-7.07 (m, 3H), 4.56 (s, 2H), 4.24-4.17 (m, 2H), 3.81 (s, 2H), 3.66-3.64 (m, 2H), 3.43 (s, 2H), 2.83 (t, 2H, J = 6.3 Hz), 1.50 (s, 9H), 1.28 (t, J = 7.0 Hz, 3H); LCMS-ESI*: calc'd for C1 9
H
29
N
2 0 4 : 349.4 (M+H*); Found: 349.0 (M+H*). Compound EH: Prepared by Method LXVI
NH
2 N
NO)
2 O N N CO 2 Et EH 15 NBoc 'H NMR (CDCs, 300 MHz): 6 (ppm) 7.30-7.06 (m, 3H), 4.66 (s, 2H), 4.54 (s, 2H), 4.10 4.21 (m, 4H), 4.032 (s, 2H), 3.62-3.34 (m, 2H), 2.79-2.81 (m, 2H), 1.69-1.65 (m, 2H), 1.50 (s, 9H), 1.43-1.48 (m, 2H), 1.22-1.28 (m, 3H), 0.89-0.96 (m, 3H); LCMS-ESI: calc'd for 20 C 2 9
H
39 N50 7 : 559.6 (M+H'); Found: 559.0 (M+H*). Example 103: Prepared by Method XL 193 WO 2010/077613 PCT/US2009/067002
NH
2 H N 0 N N Example 103 NH Example 103 was made according to Method XL. 1 H NMR (CD 3 OD, 300 MHz): d (ppm) 7.26-7.22 (m, 3H), 4.86 (s, 2H), 4.43-4.36 (m, 4H), 4.05 (s, 2H), 3.50 (t, J = 6.4 Hz, 2H), 5 3.12 (t, J = 6.1 Hz, 2H), 1.78-1.70 (m, 2H), 1.49- 1.42 (m, 2H), 0.95 (t, J = 7.5 Hz, 3H). LCMS-ESI*: calc'd for C 20
H
27
N
6 0 2 : 383.4 (M+H*); Found: 383.1 (M+H'). Example 104: Prepared by Method XLI
NH
2 H N- O N Example 104 / 10 Example 104 was made according to Method XLI. 1 H NMR (CD 3 QD, 300 MHz): d (ppm) 7.32-7.24 (m, 3H), 4.58-4.56 (m, 2H), 4.38 (t, J = 6.5 Hz, 2H), 4.26-4.24 (m, 2H), 4.03 (s, 2H), 3.79-3.71 (m, 2H), 3.21-3.10 (m, 2H), 1.80-1.68 (m, 2H), 1.47-1.39 (m, 2H), 0.96 (t,, J = 7.4 Hz, 3H). LCMS-ES*: calc'd for C 22
H
31
N
6 0 2 : 411.5 (M+H'); Found: 411.2 (M+H*). 15 Example 105: Prepared by Method XLVIII
NH
2 H O1N N Example 105 x N Example 105 was made according to Method XLVIII. 'H NMR (CD 3 0D, 300 MHz): d (ppm) 7.29-7.26 (m, 3H), 4.46-4.35 (m, 4H), 4.02 (s, 2H), 3.76-3.72 (m, 2H), 3.23-3.21 20 (m, 2H), 1.77-1.72 (m, 2H), 1.47-1.44 (m, 8H), 0.96 (t, J = 7.0 Hz, 3H); LCMS-ESI*: caic'd for C 23
H
33
N
6 0 2 : 425.5 (M+H 4 ); Found: 425.2 (M+H*). 194 WO 2010/077613 PCT/US2009/067002 Example 106: Prepared by Method XLVIII NH2 H N N O O N N Example 106 ,r N 5 Example 106 was made according to Method XLVIII. 1 H NMR (CD 3 0D, 300 MHz): d (ppm) 7.30-7.26 (m, 3H), 4.67-4.64 (m, 11H), 4.41-4.37 (m, 3H), 4:04-4.02 (m, 2H), 3.88 3.85 (m, 1H), 3.43-3.41 (m, 1H), 3.34-3.20 (m, 4H), 1.76-1.72 (m, 2H), 1.49-1.44 (m, 2H), 1.24-1.20 (m, 1H), 0.99-0.94 (m, 3H), 0.82 (t, J = 6 Hz, 2H), 0.45 (m, 2H). LCMS-ESl*: calc'd for C 2 4
H
33
N
6 0 2 : 437.2 (M+H*); Found: 437.1 (M+H*). 10 Scheme 90 OH CBZ-CI, K 2
CO
3 OH HN DMF CBZ' FB Method XLIX: Compound FB. 2-(Piperidin-4-yl)-ethanol, (520 mg, 4 mmol) was 15 dissolved in anhydrous DMF (8 mL) and to this was added K 2
CO
3 and the mixture was stirred under N 2 in an ice bath. To this was added benzyl chloroformate (623 pL, 4.4 mmol) dropwise. The reaction was allowed to warm to room temperature and then stirred for additional 90 minutes. The reaction was diluted with EtOAc and washed with saturated NaHCO 3 (aq) (2X) followed with saturated NaCl(aq). The organic extract was 20 dried over anhydrous Na 2
SO
4 and concentrated under reduced pressure. The residue was purified with silica gel chromatography (20-80% EtOAc in hexanes) to give Compound FB (0.99 g, 3.76 mmol). 'H NMR (CDCI, 300 MHz): d (ppm) 7.36 (m, 5H), 5.13 (s, 2H), 4.18 (bs, 2H), 3.72 (m, 2H), 2.79 (m, 2H), 1.73-1.52 (m, 5H), 1.27-1.18 (m, 3H). 25 Scheme 91 195 WO 2010/077613 PCT/US2009/067002 1) S0 3 -pyr, DMSO H O OH TEA, 50C N CBZ' 2) GlyOMe-HCI, NaBH(OAc) 3 CBZ'N FC FB ACN/NMP Method XLX: Compound FC. Compound FB (989 mg, 3.76 mmol) was dissolved in anhydrous DMSO (12 mL) and stirred under N 2 at 5 *C. Triethylamine (1.3 mL, 9.4 mmol) was added followed by sulfur trioxide pyridine complex (1.5 g, 9.4 mmol). The reaction 5 was stirred at 0-5 *C for 90 minutes. Ice and EtOAc were added to the reaction, followed by stirring for several minutes. The organic layer was collected and washed with saturated NaHCO 3 (aq) (2X) followed with saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 and concentrated under reduced pressure. The resulting oil was dissolved in anhydrous acetonitrile (10 mL) and NMP (3 mL). To this was added 10 glycine methyl ester hydrochloric salt (708 mg, 5.64 mmol) followed by stirring for 15 minutes. NaBH(OAc) 3 (1.59 g, 7.52 mmol) was added and the reaction was stirred for 16 hours. Then MeOH was added and the mixture was stirred for 5 minutes. The reaction was diluted with EtOAc and washed with saturated NaHCO 3 (aq) (2X) followed with saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 , filtered, and 15 concentrated under reduced pressure. The residue was purified with silica gel chromatography (0-10% MeOH in CH 2 Cl 2 ) to give Compound FC (142mg, 0.43 mmol). Scheme 92 CI NH 2 CI 1) NH/MeOH, THF, 0 0 C N NO 2 1) N ,J NO 2 S N C 2) DIPEA OSIN" 0N H SH N N N N 0 CBZ'N F O FD N CBZ 20 Method XLXI: Compound FD. 4,6-dichloro-5-nitro-2-methylthiopyrimidine (124 mg, 0.468 mmol) was dissolved in anhydrous THF (5 mL) and stirred under N 2 (g) in an ice bath. A solution of 7 N NH 3 in MeOH (73 pL, 0.51 mmol) in THF (500 pL) was added dropwise over 2-3 minutes. The reaction was stirred for 60 minutes. Additional 7 N NH 3 in MeOH solution (73 pL, 0.51 mmol) was added and the mixture was stirred for an 25 additional 60 minutes. A solution of FC (142mg, 0.42 mmol) in anhydrous THF (0.5 mL) was added to the reaction. The DIPEA (89 pL, 0.51 mmol) was added. The reaction 196 WO 2010/077613 PCT/US2009/067002 mixture was then stirred for 16 hours at room temperature, diluted with EtOAc, and washed with saturated NaHCO 3 (aq) solution (2X) followed with saturated NaCI(aq). Ther organic extract was dried over anhydrous Na 2
SO
4 and concentrated under reduced pressure. The product was purified with silica gel chromatography (20-50% EtOAc in 5 hexanes) to give Compound FD (150 mg, 0.29 mmol). 'H NMR: (CDC 3 , 300 MHz): d (ppm) 7.36 (m, 5H), 5.13 (s, 2H), 4.12 (m, 4H), 3.76 (s, 3H), 3.41 (m, 2H), 2.76 (m, 2H), 2.42 (s, 3H), 1.67 (m, 4H), 1.45 (m, 1H), 1.20 (m, 2H). LCMS-ESI*: calc'd for
C
23
H
31
N
6 OeS: 519.2 (M+H*); Found: 519.0 (M+H t ). 10 Scheme 93
NH
2
NH
2 N NO 2 N NO 2 1) peracetic acid 11 N N O , M O N N Os o 2) nBuOH, TFA 0 FD FE N N CBZ CBZ Method XLXII: Compound FE. Compound FD (150 mg, 0.29 mmol) was dissolved in anhydrous acetonitrile (10 mL) and stirred under N 2 (g) in an ice bath. Aqueous 32% peracetic acid solution (244 pL, 1.16 mmol) was added and the mixture was stirred for 2 15 hours. Saturated Na 2
S
2 0 3 (aq) solution was added and the mixture was stirred for 5 minutes. The mixture was extracted with EtOAc. The organic extract was then washed with NaHCO 3 (aq) solution followed with saturated NaCI(aq), dried over anhydrous Na 2
SO
