Title of Invention

3, 5-DISUBSTITUTED AND 3,5,7-TRISUBSTITUTED-3H-OXAZOLO AND 3H-THIAZOLO [4,5-D] PYRIMIDIN-2-ONE COMPOUNDS AND PRODRUGS THEREOF

Abstract The invention is directed to 3,5-disubstituted and 3,5,7-trisubstituted-3-H-oxazolo and 3H-thiazolo[4,5-d]pyrimidin-2-one compounds and prodrugs thereof that have immunomodulatory activity. The invention is also directed to the therapeutic or prophylactic use of such compounds and pharmaceutical compositions containing them, and to methods of treating diseases and disorders described herein, by administering effective amounts of such compounds and prodrugs.
Full Text WO 2006/066080 PCT/US2005/045589
3,5-Disubstituted and 3,5,7-Trisubstituted-3H-Oxazolo and 3H-
ThiazoIo[4,5-d]pyrimidin-2-one Compounds and Prodrugs Thereof
FIELD OF THE INVENTION
The invention is directed to 3,5-disubstituted and 3,5 J-trisubstituted-3H-
oxazolo and 3F-thiazolo[4,5-d]pyrimidin-2-one compounds and prodrags
thereof that have immunomodulatory activity. The invention is also directed to
the therapeutic or prophylactic use of such compounds and pharmaceutical
compositions containing them, and to methods of treating diseases and disorders
described herein, by administering effective amounts of such compounds and
prodrugs.
BACKGROUND OF THE INVENTION
The last few decades have seen significant efforts expended in exploring
possible therapeutic uses of guanine analogs and nucleosides thereof. A number
of nucleoside analogs are currently being marketed as antiviral drugs, including
HIV reverse transcriptase inhibitors such as AZT, ddl, ddC, d4T, 3TC and the
guanosine nucleoside analog abacavir. While not adhering to a particular theory,
nucleoside analogs may provide benefits by directly inhibiting the pathogen or
tumor, by stimulation of host immune functions, or some combination of these
or other mechanisms.
One of the studied guanosine analogs with demonstrated
immunomodulatory activity is 5-amino-3-(P-D-ribofuranosylthiazolo[4,5-
d]pyrirnidine-2,7(3H, 6H) dione (7-thia-8-oxoguanosine). For example, certain
pyrimido[4,5-d]pyrimidine nucleosides are disclosed in U.S. Patent No.
5,041,542 to Robins et al. as being effective in treatment against L1210 in BDF1
mice. It addition, 3-P-D-ribofuranosylthiazolo[4,5-d]pyrimidihes demonstrating
significant immunoactivity, including murine spleen cell proliferation and in
vivo activity against Semliki Forest virus, are disclosed U.S. Patent Nos.
5,041,426 and 4,880,784 to Robins et al. A number of publications have also
described non-glycosyl derivatives of the thiazolo[4,5-d]pyrimidine moiety.
See, e.g., U.S. Patent Nos. 5,994,321 and 5,446,045; Revankar et ah, I Het
Chem., 30,1341-49 (1993); Lewis et al, I Het. Chem., 32,547-56 (1995).

WO 2006/066080 PCT/US2005/045589
SUMMARY OF THE INVENTION
The present invention describes novel 3,5-disubstituted and 3,5,7-
trisubstituted-3H-oxazolo and 3H-miazolo[4,5-d]pyrimidin-2-one compounds,
pharmaceutically active prodrugs, pharmaceutically active metabolites,
pharmaceutically acceptable salts, and phannaceutically acceptable solvates
thereof, which are useful as unmunomodulators.
In another embodiment, the present invention encompasses a method of
treating or preventing a hepatitis C virus infection in a patient in need thereof
comprising administering to the patient a therapeutically or prophylactically
effective amount of a 3,5-disubstituted and 3,5,7-trisubstituted-3H-oxazolo and 3H-
thiazolo[4,5-d]pyrimidin-2-one compound or a prodrug thereof.
In a general aspect, the invention relates to prodrugs that are 3,5-
disubstituted and 3,5,7-trisubstituted-3H-oxazolo and 3H-thiazolo[4,5-
rfJpyrimidin-2-one compounds of Formula I

wherein
X is O or S,
Y is O or S,
R1 is H, alkyl, aryl, cycloalkyl, or heterocyclyl,
R2 is NH2, -NHC(O)R4, -NHR5, -NHNR6R7,
R3 is H,Cl,Br, or OR8,
R4 is -C1-C7-alkyl or -O(C1-C7-alkyl),
R5 is -C1-C7-aIkyl,
R6 and R7 are independently -C1-C7-alkyl or together with nitrogen form a 5- or 6-
membered heterocyclic ring,
R8 is-CHR9 R10,
R9 is H, -C1-C7-alkyl, cycloalkyl, aryl, heterocyclyl, -NR11R12, or OR5,
R10 is -C1-C7-alkyl, cycloalkyl, aryl, heterocyclyl, -NR11R12, or OR5,
R11 and R12 are independently H, -C1-C7-alkyl, or -C(O)R4,
2

WO 2006/066080 PCT/US2005/045589
wherein when X is O, Y is S, and R3 is H, Cl, Br, or OR8, R1 is not H or (5-D-ribose
or esters thereof,
wherein the above alkyl, aryl, cycloalkyl, or heterocyclyl moieties are optionally
substituted by 1-4 substituents selected from
hydrogen,
alkanoyl,
alkylamine,
amino,
aryl, cycloalkyl, heterocyclyl,
azido,
C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 alkoxy, C1-C6
alkylamine, C1-C6 dialkylamine, C2-C6 alkenyl, or C2-C6 alkynyl, wherein
each of which may be interrupted by one or more hetero atoms,
carboxyl,
cyano,
halo,
hydroxy,
mercapto,
nitro,
thioalkyl,
-N=N-NH2,
-C(O)2-(C1-C6 alkyl), -C(O)2-(aryl), -C(O)2-(cycloalkyl), -C(O)2-
(heterocydyl), -O-(C1-C6 haloalkyl), -O-(C1-C6 alkyl)aryl, -O-(C1-C6
alkyl)cycloalkyl, -O-(C1-C6 alkyl)heterocyclyl, -O-(C1-C6 alkyl)amino, -O-
(C1-C6 alkyl)alkylamino, -O-(C1-C6 alkyl)dialkylamino, -O-(C1-C6 alkyl)-
C(O)-amino, -O-(C1-C6 alkyl)-C(O)-alkylamino, -O-(C1-C6 alkyl)-S(O)2-
amino, -O-(C1-C6 alkyl)- S(O)2-alkylamino, -O-(C1-C6 alkyl)- S(O)2-
dialkylamino, -O-(C1-C6 alkyl)-C(O)-dialkylamino, -O-aryl, -O-heterocyclyl,
-NHCC(O)-C1-C6 alkyl), -NHC(O)-(C1-C6 alkenyl), -NHC(O)-(aryl),
-NHC(O)-(cycloalkyl), -NHC(0)-(heterocyclyl), -NHC(O)-(C1-C6
alkyl)aryl, -NHCX(O)-C1-C6 alkyl)cycloalkyl, -NHC(O)-(C1-C6
alkyl)heterocyclyl, -NHC(O)-C1-C6 alkyl)amino, -NHC(O)-(C1-C6
alkyl)alkylamine, -NHC(O)-(C1-C6 alkyl)dialkylamine, -NHC(0)-(C1-C6
3