4 , filtered, and concentrated under reduced pressure. The residue was added to n BuOH (5 mL) and TFA (90 pL, 1.16 mmol) and then stirred at 100 *C for 2-3 hours. The 20 mixture was concentrated under reduced pressure, dissolved in EtOAc and washed with saturated NaHCO 3 (aq) solution (2X) followed with saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 and concentrated under reduced pressure. The product was purified with silica gel chromatography (20-50% EtOAc in hexanes) to give Compound FE (108 mg, 0.20 mmol). 1 H NMR (CDC 3 , 300 MHz): F 7.36 (m, 5H), 25 5.13 (s, 2H), 4.22-4.10 (m, 6H), 3.76 (s, 3H), 3.40 (m, 2H), 2.76 (m, 2H), 1.71 (m, 6H), 1.45 (m, 3H), 1.20 (m, 2H), 0.95 (t, J = 7.2Hz, 3H). LCMS-ESI*: calc'd for C 26
H
37
N
6 0 7 : 545.3 (M+H*); Found: 545.1 (M+H+), Scheme 94 197 WO 2010/077613 PCT/US2009/067002
NH
2 NH 2 H N NO 2 N N O A0 Pd/C, H 2 T O N O N 0 MeOHITHF FE Example 107 N N N H CBZ Method XLXIII: Example 107. Compound FE (108 mg, 0.20 mmol) was dissolved in THF (4 mL) and MeOH (15 mL). To this was added 10% Pd/C and the reaction was stirred under 1 atmosphere H 2 (g) for 16 hours. The reaction was filtered reaction through 5 Celite. Concentration under reduced pressure gave Example 107 (60 mg, 0.17 mmol). 'H NMR: (CDCl 3 , 300 MHz): d (ppm) 5.15 (s, 2H), 3.97 (t, J = 6.9Hz, 2H), 3.75 (s, 2H), 3.35 (m, 2H), 2.76 (m, 2H), 1.65-1.05 (m, 13H), 0.95 (t, J = 7,2Hz, 3H). LCMS-ESl*: calc'd for C 1 7
H
29
N
6 0 2 : 349.2 (M+H*); Found: 349.1 (M+H t ). 10 Scheme 95: Example 108 NH2 NH NHH BnBr ON MIPEA N NYNr DMVF OIN N T Example 107 Example 108 N N H Method XLXIV: Example 108: Example 107 (20 mg, 0.057 mmol) was dissolved in anhydrous DMF (0.5 mL). To this was added diisopropylethylamine, DIPEA, (15 pL, 0.086 mmol) and benzyl bromide (8 pL, 0.068 mmol). The reaction was stirred for 16 15 hours. Reaction was directly purified with Prep HPLC Phenomenex Gemini 5u C 16 column and eluted with a linear gradient of 5-100% Acetonitrile containing 0.1% TFA to give Example 108 (11.2mg, 0.025 mmol). 1 H NMR: (CD 3 0D, 300 MHz): d (ppm) 7.50 (s, 5H), 4.42 (t, J = 6.3Hz, 2H), 4.30 (s, 2H), 4.20 (s, 2H), 3.69 (m, 2H), 3.51 (m, 2H), 3.00 (m, 2H), 2.03 (m, 2H), 1.80-1.46 (m, 9H), 0.98 (t, J = 7.2Hz, 3H). LCMS-ES*: caic'd for 20 C 24
H
35
N
6
O
2 : 439.3 (M+H*); Found: 439.2 (M+H*). 198 WO 2010/077613 PCT/US2009/067002 Scheme 96: OH SOC' 2 CI *HCI O A DCM CF N DMFG IN Method XLXV: Compound FG: Starting with (2-methylpyridine-5-yl)-methanol (5.07 g) in
CH
2
CI
2 (50.0 mL), 4 equivs of SOC 2 (12.0 mL) were added at 23 "C. The mixture was 5 allowed to stir overnight and was then concentrated in vacuo, giving Compound FG as a monohydrochioride salt, which was used without purification. 1 H NMR: (DMSO-d 6 , 300 MHz): 5 8.84 (s, 1H), 8.44 (d, J = 6.9 Hz, 1H), 7.86 (d, J = 7.8 Hz, 1H), 4.92 (s, 2H), 2.1 (s, 3H). 10 Scheme 97 CI HCI H 2 N O- (free base) I H N Nl FG FH Method XLXVI: Compound FH. Ethyl glycinate hydrochloride (113 mg) was slurried in DMF (3.0 mL) with K 2
CO
3 (270 mg) and the crude pyridinyl chloride (FG)(110 mg). The mixture was heated to 40 *C and allowed to stir overnight. The reaction was quenched 15 by the addition of water and was diluted with EtOAc. The mixture was washed with a 5% solution of LiCI (3 x 5 ml) to remove DMF, followed by a brine wash, and the organic extracts were dried with sodium sulfate and concentrated in vacuo. Chromatography on silica using CH 2
C
2 and 20% MeOH/CH 2 Cl 2 as eluent gave rise to the desired pyridyl aminoester product (55 mg). 'H NMR: (DMSO-d 6 , 300 MHz): 8 8.42 (s, 1 H), 7.71-7.62 (m, 20 1H), 7.25 (d, J = 7.8 Hz, 1H), 5.03 (s, 2H), 4.12-4.05 (m, 2H), 3.73 (d, J = 11.7 Hz, 2H), 2.45 (s, 3H), 1.30 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 11
H
17
N
2 0 2 : 208.26 (M+H*); Found: 208.9 (M+H*). Scheme 98 199 WO 2010/077613 PCT/US2009/067002
NH
2 CI NH 3 / MeOH N NO 2 -O N THF N O N" HSIN N N FPH SN C 1 NHFJO Method XLXVL: Compound FJ. 4,6-dichloro-5-nitro-2-methylmercaptopurine (1.0715 g, 4.502 mmol) was dissolved in 25 mL THF and cooled to 0 *C. NH 3 / MeOH was added 5 (3.5 Equiv) and the mixture was allowed to stir cold for 1 h. Aminoester (1.22 g, 4.37 mmol) was then added dropwise as a solution in 10 mL THF over 10-15 minutes, and the resulting mixture was allowed to warm to room temperature. After 3 h, the reaction was quenched with the addition of water, diluted with EtOAc and the pH was adjusted to = B using solid K 2 C0 3 . The mixture was washed with water, washed with brine then dried 10 with sodium sulfate and concentrated in vacuo. The crude product was then chromatographed on silica with a CH 2
C
2 and 20% MeOH/CH 2
CI
2 gradient over 10-15 column volumes. Sometimes mixtures of 6-chloropyrimidine and 6-aminopyrimidine products are obtained (1.02 g) and are sequentially treated with excess NH 3 in MeOH in THF over 45 minutes at room temperature and rechromatographed as above to give pure 15 6-aminopyrimidine product (716 mg). LCMS-ESI*: calc'd for C1 6
H
21
N
6 0 4 S: 392.43 (M+H*); Found: 393.0 (M+H*). Scheme 99:
NH
2 N NO NH2 N N' 0 Na 2
WO
4 N NO2
H
2 0 2 , AcOH 0 O S N N O EtOH 0 o N FJ N FK N 20 Method XLXVIII: Compound FK. To a solution a suspension of the sulfide FJ (3.68 g, 8.00 mmol) in EtOH (40 mL) at 0 "C was added sodium tungstate dehydrate (792 mg, 2.40 mmol), acetic acid (4.6 mL, 80 mmol), and hydrogen peroxide (3.4 mL, -40 mmol, 35% w/w in H20) sequentially. After 3 h, additional acetic acid (4.6 mL) and hydrogen peroxide 25 (3.4 mL) were added. The reaction was maintained at 0 "C for 16 h. A saturated solution 200 WO 2010/077613 PCT/US2009/067002 of Na 2
SO
3 (50 mL) was added carefully while at 0 *C followed by CH 2 0C 2 (75 mL). The layers were separated, and the aqueous layer was extracted with CH 2
C
2 (4 x 50 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum to give FK which was used without further purification. LCMS-ES*: cal'd for 5 sulfoxide C1 6
H
2 0
N
6 0 5 S: 408.43 (M+H); Found: 409.0 (M+H). LCMS-ESl*: calc'd for sulfone C 16
H
21 N,0 6 S: 424.43 (M+H*); Found: 425.1 (M+H*). Scheme 100:
NH
2
NH
2 N 11 NO2 2-Pentanol NO2 S N N TFA 0 N N 000 0 FKO FL 10 Method XLXIX: Compound FL. To a solution of sulfone FK (1.0 g, 2.0 mmol) in racemic 2-pentanol (10 mL) was added TFA (470 pL, 6.1 mmol). The reaction was stirred at 100 OC for 1 h. The reaction mixture was poured onto a saturated solution of NaHCO 3 (20 mL) and CH 2
C
2 (30 mL). The layers were separated, and the aqueous layer was extracted 15 with CH 2
CI
2 (30 mL). The combined organic layers were dried over MgSO 4 , filtered, and concentrated under vacuum. Purification was conducted by silica gel chromatography (1 g substrate/10 g SiO 2 ) (2-15% MeOH/CH 2 l0 2 ). LCMS-ESI*: calc'd for C 2 0 H29N 6 0 5 : 432.47
(M+H
t ); Found: 433.1 (M+H*). 20 Scheme 101:
NH