WO 2006/066080 PCT/US2005/045589
alkyl)C(0)amino, -NHC(O)-(C1-C6 alkyl)C(O)alkylamine, -NHC(O)-(C1-C6
alkyl)C(O)diaIkylamine, -NHC(O)-(C1-C6 alkyl)N(H)-(Ci-C6 alkyl)C(O)2-
(C1-C6 alkyl), -NH-(C1-C6 alkyl)-C(O)-amino, -NH-(C1-C6 alkyl)-C(O)-
alkylamino, -NH-(C1-C6 alkyl)-C(O)-dialkylamino, -NHC(O)-(C1-C6
alkySC(O)2 C1-C6 alkyl), -NHC(O)-(C1-C6 alkyl)-S-(heterocyclyl),
-NHS(O)2(C1-C6 alkyl), -NHS(O)2-(aryl), -NH-(C1-C6 alkyl)- S(O)2-
amino, -NH-(C1-C6 alkyl)- S(O)2-alkylamino, -NH-(C1-C6 alkyl)- S(O)2-
dialkylamino, -NHS(O)2-(cycloalkyl), -NHS(O)Hheterocyclyl),
-NHS(O)(C1-C6 alkyl), -NHS(O)(aryl), -NHS(O)(cycloalkyl),
-NHS(O)(heterocyclyl), -NHS(C1-C6 alkyl), -NHS(aryl),
-NHS(cycloalkyl), and -NH-S-(heterocyclyl),
wherein each of the above substituents can be further optionally substituted
by 1-5 substituents selected from
amino,
C1-C6 alkylamine, C1-C6 dialkylamine,
C1-C6 alkyl, C1-C6 alkoxy, C1-C6 alkenyl, C1-C6 hydroxyl, and C1-C6
hydroxyalkyl, each optionally substituted by
cyano,
halo, and
nitro,
or a pharmaceutically acceptable salt, hydrate, or stereoisomer thereof.
In one embodiment, the invention relates to compounds of Formula I,
wherein R2 is NH2-
In another embodiment, the invention relates to compounds of Formula I,
wherein R3 is H.
In another embodiment, the invention relates to compounds of Formula I,
wherein X is O and Y is S.
In another embodiment, the invention relates to compounds of Formula I,
wherein R1 is selected from
4

5
WO 2006/066080 PCT/US2005/045589


6
WO 2006/066080 PCT/US2005/045589


7
WO 2006/066080 PCT/US2005/045589


8
WO 2006/066080 PCT/US2005/045589


9
WO 2006/066080 PCT/US2005/045589



10
WO 2006/066080 PCT/US2005/045589

WO 2006/066080 PCT/US2005/045589

wherein
X is O or 3,
Y is O or S,
Z is O or CH2,
R2 is -NH2, -NHC(O)R4, -NHR5, -N=CHNR6R7,
R4 is -C1-C7-alkyl or -O(C1-C7-alkyl),
R5 is -C1-C7-alkyl,
R6 and R7 are independently -C1-C7-alkyl or together with nitrogen form a 5- or 6-
membered heterocyclic ring,
R13 is OH or SH,
R14 is H, -CH2OH, or -CH2-O-C(O)C1-C18alkyl,
R1S is OH, alkenyl, -OC(O)C1-C18 alkyl, -OC(O)aryl, or -OC(0)heterocyclyl,
R16, R17, R18, and R19 are independently H, halo, N3, alkyl, -(CH2)mOR20,
-(CH2)mOC(O)C1-C18 alkyl, -OC(O)aryl, -OS(O)2aryl, or R16 and R17 are an alkenyl, or
R17 and R19 combine together to form a dioxole ring,
R20 is H or alkyl,
m is 0 or 1,
n is 1 or 2,
wherein if R2 is NH2, then one of the following must be present:
Z is CH2;
either n is 2 or m is 1;
at least one of R16, R17, R18, and R19 is halo, N3, alkyl, or -(CH2)mOR20 wherein m is
1, and wherein if R17 is N3, then R18 and R19 are not H, and wherein if R17 is OH and
R16 and R19 are H, then R18 is not F; or
R16 and R17 are an alkenyl,
11

WO 2006/066080 PCT/US2005/045589
wherein the above alkyl, aryl, cycloalkyl, or heterocyclyl moieties are optionally
substituted by 1-4 substituents selected from
hydrogen,
alkanoyl,
alkylamine,
amino,
aryl, cycloalkyl, heterocyclyl,
azido,
C1-C7 alkyl, C1-C7 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 alkoxy, C1-C6
alkylamine, C1-C6 dialkylamine, C2-C6 alkenyl, or C2-C6 alkynyl, wherein
each of which may be interrupted by one or more hetero atoms,
carboxyl,
cyano,
halo,
hydroxy,
mercapto,
nitro,
thioalkyl,
-N=N-NH2,
-C(O)2-(C1-C6 alkyl), -C(O)2-(aryl), -C(O)2-(cycloalkyl), -C(O)2-
(heterocyclyl), -O-(C1-C6 haloalkyl), -O-(C1-C6 alkyl)aryl, -O-(C1-C6
alkyl)cycloalkyl, -O-(C1-C6 alkyl)heterocyclyl, -O-(C1-C6 alkyl)amino, -O-
(C1-C6 alkyl)alkylamino, -O-(C1-C6; alkyl)dialkylamino, -O-(C1-C6 alkyl)-
C(O)-amino, -O-(C1-C6 alkyl)-C(O)-alkylamino, -O-(C1-C6 alkyl)-S(O)2-
amino, -O-(C1-C6 alkyl)- S(O)2-alkylamino, -O-(C1-C6 alkyl)- S(O)2-
dialkylamino, -O-(C1-C6 alkyl)-C(0)-dialkylamino, -O-aryl, -O-heterocyclyl,
-NHC(O)-(C1-C6 alkyl), -NHC(O)-(C1-C6 alkenyl), -NHC(O)-(aryD,
-NHC(OHcycloalkyl), -NHC(OHheterocyclyl), -NHC(O)-(C1-C6
alkyl)aryl,-NHC(O)-(C1-C6 alkyl)cycloalkyl,-NHC(O)-(C1-C6
alkyl)heterocyclyl, -NHC(O)-(C1-C6 alkyl)amino, -NHC(O)-(C1-C6
alkyl)alkylamine, -NHC(O)-(C1-C6 alkyl)dialkylamine, -NHC(O)-(C1-C6
alkyl)C(O)amino, -NHC(O)-(C1-C6 alkyl)C(O)aIkylamine, -NHC(O)-(C1-C6
alkyl)C(O)dialkylamine, -NHC(O)-(C1-C6 alkyl)N(H)-(C1-C6alkyl)C(O)2-
(C1-C6 alkyl), -NH-(C1-C6 alkyl)-C(0)-amino, -NH-C(C1-C6 alkyl)-C(O)-
12


WO 2006/066080 PCT/US2005/045589
alkylamino, -NH-(C1-C6 aIkyl)-C(O)-dialkylamino, -NHC(0)-(C1-C6
alkyI)S(O)2(C1-C6 alkyl), -NHC(O)(C1-C6 alkyl)-S-(heterocyclyl),
-NHS(O)2-(C1-C6 alkyl), -NHS(O)2-(aryl), ~NH-(C1-C6 alkyl)- S(O)2-
araino, -NH-(C1-C6 alkyl)- S(O)2-alkylamino, -NH-(C1-C6 alkyl)- S(O)2-
dialkylamino, -NHS(O)2-(cycloalkyl), -NHS(O)2-(heterocyclyl),
-NHS(O)(C1-C6 alkyl), -NHS(O)(aryl), -NHS(O)(cycloalkyl), .
-NHS(O)(heterocyclyl), -lSfHS(C1-C6 alkyl), -NHS(aryl),
-NHS(cycloalkyl), and -NH-S-(heterocydyl),
wherein each of the above substituents can be further optionally substituted
by 1-5 substituents selected from
amino,
C1-C6 alkylamine, C1-C6 dialkylamine,
C1-C6alkyl, C1-C6 alkoxy, C1-C6 alkenyl, C1-C6 hydroxyl, and C1-C6
hydroxyalkyl, each optionally substituted by
cyano,
halo, and
nitro,
or a pharmaceutically acceptable salt, hydrate, or stereoisomer thereof.
In another embodiment, the invention relates to compounds of Formula II,
wherein R2 is NH2.
In another embodiment, the invention relates to compounds of Formula II,
wherein R13 is OH.
In another embodiment, the invention relates to compounds of Formula II,
wherein X is O and Y is S.
13
In another embodiment, the invention relates to compounds of Formula II
selected from