2 NH 2 H N1 N NO 2 Raney Ni O ON N OIN N O MeOH/H 2 0 N N FL Example 109 Method XLXX: Example 109. To a solution of nitro compound (730 mg, 1.5 mmol) in MeOH (10 mL) was added a Raney Nickel (-200 pL, slurry in H 2 0). The reaction vessel 25 was flushed with H 2 and then stirred under an H 2 atmosphere for 1.5 h. The mixture was 201 WO 2010/077613 PCT/US2009/067002 filtered through celite with CH 2
C
2 and MeOH (1:1). The filtrate was concentrated under vacuum and left on Iyophilizer overnight. The title product was obtained as a free base. 'H NMR (DMSO-d 6 , 300 MHz): 8 9.66 (s, broad, 0.78H), 8.40 (s, IH), 7.59 (d, J = 7.8 Hz, 1H), 7.20 (d, J = 7.8 Hz, 1H), 6.18 (s, broad, 1.5H), 5.60-5.56 (m, broad, 0.78H), 4.96 5 4.85 (m, 1H), 4.61 (s, 2H), 3.82 (s, 2H), 2.42 (s, 3H), 1.53-1.04 (m, 7H), 0.83 (t, J = 7 Hz, 3H). LCMS-ESI*: calo'd for C 1 8
H
25
N
6 0 2 : 356.42 (M+H 4 ); Found: 356.9 (M+H*). Scheme 102: Prepared Via Method XLXV: OH SOC12 CI N CF 3 DCM N CF 3 FR FS 10 1 H NMR (DMSO-d, 300 MHz): 8 8.84 (s, IH), 8.17 (d, J=8.1 Hz, 1H), 7.94 (d, J=8.4 Hz, 1 H), 4.82 (s, 2H). LCMS-ESI': calc'd for C 7
H
6
CIF
3 N 195.57 (M + H'); Found: for 3 CI 195.9 (M + H') and 37 CI 197.9 (M + H*). 15 Scheme 103: Prepared Via Method XLXVI: HCI CI
H
2 N N CF 3
F
3 C N FS
'
9 F NMR (DMSO-d 6 , 282 MHz): 8 -66.69. 'H NMR (DMSO-d, 300 MHz): 8.69 (s, 1H), 8.02 (dd, J=7.8 Hz, IH), 7.85 (d, J=7.8 Hz, 1H), 4.08 (d, 2H), 3.85 (s, 2H), 2.82 (bs, IH), 20 1.15-1.19 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 11
H
1 3
F
3
N
2 0 2 262.23 (M + H*); found: 262.9 (M + H'). Scheme 104: 202 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2 NO 'N NO 2 EtN, FT N N 2 N N N O MO N OTs THF O BM F3C(N FM Method XLXXI: Compound FM. Compound FT (6.5 mg, 0.025 mmol) was dissolved in THF (1 mL) and to this was added BM (9.6 mg, 0.025 mmol). Then triethylamine (10 pL, 5 0.075 mmol) was added and the mixture was stirred for 12 hours. The mixture was added to EtOAc and washed with saturated NaHCO 3 (aq) solution followed with saturated NaCI(aq). The organic extract was dried over anhydrous Na 2
SO
4 , filtered, and concentrated under reduced pressure. The product was then purified with Prep HPLC Phenomenex Gemini 5u C 18 column and eluted with a linear gradient of 25-100% 10 Acetonitrile containing 0.1% TFA. LCMS-ESI*: calc'd for C 19
H
2 4
F
3
N
6 05- 472.42 (M+H); Found: 473.1 (M+H*). Compound FAB: Prepared by Method XLXXI
NH
2 N NO 2 OIN' N' Os 0 Boc' N H FAB 15 Compound FAB was made from commercial N-[3-(tert-butoxylcarbonylamino)propyl] glycine ethyl ester according to Method XLXXI. To a stirred solution of tosylate (BM)(648.6 mg) in 30 mL of THF was added N-[3-(tert-butoxyicarbonyiamino)propyl glycine ethyl ester (475 mg), and resulting solution became yellow within seconds. EtN 20 (500 pL) was added and the mixture was allowed to stir overnight at 23 0 C. After quenching with water, the mixture was diluted 100% with EtOAc and partitioned with saturated brine solution. The organic layer was collected, dried with sodium sulfate, and concentrated in vacuo. After chromatography on silica gel (Eluent: DCM -+ MeOH/DCM 1:4) pure FAB was obtained as a free base (852 mg) in 98% yield. 'H NMR (DMSO d6, 25 300 MHz): d (ppm): 7.98 (s, braod, 2H); 6.79 (m, broad, 1H); 4,18-4.06 (m, 6H); 3.29 (in, 2H); 2.93-2.85 (m, 2H); 1.79-1.70 (m, 2H); 1.66-1.57 (m, 2H); 1.42-1.32 (m, 11H); 1.22 (t, 203 WO 2010/077613 PCT/US2009/067002 J = 7.0 Hz, 3H); 0.90 (t, J = 7.6 Hz, 3H). LCMS-ESl*: calc'd for C 2 0
H
35 N30 7 : 471.52 (M+H*); Found: 471.1 (M+H*). Scheme 105:
NH
2
NH
2 N NO 2 N NO 2 N O TFA O N N0 O N N 0 CM O _ 0 0 BocsN
H
2 N H 5 FAB FO Method XLXXII: Compound FO. Substrate FAB (400 mg) was dissolved in DCM (25 mL) and cooled to 0 *C. TFA (2 mL) was added. After an hour at 0 *C, the reactions progress was observed to be sluggish; More TFA (1 mL) was added and the mixture 10 continued to stir in the cold bath without any additional ice being added. At 2 h, the temperature was observed to be 6.8 *C, and the mixture was observed to be 60:40 (product;starting material). The cold bath was removed and the mixture was allowed to gradually warm to 23 0C. After - 7.5 h, reaction had progressed to 95% complete according to HPLC. Water was added and the mixture allowed to stir at 23 'C overnight. 15 Mixture was neutralized to pH = 8 with saturated NaHCO 3 , and extracted with EtOAc. The organic phase was dried with sodium sulfate and concentrated to a syrup. Crude material was not purified. LCMS-ESI*: calc'd for C 15
H
27
N
6 0 5 : 371.4 (M+H*): Found: 371.1 (M+H*). 20 Scheme 106:
NH
2
NH
2 N NO 2 Benzaidehyde N NO 2 I N N'YNaBH(OAc) 3 N O DIPEA / AcOH O H2N ~ EtOH H
H
2 N HN FO FP Method XLXXIII: Compound FP. Compound FO (free base form)(200 mg) was dissolved in EtOH and treated with benzaldehyde (65 pL), DIPEA (100 pL), and 1 drop of HOAc so 204 WO 2010/077613 PCT/US2009/067002 that the mixture was at approximately pH = 5.8. After a few minutes of stirring, NaHB(OAc) 3 (344 mg, 3 equiv based on pure FO) was added, and the mixture stirred at 23 0 C overnight. After dilution with one volume of EtOAc relative to EtOH used previously, the mixture was washed with water, followed by saturated brine. The organic 5 phase was dried with sodium sulfate, filtered, and concentrated in vacuo. Flash chromatography consistently gave rise to mixtures of unreacted starting material, desired product and a double reductive amination product. Thus, multiple runs of gravity column chromatography on silica gel using 5% MeOH in DCM were needed to obtain small amounts of purified desired product FP as a free base (77.1 mg). LCMS-ESl*: calc'd for 10 C 2 2
H
3 2
N
6
O
5 : 461.53 (M+H*); Found: 461.2 (M+H). Scheme 107:
NH
2 N 0 NH 2 N NO 2 N OH N NO 2 '1XOA N N 0 HATU O XNX N N 0 DIPEAN DMF N 0 HN N N FP FQ Method XLXXIV: Compound FQ. To stirred solution of the benzyl amine FP (47 mg) in 15 DMF (3 mL) was added 2-(4-methylpiperazin-1-yl)acetic acid (21 mg) followed by HATU (51.3 mg). The mixture was stirred for a few minutes. DIPEA (100 pL) was then added and the resulting mixture was allowed to stir at 23 *C. After 45 minutes, the starting material was observed to be consumed according to HPLC analysis, and the reaction was quenched with water and diluted with EtOAc (30 mL). The mixture was washed with 20 5% w/v aq. LiCI (3 x 20mL) then washed with saturated brine. The organic phase was dried with sodium sulfate and filtered. After concentrating in vacuo the crude product was chromatographed on an ISCO silica gel column (Eluent: 0 -+ 20% B ramp over 20 minutes: A = DCM and solvent B = MeOH/DCM 1:4) to give rise to desired product FQ (60 mg) as a free base. 'H NMR (MeOH-d 4 , 300 MHz): d (ppm) 7.36-7.23 (m, 5H); 4.71 25 4.36 (m, 2H); 4.28-4.10 (m, 6H); 4.01 (s, 1H); 3.50-3.47 (m, 2H); 3.38-3.17 (m, 4H); 2.59 (app. s, broad, 8H); 2.43-2.36 (m, 3H); 2.10-1.78 (m, 2H); 1.69 (m, broad, 2H), 1.48-1.38 (m, broad, 2H), 1.31-1.22 (t, J = 7.0 Hz, 3H), 0.99-0.93 (t, J = 7.6 Hz, 3H). LCMS-ESI*: calc'd for C 29
H
45
N
8
O
6 : 601.71 (M+H*); Found: 602.3 (M+H*). 205 WO 2010/077613 PCTIUS2009/067002 Scheme 108: Example 110: Method XLXX:
NH
2 N - NO 2 NH 2 O N Raney Ni N I
H
2 O N N N MeOH/H 2 0 N N N N N_,N -Q I Example 110 Example 110 was prepared according to Method XLXX. Prep HPLC was utilized to 5 isolate desired Example 110 as a free base (Eluent: CH 3 CN / H 2 0 gradient). 'H NMR (DMSO-d, 300 MHz): d (ppm) 9.64-9.62 (d, broad, J= 6.9 Hz, 1H), 7.72-7.64 (m, broad, 1H), 7.36-7.15 (m, 5H); 6.12 (s, 2H), 4.67 (s, 1H); 4.51 (d, J = 49.8 Hz, 2H), 4.04-3.87 (m, 4H), 3.50-3.23 (m, 2H), 3.12 (s, 2H), 2.37-2.27, (d, broad, J = 30.3 Hz, 8H), 2.13 (s, 3H); 1.85 (m, 2H); 1.75-1.50 (m, broad, 4H), 1.36-1.14 (m, 2H), 0.89-0.80 (t, J = 7.6 Hz, 3H). 10 LCMS-ESI*: calc'd for C 2 7
H
41
N
8 0 3 : 525.74 (M+H 4 ); Found: 525.3 (M+H*). Scheme 109: Prepared Via Method XLXIX
NH
2
NH
2 N NO 2 Method XLXIX N) NO2N S N N 0 OIN N 00 0 FK N O The sulfoxide/sulfone mixture (FK) was advanced with Method XLXIX using to install the 15 (S)-(+)-2-pentanol side chain. LCMS-ESl*: calc'd for CH 27 N'0 5 : 418.45 (M+H*); Found: 419.1 (M+H'). Scheme 110: Example 111, Method XLXX 206 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2 N 1 N O 2 N N O NO ( 0 Method XLXX 0 N 0 N FW N Example 111 Method XLXX was used to produce the final product. 'H NMR (DMSO-d 6 , 300 MHz): 5 9.67 (s, 1H), 8.42 (s, 1H), 7.61 (d, J = 7.8 Hz, 1H), 7.20 (d J = 7.8 Hz, 1H), 6.22 (s, broad, 5 2H), 4.62 (s, 2H), 4.10-4.06 (m, 2H), 3.83 (s, 2H), 2.43 (s, 3H), 1.63-1.53 (m, 2H), 1.40 1.30 (m, 2H), 0.88 (t, J = 7 Hz, 3H). LCMS-ES I: calo'd for C 17
H
2 3
N
6 0 2 : 342.4 (M+H*); Found: 343.2 (M+H*). Scheme 111 NH2 NH 2 N H -2 H NMnO 2 NM2N O N N ON O-N N: DMSO/ H 2 0 0 N N 10 ExamplelI Example 112 N Method XLXXV: Example 112. A solution of Example 111 (10.0 mg) in DMSO (2.9 mL) was treated with H 2 0 (750 pL) followed by MnO 2 (85%, activated, from Sigma Aldrich)(126 mg) at 23 *C. After 5 h, the reaction was filtered through a 0.45 micron 15 Teflon filter cartridge. The filtrate was directly loaded onto a Teledyne isco 'gold' 5.5 gram column and flashed (Eluent: 0.05% w/v aq. HCI/CH 3 CN 95:5 - 0:100), giving Example 112 (4.7 mg, 41% yield) as a white solid in monohydrochloride form. 'H NMR (CD 3 0D, 300 MHz): S (ppm) 8.80 (s, 1H), 8.57 (d, J = 8.2 Hz, 1H), 7.88 (d, J = 8.2 Hz, IH), 5.59 (s, 2H), 4.33 (t, J = 7.6 Hz, 2H), 2.76 (s, 3H), 1.73 (tt, J = 7.6 Hz, 7.6 Hz, 2H), 1.46 (qt, J = 20 7.6 Hz, 7.6 Hz, 2H), 0.96 (t, J = 7.6 Hz, 3H). LCMS-ESI: calc'd for C 17
H
2 1
N
8 0 3 : 357.2 (M+H*); Found: 357.2 (M+H'). Example 113: Prepared by Method XLXIV 207 WO 20101077613 PCT/US20091067002
NH
2 N O Example 113 N Example 113 was prepared according to Method XLXIV: 1 H NMR (CD 3 OD, 300 MHz): d 4.45 (t, J = 6.3Hz, 2H), 4.24 (s, 2H), 3.69 (m, 4H), 3.02 (m, 4H), 2.07 (m, 2H), 1.82-1.49 (m, 9H), 1.06 (m, 1H), 1.00 (t, J = 7.2Hz, 3H), 0.78 (m, 2H), 0.44 (m, 2H). LCMS-ESI*: 5 calc'd for C21H 35
N
6 0 2 : 403.3 (M+H*); Found: 403.2 (M+H*). Scheme 112: Prepared Via Method XLXIX
NH
2 NH 2 N NO 2 Method XLXIX N NO 2 NN0 1 -Y0 S' N N O O N N1 0 N FK N FU Compound FU. The sulfoxide/sulfone mixture (FK) was advanced with Method XLXIX 10 using tetrahydrofurfurol to install the tetrahydrofurfuryl side chain, LCMS-ESI*: calc'd for
C
20
H
27
N
6 0 6 : 446.46 (M+H*); Found: 447.1 (M+H*). Scheme 113: Example 114, Method XLXX
NH
2 NH N> NO 2 N> N O O N Method XLXX I 0 NX N O N N' 0-- 0___ T1 N FU N Example 114 15 Method XLXX was used to produce the final product. 1 H NMR (DMSO-d 6 , 300 MHz): a 9.63 (s, broad,1 H), 8.41 (s, 1H), 7.55-7.62 (m, 1H), 7.19 (d, J = 8 Hz, IH), 6.25 (s, 2H), 4.62 (s, 2H), 4.24-3.96 (m, 3H), 3.83 (s, 2H), 3.77-3.69 (m, 1H), 3.66-3.58 (m, 1H), 2.43 208 WO 2010/077613 PCT/US2009/067002 (s, 3H), 1.93-1.72 (m, 3H), 1.62-1.48 (m, 1H). LCMS-ESI*: calc'd for C 1 8
H
2 3
N
6 0 3 : 370.41 (M+H*); Found: 371.0 (M+H*). Scheme 114: Prepared Via Method XLXIX
NH
2 NH 2 N NO 2 Method XLXIX N NO 2 0N N o N N O 5 FK FV The sulfoxide/sulfone mixture (FK) was advanced with Method XLXIX using tetrahydrofuran-3-methanol to install the alkoxy side chain. LCMS-ESl*: calc'd for
C
20
H
27
N
6 03: 446.46 (M+H*); Found: 447.1 (M+H*). 10 Scheme 115: Example 115, Method XLXX
NH
2 NH 2 N N 2 N ' N O A N 2 ~ Method XLXX N O3 "0 O N' O0 N- FV Example 115 Method XLXX was used to produce the final product. 1 H NMR (DMSO-d 6 , 300 MHz): 5 15 9.69 (s, broad, 1H), 8.42 (s, 1H), 7.61 (d, J = 7.8 Hz, 1H), 7.19-7.22 (d J = 7.5, 1H), 6.25 (s, broad, 2H), 4.62 (s, 2H), 4.1-3.95 (m, 4H), 3.83 (s, 2H), 3.75-3.69 (m, 3H), 3.64-3.57 (m, 2H), 3.46-3.43 (m, 2H), 2.43 (s, 3H), 2.02-1.88 (m, 2H), 1.62-1.50 (m, 2H), 1.22 (s, broad, 1H). LCMS-ESl*: calo'd for C 1 8 H23N 6
O
3 : 370.41 (M+H*); Found: 371.0 (M+H*). 20 Scheme 116: Prepared Via Method XLXIX 209 WO 2010/077613 PCT/US2009/067002
NH
2
NH
2 N NO 2 Method XLXIX - N NO 2 S N N O N N N 0 FK O FX Starting from the sulfone/sulfoxide mixture (FK), Method XLXIX was used to install the chiral 2-pentoxy side chain. LCMS-ESI*: calc'd for C 20
H
27
N
6 0 5 : 432.47 (M+H*); Found: 5 4332 (M+H*). Scheme 117: Example 116, Method XLXX
NH
2 NH 2 N 1,N 2 MN N o Method XLXXNN FX Example 116 10 Method XLXX was used to produce the final product. 'H NMR (DMSO-d, 300 MHz): 5 9.66 (s, 1H), 8.40 (s, 1H), 7.59 (d, J = 8.4 Hz, IH), 7.20 (d, J = 8.1 Hz, 1H), 6.18 (s, broad, 2H), 4.94-4.87 (m, 1H), 4.61 (s, 2H), 3.83 (s, 2H), 2.42 (s, 3H), 1.58-1.07 (m, 7H), 0.84 (t, J = 7 Hz, 3H). calc'd for C 2 0
H
2 7
N
6 0 5 : 356.42 (M+H*); Found: 357.1 (M+H'). 15 Scheme 118: Example 117, Method XLXX
NH
2 NH 2 0 Method XLXX I N -- y 'N
F
3 C N FM F 3 C N Example 117 The final compound was synthesized using Method XLXX. 'H NMR (DMSO-d 6 , 300 MHz): 6 9.70 (s, 1H), 8.73 (s, 1H), 8.01-7.98 (s, J = 7.8 Hz, 1H), 7.86 (d, J = 7.8 Hz, 1H), 20 6.25 (s, broad, 2H), 4.75 (s, 2H), 4.00 (m, 5H), 1.54-1.51 (m, 2H), 1.32-1.22 (m, 4H), 210 WO 2010/077613 PCT/US2009/067002 0.84-0.86 (t, J = 7 Hz, 3H). LCMS-ESI*: calc'd for C 17
H
20
F
3 NeO 2 : 396.37 (M+H*); Found: 397.1 (M+H*). Compound FY: Prepared by Method XLXVIl
NH
2 N
NO
2 0 F3C N N 5 3 0 FY Compound FY was prepared from FT and isolated as a free base. 1 H NMR (DMSO d 6 , 300 MHz): d (ppm) 8.71 (s, 1H), 8.53-8.41 (d, broad, J = 38.1 Hz, 1H); 8,22 (s, broad, 2H), 8.04-8.01 (d, J = 7.5Hz, 1H), 7.89-7.76 (d, J=7.5 Hz, 1H), 4.81 (s, 2H), 4.19 (s, 2H), 10 4.15-4.08 (m, 2H); 2.27 (s, 3H), 1.19-1.15 (t, J = 7.0 Hz, 3H). LCMS-ESI*: calc'd for
C
1 6 H1 8
F
3
N
6 0 4 S: 447.4 (M+H*); Found: 446.9 (M+H). Compound FZ: Prepared by Method XLXVIII
NH
2 N NO 2 N N O 000
F
3 C NFZ 15 Compound FZ was prepared from FY according to Method XLXVIll. MS-ESIF: calc'd for C1 6 1 1
F
3
N