14
WO 2006/066080 PCT/US2005/045589

WO 2006/066080 PCT/US2005/045589

In a another general aspect, the invention relates to 3,5-disubstituted and
3,5,7-trisubstituted-3#-oxazolo and 3H-thiazolo[4,5-d]pyrimidin-2-one compounds
selected from
15



16
WO 2006/066080 PCT/US2005/045589

WO 2006/066080 PCT/US2005/045589
The invention is also directed to pharmaceutically active metabolites,
phannaceutically acceptable salts, and pharmaceutically acceptable solvates of the
compounds or metabolites of the Formula I prodrugs, Formula II compounds, and
other compounds of the invention. Advantageous methods of making the
compounds of the invention are also described.
The Formula I prodrugs, Formula II compounds, and other compounds of
the invention are useful as immune system enhancers and have certain immune
system properties including modulation, mitogenicity, augmentation, and/or
potentiation or they are intermediates for compounds that have these properties.
The compounds are expected to express effects on at least the natural killer,
macrophages, dendritic or lymphocyte cells of the immune system of a host.
Because of these properties they are useful as antiviral and antitumor agents or
as intermediates for antiviral and antitumor agents. They can be used to treat an
affected host by serving as the active ingredients of suitable pharmaceutical
compositions.
In one aspect of the invention, Formula I prodrugs, Formula II compounds,
and other compounds of the invention are utilized to treat the full range of viral
diseases in mammals, including humans, by administering to the mammal a
therapeutically effective amount of the compounds. Viral diseases contemplated to
be treated with compounds of the invention include acute and chronic infections
caused by both RNA and DNA viruses. Without limiting in any way the range of
viral infections that may be treated, Formula I prodrugs, Formula II compounds, and
other compounds of the invention are particularly useful in the treatment of
infections caused by adenoviras, cytomegalovirus, hepatitis A virus (HAV),
hepatitis B virus (HBV), fiaviviruses including Yellow Fever virus and hepatitis C
virus (HCV), herpes simplex type 1 and 2, herpes zoster, human herpesvirus 6,
human immunodeficiency virus (HIV), human papilloma virus (HPV), influenza A
virus, influenza B virus, measles, parainfluenza virus, polidvirus, poxvirus
(including smallpox and monkeypox virus), rhinovirus, respiratory syncytial virus
(RSV), multiple families of viruses that cause hemorrhagic fevers, including the
Arenaviruses (LCM, Junin virus, Machup virus, Guanarito virus, and Lassa Fever),
the Bunyaviruses (Hanta viruses and Rift Valley Fever) and Filoviruses (Ebola and
Marburg virus), a range of viral encephalitides including West Nile virus, LaCrosse
17

WO 2006/066080 PCT/US2005/045589
virus, California Encephalitis virus, Venezuelan Equine Encephalitis virus, Eastern
Equine Encephalitis virus, Western Equine Encephalitis virus, Japanese Encephalitis
virus, Kysanur Forest virus, and tickborne viruses such as Crimean-Congo
Hemorrhagic fever virus.
In another aspect of the invention, Formula I prodrugs, Formula II
compounds, and other compounds of the invention are utilized to treat bacterial,
fungal, and protozoal infections in mammals by administering to the mammal a
therapeutically effective amount of the compounds. The full range of pathogenic
microorganisms is contemplated to be treatable by the compounds of the present
invention, including without limitation those organisms that are resistant to
antibiotics. The ability of compounds to activate multiple components of the
immune system bypasses resistance mechanisms commonly found to reduce
susceptibility to antibiotics, and thus treatment of infections in a mammal caused
by such resistant microorganisms by Formula I prodrags, Formula II
compounds, and other compounds of the invention is a particular utility of the
present invention.
In another aspect of the invention, Formula I prodrugs, Formula II
compounds, and other compounds of the invention are utilized to treat tumors in
mammals by administering to the mammal a therapeutically effective amount of
the compounds. Tumors or cancers contemplated to be treated include but are
not limited to those caused by virus, and the effect may involve inhibiting the
transformation of virus-infected cells to a neoplastic state, inhibiting the spread
of viruses from transformed cells to other normal cells, and/or arresting the
growth of virus-transformed cells. The compounds of the invention are expected
to be useful against a broad spectrum of tumors including but not limited to
carcinomas, sarcomas, and leukemias. Included in such a class are mammary,
colon, bladder, lung, prostate, stomach, and pancreas carcinomas and
lymphoblastic and myeloid leukemias.
In another aspect of the invention, a method of treating a mammal
comprises administering a therapeutically and/or prophylactically effective
amount of a pharmaceutical containing a compound of the invention. In this
aspect the effect may relate to modulation of some portion of the mammal's
immune system, especially modulation of cytokine activities of Thl and Th2,
18

WO 2006/066080 PCT/US2005/045589
including but not restricted to the interleukin family, e.g., EL-1 through IL-12,
and other cytokines such as TNF alpha, and interferons including interferon
alpha, interferon beta, and interferon gamma, and their downstream effectors.
Where modulation of Till and Th2 cytokines occurs, it is contemplated that the
modulation may include stimulation of both Thl and Th2, suppression of both
Thl and Th2, stimulation of either Till or Th2, and suppression of the other, or a
bimodai modulation in which one effect on Thl/Th2 levels (such as generalized
suppression) occurs at a high concentration, while another effect (such as
stimulation of either Thl or Th2 and suppression of the other) occurs at a lower
concentration.
In another aspect of the invention, pharmaceutical compositions
containing a Formula I prodrug, Formula II compound, or other compound of the
invention are administered in a therapeutically effective dose to a mammal that
is receiving anti-infective drugs not included in the compounds of the invention.
In a preferred aspect of this invention, the pharmaceutical compositions
containing a Formula I prodrug, Formula II compound, or other compound of the
invention are administered in a therapeutically effective dose with anti-infective
drug(s) that act directly upon the infectious agent to inhibit the growth of or kill
the infectious agent
In another aspect, the invention encompasses a method for treating or
preventing hepatitis C virus infection in a mammal in need thereof, preferably in a
human in need thereof
In another aspect, the invention encompasses a method for treating or
preventing hepatitis C virus infection in a patient in need thereof, comprising
administering to the patient a therapeutically or prophylactically effective amount of
a Formula I prodrug, Formula II compound, or other compound of the invention and
a pharmaceutically acceptable excipient, carrier, or vehicle.
Baaanother aspect, theinventionencompasses a method for treating or
preventing hepatitis C virus infection in a patient in need thereof, comprising
administering to the patient a therapeutically or prophylactically effective amount of
a compound of a Formula I prodrug, Formula II compound, or other compound of
the invention and an additional therapeutic agent, preferably an additional antiviral
agent or anti-tumor agent as appropriate for the intended use.
19