6 0 6 S: 478.4 (M+H*); Found: 478.9 (M+H*). Scheme 119: Compound FAA Prepared Via Method XLXIX
NH
2
NH
2 N NO 2 > NO2 I Method XLXIX N 2 NS N O N 000 0 0 20 F 3 C N FZ F 3 C N FAA 211 WO 2010/077613 PCT/US2009/067002 The sulfoxide/sulfone mixture (FZ) was advanced with Method XLXIX using tetrahydropyran-4-methanol to install the alkoxy side chain of Compound FAA. LCMS ESI*: calc'd for C 20
H
27
N
6 0 6 : 446.46 (M+H*); Found: 447.1 (M+H*). Scheme 120: Example 118, Method XLXX
NH
2
NH
2 H N NO 2 N N O A N O Method XLXX 0 OIN N"r 01 N" Nr 0 O o
F
3 C N FAA F 3 C N Example 118 Method XLXX was used to produce the final product. 1 H NMR (DMSO-d 6 , 300 MHz): d 10 9.73 (s, broad, IH), 8.71 (d, J = 13.8 Hz, 1H), 8.00-7.82 (m, 2H), 6.27 (s, 2H), 5.73 (s, broad, 1H), 4.75 (s, 2H), 4.58 (m, 2H), 3.96 (s, 2H), 3.89-3.77 (m, 2H), 3.27-3.16 (m, 2H), 1.56-1.42 (m, 3H), 1.26-1.08 (m, 2H). LCMS-ESI*: calc'd for C, 9
H
22
F
3
N
6 0 3 : 438.4 (M+H*); Found: 439.0 (M+H*). 15 212 WO 2010/077613 PCT/US2009/067002 Prophetic Examples As with the examples herein described, the following compounds may be prepared using analogous synthetic methods:
NH
2
NH
2 H ' O' - : F3C "I .. N O NoN NH NNN NN It N ON
F
3 C O N N N NHN N - N NH 2 H N N N HN N NO N NTO C N
NH
2 NH N N
N
2 H N N N I' NO N N OO N N NN NN
NNH
2 H N N~r OI N NO O'N N 5H 213 WO 2010/077613 PCT/US2009/067002 General Scheme Pyrimidi nodiazepinone Derivatives
NH
2 Y -CO 2
CH
3 N NO 2 HN'Z + | Step 1 N X (R) D
(L
2
-R
2 )m
NH
2
NH
2
NO
2 NO2 N N y-CO 2
CH
2 N y-CO2 Step 2 sN N S N N-ZSe2 S N N'Z XtQ (R)n X(R )n DoD (D0
(L
2
-R
2 )m (L 2
-R
2 )m NH2 N NO 2 y-CO 2
CH
3 N NH 2 HO R' AL - C2H
N
Step 3 R N N'Z' Step4 R t I~L' N N'Z X R (R')n D
(L
2
-R
2 )m
(L
2
-R
2 )m 214 WO 2010/077613 PCT/1JS2009/067002 Prophetic Examples The following compounds may be prepared using analogous synthetic methods:
NI
1 2 0NH2 H 0 NN 1N N N V NN CrH
NH
2 H 0 0 -0N N OIN~O N C
NH
2 H0
NH
2 H 0 N
N
N 1 N NoN NH2 N NH2 0 H N NI, N "11"'N NNN '-r No NH2H 0
NH
2 HO NN NN 215 WO 2010/077613 PCT/US2009/067002
NH
2 HN N N N ONN AO J N NNQ N C H2NH2H 0 N N r kN N O~ N N
CF
3
NH-
2 H0 NI1 N N N N N O!N (O O01H 00 1 CH 5 Biological Examples PBMC Assay Protocol Assays were conducted to determine cytokine stimulation at 24 hours from human Peripheral Blood Mononuclear Cell (PMBC) using the compounds of the present invention. The assays were run in duplicate, with 8-point, half-log dilution curves. The 10 compounds of the present invention were diluted from 10 mM DMSO solution. Cell supernatants are assayed directly for IFNa and 1:10 dilution for TNFa. The assays were performed in a similar fashion as described in Bioorg. Med. Chem. Lett. 16, 4559, (2006). Specifically, cryo-preserved PBMCs were thawed and seeded 96 well plates with 750,000 cells/well in 190pLiweIl cell media. The PBMCs were then incubated for 1 hour at 37*C at 15 5% C02. Then, the compounds of the present invention were added in 10pL cell media 216 WO 2010/077613 PCT/US2009/067002 at 8 point, half-log dilution titration. The plates were incubated at 370C and 5% C02 for 24 hours and then spinned at 1200rpm for 10min, which was followed by collecting supernatant and storing the same at -80*C. Cytokine secretion was assayed with Luminex and Upstate multi-plex kits, using a Luminex analysis instrument. The IFN-a 5 MEC value for a compound was the lowest concentration at which the compound stimulated IFN-a production at least 3-fold over the background as determined using the assay method above. The compounds of the present invention have IFN-a MEC values (pM) in the range of > 0.03 M or = 0.03 [M. In one embodiment, the compounds of the present 10 invention have IFN MEC values of < 0.01 pM. Table 1 shows IFN MEC values for the compounds disclosed in Examples 1-118 of the present application. Table I Example IFN MEC 1 > 0.03 2 = 0.03 3 > 0.03 4 = 0.03 5 > 0.03 6 > 0.03 7 > 0.03 8 > 0.03 9 =0.03 10 > 0.03 11 > 0.03 12 > 0.03 13 > 0.03 14 > 0.03 15 >0.03 16 > 0.03 17 > 0.03 18 >0,03 19 > 0.03 20 > 0.03 21 = 0.03 22 > 0.03 23 >0,03 24 = 0.03 25 = 0.03 26 > 0.03 27 > 0.03 28 > 0.03 29 > 0.03 30 =0.03 31 =0.03 32 > 0.03 217 WO 2010/077613 PCT/US2009/067002 Example IFN MEC 33 > 0.03 34 > 0.03 35 >0.03 36 > 0.03 37 =0.03 38 =0.03 39 =0.03 40 =0.03 41 =0.03 42 > 0.03 43 =0.03 44 > 0.03 45 > 0.03 46 >0.03 47 > 0.03 48 = 0.03 49 =0.03 50 >0.03 51 = 0.03 52 =0,03 53 > 0.03 54 > 0.03 55 = 0.03 56 =0.03 57 >0.03 58 > 0.03 59 = 0.03 60 > 0.03 61 = 0.03 62 > 0.03 63 > 0.03 64 > 0.03 65 = 0.03 66 > 0.03 67 > 0.03 68 =0.03 69 > 0.03 70 = 0.03 71 = 0.03 72 =0.03 73 > 0.03 74 > 0.03 75 >0.03 76 >0.03 77 >0.03 78 > 0.03 79 =0.03 80 >0.03 81 > 0.03 218 WO 2010/077613 PCT/US2009/067002 Example IFN MEC 82 = 0.03 83 = 0.03 84 = 0.03 85 > 0.03 86 = 0.03 87 =0.03 88 = 0.03 89 =0.03 90 >0.03 91 > 0.03 92 >0.03 93 =0.03 94 = 0.03 95 = 0.03 96 = 0.03 97 =0.03 98 = 0.03 99 = 0.03 100 = 0.03 101 > 0.03 102 = 0.03 103 = 0.03 104 = 0.03 105 =0.03 106 > 0.03 108 >-0.03 109 > 0.03 110 = 0.03 111 > 0.03 112 > 0.03 113 = 0.03 114 > 0.03 115 > 0.03 116 > 0.03 117 > 0.03 118 > 0.03 The specific pharmacological responses observed may vary according to and depending on the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of administration 5 employed, and such expected variations or differences in the results are contemplated in accordance with practice of the present invention. Suppression of HCV replicons by exudates of primary leukocytes treated with these compounds can then be measured by the procedure of Thomas, et al. (Antimicrob. Agents Chemother. 2007, 51, 2969-2978), which is herein incorporated by reference. Alternatively, 219 WO 2010/077613 PCT/US2009/067002 the effectiveness of these compounds for suppressing HCV replicons in the presence of PBMCs and pDCs can be determined by the procedure of Goldchild, et aL. (J. Biomol. Screen. 2009, 14, 723-730), which is herein incorporated by reference. The compounds of Formula la, 11, or Ila may also be tested for their ability to 5 induce expression of immunomodulatory cytokines Cynomolgus monkeys (Example B3), mice (Example B4) and healthy woodchucks (Example B5). Moreover, as described in Example B6, the compounds of Formula [a, 11, or Ila may also be tested for their ability to cause seroconversion against Woodchuck Hepatitis Virus (WHV) in chronically infected Eastern Woodchucks (Marmota monax) which is an art-recognized model system for 10 HBV infection in human beings (see, e.g., Tennant, B. C., Animal models of hepatitis B virus infection, Clin. Liver Dis. 3:241-266 (1999); Menne, S., and P. J. Cote, The woodchuck as an animal model for pathogenesis and therapy of chronic hepatitis B virus infection, World J.Gastroenterol. 13:104-124 (2007); and Korba BE, et a., Treatment of chronic WHV infection in the Eastern woodchuck (M. monax) with nucleoside analogues 15 is predictive of therapy for chronic hepatitis B virus infection in man, Hepatology, 31: 1165-1175 (2000)). Example B3: Induction of Interferon Alpha by Compounds in Cynomolqus Monkeys A dose of a compound of Formula Il is administered orally or iv to cynomolgus 20 monkeys (3 or more animals per dose group) and serum is collected at 4 hours and 8 hours after dosing. Serum samples are analyzed for levels of interferon-alpha by ELISA. Prior to dosing, serum interferon-alpha levels are usually near or below the level of detection in each animal. The limit of quantitation (LOQ) for IFN-a based on cynomolgus monkey IFN-a standard is about 625 pg/mL. 25 Additionally, multiple doses of a compound may be administered to Cynomolgus monkeys, and the concentrations of interferon alpha were measured. Example B4: Induction of Cytokines by Compounds in Mice A compound of Formula il may be dosed once or more per day for 14 days usually by 30 oral gavage, at 0.5 mg/kg or 2.5 mg/kg, in CD-1 mice. Mouse serum samples are collected at day 1 and day 14, and serum cytokine levels are determined using the following method. Samples are thawed on ice and diluted 2 fold in assay diluent. The assay for interferon-a is done by ELISA (VeriKineTM Mouse Interferon Alpha (Mu-IFN-a) ELISA Kit, Product Number: 42100-1, PBL Biomedical Laboratories, New Brunswick, 35 New Jersey) and the other serum cytokines are assayed with Luminex and Millipiex bead 220 WO 2010/077613 PCT/US2009/067002 kits. Cytokine levels are determined using a nonlinear five point parameter curve for interpolation of data using the fit = (A+((B-A)/(1+(((B-E)/(E-A))*((x/C)^D))))). Example B5: Induction of Cytokines by Compounds in Healthy Woodchucks 5 A compound of Formula I may be administered orally to adult, WHV-negative woodchucks at one or more different doses. Three male woodchucks receive a compound of Formula I at about 0.1 to about 0.05 mg/kg and three other male woodchucks at higher doses. Whole blood samples (4 mls) are collected from each woodchuck prior to dosing at TO, and then at 4, 8, 12, and 24 hours post-dose using 10 EDTA-containing collection tubes. The induction of an immune response in woodchucks following administration of a compound are determined by measuring the mRNA expression of cytokines and interferon-inducible genes in whole blood samples collected at different time points. Total RNA is isolated using the QlAamp RNA Blood Mini Kit (Qiagen) according to the 15 manufacturer's specifications. RNA is eluted into 40 p nuclease-free water and stored at 70 0 C. The concentration of RNA is determined spectrophotometrically at OD 260 nm. Two pg of RNA are treated with DNase I (Invitrogen) and reverse transcribed to cDNA with MultiScribe Reverse Transcriptase (Applied Biosystems) using random hexamers. Triplicates of 2 pl cDNA were amplified by real time PCR on an ABI PRISM 7000 20 Sequence Detection instrument (Applied Biosystems) using SYBR GREEN Master Mix (Applied Biosystems) and woodchuck-specific primers. Amplified target genes include IFN-a, IFN-y, TNF-a, IL-2, IL-6, IL-10 IL-12, 2'5'-OAS, IDO, and MxA. Woodchuck B-actin mRNA expression is used to normalize target gene expression. Transcription levels of woodchuck cytokines and interferon-inducible genes are represented by the formula 25 2ACt, where ACt indicates the difference in the threshold cycle between P-actin and target gene expression. Results may be further represented as a fold-change from the transcription level at TO. Example B6: Seroconversion in Woodchucks Chronically Infected with Woodchuck 30 Hepatitis Virus (WHV) A compound of Formula 11 or placebo is administered orally to five woodchucks per group that are chronic carriers of woodchuck hepatitis virus (WHV). The compound may be administered at a dose of about 1 to abouto.5 mg/kg/day for 28 days. Blood samples are collected prior to dosing and multiple times during and after the 28 day 35 dosing period. Antiviral activity of the compound is assessed by comparing the serum 221 WO 2010/077613 PCT/US2009/067002 WHV DNA of treated WHV carrier woodchucks with control WHV carrier woodchucks receiving vehicle. The ability of the compound to cause seroconversion in chronically infected animals is assessed by comparing the serum antibody levels against the woodchuck hepatitis virus surface antigen (anti-WHsAg) in infected animals to the anti 5 WHsAg antibody levels in placebo treated animals. The woodchucks used in this study are bom to WHV-negative females and reared in environmentally controlled laboratory animal facilities. Woodchucks are inoculated at 3 days of age with 5 million woodchuck infectious doses of a standardized WHV inoculum (cWHV7P1 or WHV7P2). Woodchucks selected for use develope WHV surface antigen 10 (WHsAg) serum antigenemia and became chronic WHV carriers. The chronic carrier status of these woodchucks is confirmed prior to initiation of drug treatment. Serum WHV DNA concentrations are measured before treatment, during treatment, and during the post-treatment follow-up period at frequent intervals. WHV viremia in serum samples is assessed by dot blot hybridization using three replicate 15 volumes (10 pl) of undiluted serum (sensitivity, 1.0 x 10 WHV genome equivalents per ml [WHVge/ml]) compared with a standard dilution series of WHV recombinant DNA plasmid (pWHV8). Levels of Woodchuck Hepatitis Virus surface antigen (WHsAg) and antibodies to WHsAg (anti-WHs) are determined before treatment, during treatment, and during the 20 post-treatment follow-up period at frequent intervals, using WHV-specific enzyme immunoassays. Antiviral activity of a compound of Formula If is assessed by comparing the serum WHV DNA and the hepatic WHV nucleic acids of treated WHV carrier woodchucks with control WHV carrier woodchucks receiving vehicle. 25 Immune stimulatory activity of a compound required to cause seroconversion is assessed by comparing the serum levels of WHsAg and antibodies to WHsAg (anti WHsAg). Although specific embodiments of the present invention are herein illustrated and described in detail, the invention is not limited thereto. The above detailed descriptions are 30 provided as exemplary of the present invention and should not be construed as constituting any limitation of the invention. Modifications will be obvious to those skilled in the art, and all modifications that do not depart from the spirit of the invention are intended to be included with the scope of the appended claims. 222 H:\dar\Intenvoven\NRPortbl\DCC\DAR\7909048-I.docx- 1/06/2015 Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. 5 The reference in this specification to any prior publication (or information derived from it), orto any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates. 10 -223-