WO 2006/066080 PCT/US2005/045589
In a preferred aspect of the invention, a pharmaceutical composition
comprising a therapeutically effective amount of a Formula I prodrug provides
for improved oral availability and administration as an immunomodulator. In
another preferred aspect of the invention, a pharmaceutical composition
comprising a therapeutically effective amount of a Formula I prodrug of the
invention provides for masking the active structure as the agent passes through
lymphoid tissue lining the stomach, thereby minimizing activation of this tissue
and allowing for improved oral tolerability.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a plot of pg/ml EFN-a induced in human PBMCs from
compound 134 vs. pg/ml IFN-a induced by an identical concentration of isatoribine.
Figure 2 shows a plot of pg/ml IFN-a induced in human PBMCs from
compound 122 vs. pg/ml IFN-a induced by an identical concentration of isatoribine.
DETAILED DESCRIPTION OF THE
INVENTION AND PREFERRED EMBODIMENTS
Where the following terms are used in this specification, they are used as
defined below:
The terms "comprising" and "including" are used herein in their open,
non-limiting sense.
The term "pyrirnidine" refers to nitrogenous monocyclic heterocycles.
The term "alkyl", as used herein, unless otherwise indicated, includes saturated
monovalent hydrocarbon radicals having straight, branched, or cyclic moieties
(including fused and bridged bicyclic and spirocyclic moieties), or a combination of
the foregoing moieties. For an alkyl group to have cyclic moieties, the group must
have at least three carbon atoms.
The term "alkenyl", as used herein, unless otherwise indicated, includes alkyl
moieties having at least one carbon-carbon double bond wherein alkyl is as defined
above and including E and Z isomers of said alkenyl moiety.
The term "alkynyl", as used herein, unless otherwise indicated, includes alkyl
moieties having at least one carbon-carbon triple bond wherein alkyl is as defined
above.
20

WO 2006/066080 PCT/US2005/045589
The term "alkoxy", as used herein, unless otherwise indicated, includes 0-
alkyl groups wherein alkyl is as defined above.
The term "Me" means methyl, "Et" means ethyl, "Ac" means acetyl, "Bz"
means benzoyl, and "Tol" means toluoyl.
The term "cycloalkyl", as used herein, unless otherwise indicated refers to a
non-aromatic, saturated or partially saturated, monocyclic or fused, spirb or unfused
bicyclic or tricyclic hydrocarbon referred to herein containing a total of from 3 to 10
carbon atoms, preferably 5-8 ring carbon atoms. Exemplary cycloalkyls include
monocyclic rings having from 3-7, preferably 3-6, carbon atoms, such as
cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
Illustrative examples of cycloalkyl are derived from, but not limited to, the
following:

The term "aryl", as used herein, unless otherwise indicated, includes an
organic radical derived from an aromatic hydrocarbon by removal of one hydrogen,
such as phenyl or naphihyl.
The term "heterocyclyl" or "heterocyclic", as used herein, unless otherwise
indicated, includes aromatic (e.g., a heteroaryl) and non-aromatic heterocyclic groups
containing one to four heteroatoms each selected from O, S and N, wherein each
heterocyclic group has from 4-10 atoms in its ring system, and with the proviso that
the ring; of Wtf groupdoesi not contain two adjacent O or S atoms. Non-aromatic
heterocyclic groups include groups having only 4 atoms in their ring system, but
aromatic heterocyclic groups must have at least 5 atoms in their ring system. The
heterocyclic groups include benzo-fused ring systems. An example of a 4 membered
heterocyclic group is azetidinyl (derived from azetidine). An example of a 5
membered heterocyclic group is thiazolyl and an example of a 10 membered
21

WO 2006/066080 PCT/US2005/045589

22
heterocyclic group is quinolinyl. Examples of non-aromatic heterocyclic groups are
pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl,
tetraliydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino,
tfuomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl,
homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-
tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl,
dioxanyl, 1,3-dioxoIanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl,
dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-
azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl.
Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl,
pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl,
oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl,
benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl,
isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl,
benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl,
naphthyridinyl, and fturopyridinyl. The foregoing groups, as derived from the groups
listed above, may be C-attached or N-attached where such is possible. For instance, a
group derived from pyrrole may be pyrrol- 1-yl (N-attached) or pyrrol-3-yl (C-
attached). Further, a group derived from imidazole may be imidazol-1-yl (N-attached)
or imidazol-3-yl (C-attached). The 4-10 membered heterocyclic may be optionally
substituted on any ring carbon, sulfur, or nitrogen atom(s) by one to two oxo, per
ring. An example of a heterocyclic group wherein 2 ring carbon atoms are substituted
with oxo moieties is 1,1-dioxo-thiomorpholinyl. Other illustrative examples of 4-10
membered heterocyclic are derived from, but not limited to, the following:


WO 2006/06080 PCT/US2005/045589

The term "imnunomodulator" refers to natural or synthetic products capable
of modifying the normal or aberrant immune system through stimulation or
suppression.
The term "preventing" refers to the ability of a compound or composition of
the invention to prevent a disease identified herein in patients diagnosed as having
the disease or who are at risk of developing such disease. The term also
encompasses preventing further progression of the disease in patients who are
already suffering from or have symptoms of such disease.
The term "patient" or "subject" means an animal (e.g.t cow, horse, sheep,
pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, guinea pig, etc.) or a
mammal, including chimeric and transgenic animals and mammals. In the treatment
or prevention of HCV infection, the term "patient" or "subject" preferably means a
monkey or a human, most preferably a human. In a specific embodiment the patient
or subject is infected by or exposed to the hepatitis C virus. In certain embodiments,
the patient is a human infant (age 0-2), child (age 2-17), adolescent (age 12-17),
adult (age 18 and up) or geriatric (age 70 and up) patient, hi addition, the patient
includes immunocompromised patients such as HIV positive patients, cancer
patients, patients undergoing immunotherapy or chemotherapy. In a particular
embodiment, the patient is a healthy individual, i.e., not displaying symptoms of
other viral infections.
The term a "therapeutically effective amount" refers to an amount of the
compound of the invention sufficient to provide a benefit in the treatment or
prevention of viral disease, to delay or minimize symptoms associated with viral
infection or viral-induced disease, or to cure or ameliorate the disease or infection or
23

WO 2006/066080 PCT/US2005/045589
cause thereof. In particular, a therapeutically effective amount means an amount
sufficient to provide a therapeutic benefit in vivo. Used in connection with an
amount of a compound of the invention, the term preferably encompasses a non-
toxic amount that improves overall therapy, reduces or avoids symptoms or causes
of disease, or enhances the therapeutic efficacy of or synergies with another
therapeutic agent.
The term a "prophylactically effective amount" refers to an amount of a
compound of the invention or other active ingredient sufficient to result in the
prevention of infection, recurrence or spread of viral infection. A prophylactically
effective amount may refer to an amount sufficient to prevent initial infection or the
recurrence or spread of the infection or a disease associated with the infection. Used
in connection with an amount of a compound of the invention, the term preferably
encompasses a non-toxic amount that improves overall prophylaxis or enhances the
prophylactic efficacy of or synergies with another prophylactic or therapeutic agent.
The term "in combination" refers to the use of more than one prophylactic
and/or therapeutic agents simultaneously or sequentially and in a manner that their
respective effects are additive or synergistic.
The term "treating" refers to:
(i) preventing a disease, disorder, or condition from occurring in an
animal that may be predisposed to the disease, disorder and/or condition, but has not
yet been diagnosed as having it;
(ii) inhibiting the disease, disorder, or condition, i.e., arresting its
development; and
(iii) relieving the disease, disorder, or condition, i.e., causing regression of
the disease, disorder, and/or condition.
The terms "a" and "p*" indicate the specific stereochemical configuration of a
substituent at an asymmetric carbon atom in a chemical structure as drawn.
The compounds of the invention may exhibit the phenomenon of
tautomerism. While the formula drawings cannot expressly depict all possible
tautomeric forms, it is to be understood they are intended to represent any
tautomeric form of the depicted compound and are not to be limited merely to a
specific compound form depicted by the formula drawings. For example, it is
understood for Formula II that regardless of whether or not the substituents are
24

WO 2006/066080 PCT/US2005/045589
shown in their enol or their keto form, they represent the same compound (as
shown in the example below).