Claims (9)

1. A compound selected from the group consisting of: NH2 NO 2 N'No? N N CO2Et NNAN NH 2 MeO2S- N N' CO Et NH2 NO 2 MeS' N' N ACO2Et N NH N A N0 2 e N CO 2 Et NHN NN N2 MeO 2 S-*N N CO 2 Et - 224 - NH 2 ANO,, N MeS N N' C C 2 Et NH 2 N NO, N N CO 2 E N ,L N O NH A,, o NO MeS N 'NGCOEt andN N< NH 2 , "NO 2 MeS' N "N O 2 Et N A25 N and ,N > ora salt thereof.
2. The compound of claims 1, selected from the group consisting of: NN' -225 - NH 2 N02 N MeO 2 S' N N" CO 2 Et N AK. N0 2 MeSA NN N N CO:Et and or a salt thereof.
3, The compound of claim 1, selected from the group consisting of: NH 2 N NO -51'N N COGEt N NH 2 NO 0k.N N "C02Et NH 2 N 0 2 NN~ NXN tA~ CoEt N and > A~ or a salt thereof, - 226- H:Worlagtev.o:CNRPafCind t6tCDA' 920 1cx6:i 'j5YN!<5
4. The compound of claim 1, selected from the group consisting of: NH, N NO 2 MeO 2 S' N N CO 2 Et N NH 2 N' ,NO 2 NO MeS< N N NCO Et N> N> N> NH 2 N' N0 MeO 2 S N N COEt > NH 2 N0 N' ' > NO MeS' N<'N NN C 2 Et NH NH2 N NO 2 MeOS N N CNO2Et N N N NH N XeS N" 'N and N or a salt thereof. 227 -
5. The compound of claim 1, having the structure: NH 2 NO O N N 'CO 2 Et or a salt thereof.
6. A compound selected from the group consisting of: N' N H 0 and or a salt thereof.
7. The compound of claim 6, having the structure: N N.. or a salt thereof.
8. The compound of claim 6, having the structure 0 Ax 2 9< OH -228 -
9. A compound selected from the group consisting of: NH 2 NO 2 N HH, NH N'CO NH 2 N and "0" N 'OH or a salt or ester thereof. -229-
AU2015203139A 2008-12-09 2015-06-12 Modulators of toll-like receptors Active AU2015203139B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2015203139A AU2015203139B2 (en) 2008-12-09 2015-06-12 Modulators of toll-like receptors