Some of the inventive compounds may exist as single stereoisomers (i.e.,
essentially free of other stereoisomers), racemates, and/or mixtures of
enantiomers and/or diastereomers. All such single stereoisomers, racemates and
mixtures thereof are intended to be within the scope of the present invention.
Preferably, the inventive compounds that are optically active are used in
optically pure form-
As generally understood by those skilled in the art, an optically pure
compound having one chiral center (i.e., one asymmetric carbon atom) is one mat
consists essentially of one of the two possible enantiomers (i.e., is enantiomerically
pure), and an optically pure compound having more than one chiral center is one that
is both diastereomerically pure and enantiomerically pure. Preferably, the
compounds of the present invention are used in a form that is at least 90% optically
pure, that is, a form that contains at least 90% of a single isomer (80% enantiomeric
excess ("e.e.") or diastereomeric excess ("d.e.")), more preferably at least 95% (90%
e.e. or d.e.), even more preferably at least 97.5% (95% e.e. or d.e.), and most
preferably at least 99% (98% e.e. or d.e.).
Additionally, Formula I prodrugs, Formula II compounds, and other
compounds of the invention are intended to cover solvated as well as unsolvated
forms of the identified structures. For example, Formula I includes compounds of
the indicated structure in both hydrated and non-hydrated forms. Other examples of
solvates include the structures in combination with isopropanol, ethanol, methanol,
DMSO, ethyl acetate, acetic acid, or ethanolamine.
In addition to Formula I prodrugs, Formula II compounds, and other
compounds of the invention, the invention includes pharmaceutically active
metabolites and pharmaceutically acceptable salts of such compounds and
metabolites.
25

WO 2006/066080 PCT/US2005/045589
"A phannaceutically acceptable prodrug" is a compound that may be
converted under physiological conditions or by solvolysis to the specified compound
or to a pharmaceutically acceptable salt of such compound prior to exhibiting its
pharmacological effect (s). Typically, the prodrug is formulated with the
objective(s) of improved chemical stability, improved patient acceptance and
compliance, improved bioavailability, prolonged duration of action, improved organ
selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased
side effects (e.g., toxicity). The prodrug can be readily prepared using methods
known in the art, such as those described by Burger's Medicinal Chemistry and
Drug Chemistry, 1,172-178,949-982 (1995). See also Bertolini et al., /. Med.
Chem., 40, 2011-2016 (1997); Shan, et al., I Pliann. Set, 86 (7), 765-767;
Bagshawe, Drug Dev. Res., 34,220-230 (1995); Bodor, Advances in Drug Res., 13,
224-331 (1984); Bundgaard, Design ofProdrugs (Elsevier Press 1985); Larsen,
Design and Application of Prodrugs, Drug Design and Development (Rrogsgaard-
Larsen et al., eds., Harwood Academic Publishers, 1991); Dear et al., /.
Chromatogr. B, 748,281-293 (2000); Spraul et al., /. Pharmaceutical & Biomedical
Analysis, 10,601-605 (1992); andProx et al., Xenobiol, 3,103-112 (1992).
"A phannaceutically active metabolite" is intended to mean a
pharmacologically active product produced through metabolism in the body of a
specified compound or salt thereof. After entry into the body, most drugs are
substrates for chemical reactions that may change their physical properties and
biologic effects. These metabolic conversions, which usually affect the polarity of
the compounds of the invention, alter the way in which drugs are distributed in and
excreted from the body. However, in some cases, metabolism of a drug is required
for therapeutic effect. For example, anticancer drugs of the anti-metabolite class
must be converted to their active forms after they have been transported into a
cancer cell.
Since most drugs undergo metabolic transformation of some kind, the
biochemical reactions that play a role in drug metabolism may be numerous and
diverse. The main site of drug metabolism is the liver, although other tissues may
also participate.
A feature characteristic of many of these transformations is that the
metabolic products, or "metabolites," are more polar than the parent drags, although
26

WO 2006/066080 PCT/US2005/045589
a polar drug does sometime yield a less polar product. Substances with high
lipid/water partition coefficients, which pass easily across membranes, also diffuse
back readily from tubular urine through the renal tubular cells into the plasma.
Thus, such substances tend to have a low renal clearance and a long persistence in
the body. If a drag is metabolized to a more polar compound, one with a lower
partition coefficient, its tubular reabsorption will be greatly reduced. Moreover, the
specific secretory mechanisms for anions and cations in the proximal renal tubules
and in the parenchymal liver cells operate upon highly polar substances.
As a specific example, phenacetin (acetophenetidin) and acetanilide are both
mild analgesic and antipyretic agents, but are transformed within the body to a more
polar and more effective metabolite, p-hydroxyacetanilid (acetaminophen), which is
widely used today. When a dose of acetanilide is given to a person, the successive
metabolites peak and decay in the plasma sequentially. During the first hour,
acetanilide is the principal plasma component In the second hour, as the acetanilide
level falls, the metabolite acetaminophen concentration reaches a peak. Finally,
after a few hours, the principal plasma component is a further metabolite that is inert
and can be excreted from the body. Thus, the plasma concentrations of one or more
metabolites, as well as the drug itself, can be pharmacologically important.
"A pharmaceutically acceptable salt" is intended to mean a salt that retains
the biological effectiveness of the free acids and bases of the specified compound
and that is not biologically or otherwise undesirable: A compound of the invention
may possess a sufficiently acidic, a sufficiently basic, or both functional groups, and
accordingly react with any of a number of inorganic or organic bases, and inorganic
and organic acids, to form a pharmaceutically acceptable salt. Exemplary
pharmaceutically acceptable salts include those salts prepared by reaction of the
compounds of the present invention with a mineral or organic acid or an inorganic
base, such as salts including sulfates, pyrosulfates, bisulfates, sulfites, bisulfites,
phosphates, monohydrogenphosphates, dihydfogehphosphates, metaphosphates,
pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates,
caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates,
oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-
1,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates,
dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates,
27

WO 2006/066080 PCT/US2005/045589
xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates,
lactates, Y-hydroxybutyrates, glycolates, tartrates, methane-sulfonates,
propanesulfonates, naphthalene- 1-sulfonates, naphthalene-2-sulfonates, and
mandelates.
If the inventive compound is a base, the desired pharmaceutically
acceptable salt may be prepared by any suitable method available in the art, for
example, treatment of the free base with an inorganic acid, such as hydrochloric
acid, hydrobromic acid, sulfuric acid, nitric add, phosphoric acid and the like, or
with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic
acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid,
salicylic acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid,
an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as
aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid or
cinnamic acid, a sulfonic acid, such as p-toluenesulfonic acid or ethanesulfonic
acid, or the like.
If the inventive compound is an acid, the desired pharmaceutically
acceptable salt may be prepared by any suitable method, for example, treatment
of the free acid with an inorganic or organic base, such as an amine (primary,
secondary or tertiary), an alkali metal hydroxide or alkaline earth metal
hydroxide, or the like. Illustrative examples of suitable salts include organic
salts derived from amino acids, such as glycine and arginine, ammonia, primary,
secondary, and tertiary amines, and cyclic amines, such as piperidine,
morpholine and piperazine, and inorganic salts derived from sodium, calcium,
potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
In the case of agents that are solids, it is understood by those skilled hi the art
that the inventive compounds and salts may exist in different crystal or polymorphic
forms, all of which are intended to be within the scope of the present invention and
specified formulas.
METHODS OF TREATMENT AND PREVENTION OF
HEPATITIS C VIRAL INFECTIONS
The present invention provides methods for treating or preventing a hepatitis
C virus infection in a patient in need thereof.
28

WO 2006/066080 PCT/US2005/045589
The present invention further provides methods for introducing a
therapeutically effective amount of a Formula I prodrug, Formula II compound, or
other compound of the invention or combination of such compounds into the blood
stream of a patient in the treatment and/or prevention of hepatitis C viral infections.
The magnitude of a prophylactic or therapeutic dose of a Formula I prodrug,
Formula II compound, or other compound of the invention or a pharmaceutically
acceptable salt, solvate, or hydrate, thereof in the acute or chronic treatment or
prevention of an infection will vary, however, with the nature and severity of the
infection, and the route by which the active ingredient is administered. The dose,
and in some cases the dose frequency, will also vary according to the infection to be
treated, the age, body weight, and response of the individual patient Suitable dosing
regimens can be readily selected by those skilled in the art with due consideration of
such factors.
The methods of the present invention are particularly well suited for human
patients. In particular, the methods and doses of the present invention can be useful
for immunocompromised patients including, but not limited to cancer patients, HIV
infected patients, and patients with an immunodegenerative disease. Furthermore,
the methods can be useful for immunocompromised patients currently in a state of
remission. The methods and doses of the present invention are also useful for
patients undergoing other antiviral treatments. The prevention methods of the
present invention are particularly useful for patients at risk of viral infection. These
patients include, but are not limited to health care workers, e.g. doctors, nurses,
hospice care givers; military personnel; teachers; childcare workers; patients
traveling to, or living in, foreign locales, in particular third world locales including
social aid workers, missionaries, and foreign diplomats. Finally, the methods and
compositions include the treatment of refractory patients or patients resistant to
treatment such as resistance to reverse transcriptase inhibitors, protease inhibitors,
etc.
Doses
Toxicity and efficacy of the compounds of the invention can be determined
by standard pharmaceutical procedures in cell cultures or experimental animals, e.g.,
for determining the LD50 (the dose lethal to 50% of the population) and the ED50
(the dose therapeutically effective in 50% of the population). The dose ratio
29

WO 2006/066080 PCT/US2005/045589
between toxic and therapeutic effects is the therapeutic index and it can be expressed
as the ratio LD50/ED50.
The data obtained from the cell culture assays and animal studies can be used
in formulating a range of dosage of the compounds for use in humans. The dosage
of such compounds lie preferably within a range of circulating concentrations that
include the ED50 with little or no toxicity. The dosage may vary within this range
depending upon the dosage form employed and the route of administration utilized.
For any compound used in the method of the invention, the therapeutically effective
dose can be estimated initially from cell culture assays. A dose may be formulated
in animal models to achieve a circulating plasma concentration range that includes
the IC50 (i.e., the concentration of the test compound that achieves a half-maximal
inhibition of symptoms) as determined in cell culture; alternatively, the dose of the
compounds may be formulated in animal models to achieve a circulating plasma
concentration range of the compound that corresponds to the concentration required
to achieve a fixed magnitude of response. Such information can be used to more
accurately determine useful doses in humans. Levels in plasma may be measured,
for example, by high performance liquid chromatography.
The protocols and compositions of the invention are preferably tested in
vitro, and then in vivo, for the desired therapeutic or prophylactic activity, prior to
use in humans. For example, in vitro assays which can be used to determine
whether administration of a specific therapeutic protocol is indicated, include in
vitro cell culture assays in which cells that are responsive to the effects of Formula I
prodrags, Formula II compounds, and other compounds of the invention are exposed
to the ligand and the magnitude of response is measured by an appropriate
technique. The assessment of the compounds is then evaluated with respect to the
compound potency, and the degree of conversion between the Formula I prodrug
and Formula II parent compound. Compounds for use in methods of the invention
can be tested in suitable animal model systems prior to testing in humans, including
but not limited to in rats, mice, chicken, cows, monkeys, rabbits, hamsters, etc. The
compounds can then be used in the appropriate clinical trials.
The magnitude of a prophylactic or therapeutic dose of a Formula I prodrug,
Formula II compound, or other compound of the invention or a pharmaceutically
acceptable salt, solvate, or hydrate thereof hi the acute or chronic treatment or
prevention of an infection or condition will vary with the nature and severity of the
30

WO 2006/066080 PCT/US2005/045589
infection, and the route by which the active ingredient is administered. The dose,
and perhaps the dose frequency, will also vary according to the infection to be
treated, the age, body weight, and response of the individual patient. Suitable dosing
regimens can be readily selected by those skilled in the art with due consideration of
such factors. In one embodiment, the dose administered depends upon the specific
compound to be used, and the weight and condition of the patient Also, the dose
may differ for various particular compounds of the invention; suitable doses can be
predicted on the basis of the aforementioned in vitro measurements and on the basis
of animal studies, such that smaller doses will be suitable for those compounds that
show effectiveness at lower concentrations than other compounds when measured in
the systems described or referenced herein. In general, the dose per day is in the
range of from about 0.001 to 100 mg/kg, preferably about 1 to 25 mg/kg, more
preferably about 5 to 15 mg/kg. For treatment of humans infected by hepatitis C
viruses, about 0.1 mg to about 15 g per day is administered in about one to four
divisions a day, preferably 100 mg to 12 g per day, more preferably from 100 mg to
8000 mg per day.
Additionally, the recommended daily dose ran can be administered in cycles
as single agents or in combination with other therapeutic agents. In one
embodiment, the daily dose is administered in a single dose or in equally divided
doses. In a related embodiment, the recommended daily dose can be administered
once time per week, two times per week, three times per week, four times per week
or five times per week.
In a preferred embodiment, the compounds of the invention are administered
to provide systemic distribution of the compound within the patient. In a related
embodiment, the compounds of the invention are administered to produce a systemic
effect in the body.
In another embodiment the compounds of the invention are administered via
oral, mucosal (including sublingual, buccal, rectal, nasal, or vaginal), parenteral
(including subcutaneous, intramuscular, bolus injection, intraarterial, or
intravenous), transdermal, or topical administration. In a specific embodiment the
compounds of the invention are administered via mucosal (including sublingual,
buccal, rectal, nasal, or vaginal), parenteral (including subcutaneous, intramuscular,
bolus injection, intraarterial, or intravenous), transdermal, or topical administration.
In a further specific embodiment, the compounds of the invention are administered
31