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
US12106108P 2008-12-09 2008-12-09
US61/121,061 2008-12-09
US17040409P 2009-04-17 2009-04-17
US61/170,404 2009-04-17
US22438609P 2009-07-09 2009-07-09
US61/224,386 2009-07-09
US22737809P 2009-07-21 2009-07-21
US61/227,378 2009-07-21
US24263509P 2009-09-15 2009-09-15
US61/242,635 2009-09-15
PCT/US2009/067002 WO2010077613A1 (en) 2008-12-09 2009-12-07 Modulators of toll-like receptors
AU2009333559A AU2009333559B2 (en) 2008-12-09 2009-12-07 Modulators of toll-like receptors
AU2015203139A AU2015203139B2 (en) 2008-12-09 2015-06-12 Modulators of toll-like receptors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2009333559A Division AU2009333559B2 (en) 2008-12-09 2009-12-07 Modulators of toll-like receptors

Publications (2)

Publication Number Publication Date
AU2015203139A1 true AU2015203139A1 (en) 2015-07-09
AU2015203139B2 AU2015203139B2 (en) 2017-08-31

Family

ID=53612099

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2015203139A Active AU2015203139B2 (en) 2008-12-09 2015-06-12 Modulators of toll-like receptors

Country Status (1)

Country Link
AU (1) AU2015203139B2 (en)

Also Published As

Publication number Publication date
AU2015203139B2 (en) 2017-08-31

Similar Documents

Publication Publication Date Title
US11110091B2 (en) Modulators of toll-like receptors
AU2010310813B2 (en) Derivatives of purine or deazapurine useful for the treatment of (inter alia) viral infections
AU2015203139B2 (en) Modulators of toll-like receptors
OA16225A (en) Modulators of toll-like receptors.

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)