WO 2006/066080 PCT/US2005/045589
via oral administration. In a further specific embodiment, the compounds of the
invention are not administered via oral administration.
Different therapeutically effective amounts may be applicable for different
infections, as will be readily known by those of ordinary skill in the art. Similarly,
amounts sufficient to treat or prevent such infections, but insufficient to cause, or
sufficient to reduce, adverse effects associated with conventional therapies are also
encompassed by the above described dosage amounts and dose frequency schedules.
Combination Therapy
Specific methods of the invention further comprise the administration of an
additional therapeutic agent {i.e., a therapeutic agent other than a compound of the
invention). In certain embodiments of the present invention, the compounds of the
invention can be used in combination with at least one other therapeutic agent.
Therapeutic agents include, but are not limited to antibiotics, antiemetic agents,
antidepressants, and antifungal agents, anti-inflammatory agents, antiviral agents,
anticancer agents, immunomodulatory agents, |}-interferons, alkylating agents,
hormones or cytokines. In a preferred embodiment the invention encompasses the
administration of an additional therapeutic agent that is HCV specific or
demonstrates anti-HCV activity.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with antibiotics. For
example, they can be formulated with a macrolide (e.g., tobramycin (Tobi®)), a
cephalosporin (e.g., cephalexin (Keflex®), cephradine (Velosef®), cefuroxime
(Ceftin®), cefprozil (Cefzil®), cefaclor (Ceclor®), cefixime (Suprax®) or .
cefadroxil (Duricef®)), a clarithromycin (e.g., clarithromycin (Biaxin®)), an
eryflironiycin (e.g., erythromycin (EMycin®)), a penicillin (e.g., penicillin V (V-
Cillin K® or Pen Vee K®)) or a quinolone (e.g., ofloxacin (Floxin®), ciprofloxacin
(Cipro®) or norfloxacin (Noroxin®)),aminoglycoside antibiotics (e.g., apramycin,
arbekacin, bambermycins, butirosin, dibekacin, neomycin, neomycin, undecylenate,
netilmicin, paromomycin, ribostamycin, sisomicin, and spectinomycin), amphenicol
antibiotics (e.g., azidamfenicol, chloramphenicol, fiorfenicol, and thiamphenicol),
ansamycin antibiotics (e.g., rifamide and rifampin), carbacephems (e.g., loracarbef),
carbapenems (e.g., biapenem and imipenem), cephalosporins (e.g., cefaclor,
cefadroxil, cefamandole, cefatrizine, cefazedone, cefozopran, cefpimizole,
cefpiramide, and cefpirome), cephamycins (e.g., cefbuperazone, cefmetazole, and
32

WO 2006/066080 PCT/US2005/045S89
cefminox), monobactams (e.g., aztreonam, carumonam, and tigemonam),
oxacephems (e.g., flomoxef, and moxalactam), penicillins (e.g., amdinocillin,
amdinocillin pivoxil, amoxicillin, bacampicillin, benzylpenicillinic acid,
benzylpenicillin sodium, epicillin, fenbenicillin, floxacillin, penamccillin,
penethamate hydriodide, penicillin o-benethamine, penicillin 0, penicillin V,
penicillin V benzathine, penicillin V hydrabamine, penimepicycline, and
phencihicillinpotassium), lincosamides (e.g., clindamycin, and Iincomycin),
amphomycin, bacitracin, capreomycin, colistin, enduracidin, enviomycin,
tetracyclines (e.g,, apicycline. chlortetracycline, clomocycline, and demeclocycline),
2,4-diaminopyrimidines (e.g., brodimoprim), nitrofurans (e.g., furaltadone, and
fuiazolium chloride), quinolones and analogs thereof (e.g., cinoxacin,, clinafloxacin,
flumequine, and grepagloxacin), sulfonamides (e.g., acetyl sulfamethoxypyrazine,
benzylsulfamide, noprylsulfamide, phthalylsulfacetamide, sulfachrysoidine, and
sulfacytine), sulfones (e.g., diathymosulfone, glucosulfone sodium, and solasulfone),
cycloserine, mupirocin and tuberin.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can also be administered or formulated in combination with an antiemetic
agent Suitable antiemetic agents include, but are not limited to, metoclopromide,
domperidone; prochlorperazine, promethazine, chlorpromazine, trimethobenzamide,
ondansetron, granisetron, hydroxyzine, acethylleucine monoethanolamine,
alizapride, azasetron, benzquinamide, bietanautine, bromopride, buclizine,
clebopride, cyclizine, dimenhydrinate, diphenidol, dolasetron, meclizine,
methallatal, metopimazine, nabilone, oxyperndyl, pipamazine, scopolamine,
sulpiride, tetrahydrocannabinols, thiethylperazjne, thioproperazine, tropisetron, and
mixtures thereof.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an antidepressant
Suitable antidepressarifs include, but are not limited to, binedaline, caroxazone,
citalopram, dimethazan, fencamine, indalpine, indeloxazine hydrocholoride,
nefopam, nomifensine, oxitriptan, qxypertine, paroxetine, sertraline, thiazesim,
trazodone, benmoxine, iproclozide, iproniazid, isocarboxazid, nialamide, octamoxin,
phenelzine, cotmine, rolicyprine, rolipram, maprotiline, metralindole, mianserin,
mirtazepine, adinazolam, amitriptyline, amitriptylinoxide, amoxapine, butriptyline,
clomipramine, demexiptiline, desipramine, dibenzepin, dimetacrine, dothiepin,
33

WO 2006/066080 PCT/US2005/045589
doxepin, fluacizine, imipramine, imipramine N-oxide, iprindole, lofepramine,
melitracen, metapramine, nortriptyline, noxiptilin, opipramol, pizotyline,
propizepine, protriptyline, quinupramine, tianeptine, trimipramitie, adrafinil,
benactyzine, bupropion, butacetin, dioxadrol, duloxetine, etoperidone, febarbamate,
femoxetine, fenpentadiol, fluoxetine, fluvoxamine, hematoporphyrin, hypericin,
levophacetoperane, medifoxatnine, milnacipran, minaprine, moclobemide,
nefazodone, oxaflozane, piberaline, prolintane, pyrisuccideanol, ritanserin,
roxindole, rubidium chloride, sulpiride, tandospirone, thozalinone, tofenacin,
toloxatone, tranylcypromine, L-tryptophan, venlafaxine, viloxazine, and zimeldine.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an antifungal
agent. Suitable antifungal agents include but are not limited to amphotericin B,
itraconazole, ketoconazole, fluconazole, intrathecal, flucytosine, miconazole,
butoconazole, clotrimazole, nystatin, terconazole, tioconazole, ciclopirox, econazole,
haloprogrin, naftifine, terbinafine, undecylenate, and griseofuldin.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an anti-
inflammatory agent Useful anti-inflammatory agents include, but are not limited to,
non-steroidal anti-inflammatory drugs such as salicylic acid, acetylsalicylic acid,
methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, acetaminophen,
indomethacin, sulindac, etodolac, niefenamic acid, meclofenamate sodium, tolmetin,
ketorolac, dichlofenac, ibuprofen, naproxen, naproxen sodium, fenoprofen,
ketoprofen, flurbinprofen, oxaprozin, piroxicam, meloxicam, ampiroxicam,
droxicam, pivoxicam, tenoxicam, nabumetome, phenylbutazone, oxyphenbutazone,
antipyrine, aminopyrine, apazone and nimesulide; leukotriene antagonists including,
but not limited to, zileuton, aurothioglucose, gold sodium thiomalate and auranofin;
steroids including, but not limited to, alclometasone diproprionate, amcinonide,
beclomethasone dipropionate, betametasone, betamethasone benzoate,
betamethasone diproprionate, betamethasone sodium phosphate, betamethasone
valerate, clobetasol proprionate, clocortolone pivalate, hydrocortisone,
hydrocortisone derivatives, desonide, desoximatasone, dexamethasone, fmnisolide,
flucoxinolide, flurandrenolide, halcinocide, medrysone, methylprednisolone,
- methprednisolone acetate, methylprednisolone sodium succinate, mometasone
furoate, paratnethasone acetate, prednisolone, prednisolone acetate, prednisolone
34

WO 2006/066080 PCT/US2005/045S89
sodium phosphate, prednisolone tebuatate, prednisone, triamcinolone, triamcinolone
acetonide, triamcinolone diacetate, and triamcinolone hexacetonide; and other anti-
inflammatory agents including, but not limited to, methotrexate, colchicine,
allopurinol, probenecid, sulfinpyrazone and benzbromarone.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with another antiviral
agent Useful antiviral agents include, but are not limited to, protease inhibitors,
nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase
inhibitors and nucleoside analogs. The antiviral agents include but are not limited to
zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifiuridine, levovirin, •
viramidine and ribavirin, as well as foscarnet, amantadine, rimantadine, saquinavir,
indinavir, amprenavir, lopinavir, ritonavir, the alpha-interferons; beta-interferons;
adefovir, clevadine, entecavir, pleconaril.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an
immunomodulatory agent. Immunomodulatory agents include, but are not limited
to, methothrexate, lefiunomide, cyclophosphamide, cyclosporine A, mycophenolate
mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar,
malononitriloamindes (e.g., leflunamide), T cell receptor modulators, and cytokine
receptor modulators, peptide mimetics, and antibodies (e. g., human, humanized,
chimeric, monoclonal, polyclonal, Fvs, ScFvs, Fab or F(ab)2 fragments or epitope
binding fragments), nucleic acid molecules (e.g., antisense nucleic acid molecules
and triple helices), small molecules, organic compounds, and inorganic compounds.
Examples of T cell receptor modulators include, but are not limited to, anti-T cell
receptor antibodies (e.g., anti-CD4 antibodies (e.g., CM-T412 (Boeringer), EDEC-
CE9.1® (IDEC and SKB), mAB 4162W94, Orthoclone and OKTcdr4a (Janssen-
Cilag)), anti-CD3 antibodies (e.g., Nuvion (Product Design Labs), OKT3 (Johnson
&7dhnson), or Rituxan (IDEC)), anti-CD5 antibodies (e.g., ananti-CD5 ricin-linked
immunoconjugate), anti-CD7 antibodies (e.g., CHH-380 (Novartis)), anti-CD8
antibodies, anti-CD40 ligandmonoclonal antibodies (e.g., IDEC-131 (IDEC)), anti-
CD52 antibodies (e.g., CAMPATH1H (Ilex)), anti-CD2 antibodies, anti-CDlla
antibodies (e.g., Xanelim (Genentech)), and anti-B7 antibodies (e.g., DDEC-114
(EDEC)) and CTLA4-immunoglobulin. Examples of cytokine receptor modulators
include, but are not limited to, soluble cytokine receptors (e.g., the extracellular
35

WO 2006/066080 PCT/US2005/045589
domain of a TNF-a receptor or a fragment thereof, the extracellular domain of an
IDL-b receptor or a fragment thereof, and the extracellular domain of an IL-6
receptor or a fragment thereof), cytokines or fragments thereof (e.g., interleukin
(DL)-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, EL-11, DL-12, IL-15, TNF-a,
interferon (IFN)-a, IFN-p, IFN-y, and GM-CSF), anti-cytokine receptor antibodies
(e.g., anti-EFN receptor antibodies, anti-IL-2 receptor antibodies (e.g., Zenapax
(Protein Design Labs)), anti-IL-4 receptor antibodies, anti-DL-6 receptor antibodies,
anti-IL-10 receptor antibodies, and anti-IL-12 receptor antibodies), anti-cytokine
antibodies (e.g., anti-IFN antibodies, anti-TNF-a antibodies, anti-BL-ip antibodies,
anti-IL-6 antibodies, anti-IL-8 antibodies (e.g., ABX-IL-8 (Abgenix)), and anti-IL-
12 antibodies).
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an agent which
inhibits viral enzymes, including but not limited to inhibitors of HCV protease, such
as BELN 2061 and inhibitors of NS5b polymerase such as NM107 and its prodrug
NM283 (Idenix Pharmaceuticals, Inc., Cambridge, MA).
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an agent which
inhibits HCV polymerase such as those described in Wu, Curr Drug Targets Infect
Disord. 2003;3(3):20749 or in combination with compounds that inhibit the
helicase function of the virus such as those described in Bretner M, et al
Nucleosides Nucleotides Nucleic Adds. 2003;22(5-8):1531, or with inhibitors of
other HCV specific targets such as those described in Zhang X. IDrugs.
2002;5(2):154-8.
The Formula I prodrugs, Formula n compounds, and other compounds of the
invention can be administered or formulated in combination with an agent which
inhibits viral replication.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with cytokines.
Examples of cytokines include, but are not limited to, interleukin-2 (IL-2),
interleukin-3 (IL-3), interleukin4 (IL-4), interleukin-5 (IL-5), interleukin-6 (IL-6),
interleukin-7 (IL-7), interleukin-9 (IL-9), interleukin-10 (IL-10), interleukin-12 (EL-
12), interleukin 15 (DL-15), ihterleukin 18 (EL-18), platelet derived growth factor
(PDGF), erythropoietin (Epo), epidermal growth factor (EGF), fibroblast growth
36

WO 2006/066080 PCT/US2005/045589
factor (FGF), granulocyte macrophage stimulating factor (GM-CSF), granulocyte
colony stimulating factor (G-CSF), macrophage colony stimulating factor (M-CSF),
prolactin, and interferon (IFN), e.g., IFN-alpha, and IFN-gamma).
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with hormones.
Examples of hormones include, but are not limited to, luteinizing hormone releasing
hormone (LHRH), growth hormone (GH), growth hormone releasing hormone,
ACTH, somatostatin, somatotropin, somatomedin, parathyroid hormone,
hypothalamic releasing factors, insulin, glucagon, enfcephalins, vasopressin,
calcitonin, heparin, low molecular weight heparins, heparinoids, synthetic and
natural opioids, insulin thyroid stimulating hormones, and endorphins.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with P-interferons
which include, but are not limited to, interferon beta-la, interferon beta-lb.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with a-interferons
which include, but are not limited to, interferon alpha-1, interferon alpha-2a
(roferon), interferon alpha-2b, intron, Peg-Intron, Pegasys, consensus interferon
(infergen) and albuferon.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an absorption
enhancer, particularly those which target the lymphatic system, including, but not
limited to sodium glycocholate; sodium caprate; N-lauryl-y-D-maltopyranoside;
EDTA; mixed micelle; and those reported m Muranishi Crit. Rev. Ther. Drug
Carrier Syst., 7-1-33, which is hereby incorporated by reference in its entirety.
Other known absorption enhancers can also be used. Thus, the invention also
encompasses a pharmaceutical composition comprising one or more Formula I
prodrugs, Formula II compounds, and other compounds of the invention and one or
more absorption enhancers.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention can be administered or formulated in combination with an alkylating
agent. Examples of alkylating agents include, but are not limited to nitrogen
mustards, emyleriimines, methyhnelamines, alkyl sulfonates, nitrosoureas, triazenes,
mechloremamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil,
37

WO 2006/066080 PCT/US2005/045589
hexamethylmelaine, thiotepa, busulfan, carmustine, streptozocin, dacarbaziae and
temozolomide.
The Formula I prodrugs, Formula II compounds, and other compounds of the
invention and the other therapeutics agent can act additively or, more preferably,
synergistically. In a preferred embodiment, a composition comprising a compound
of the invention is administered concurrently with the administration of another
therapeutic agent, which can be part of the same composition or in a different
composition from that comprising the compounds of the invention. In another
embodiment, a compound of the invention is administered prior to or subsequent to
administration of another therapeutic agent. In a separate embodiment, a compound
of the invention is administered to a patient who has not previously undergone or is
not currently undergoing treatment with another therapeutic agent, particularly an
antiviral agent.
In one embodiment, the methods of the invention comprise the
administration of one or more Formula I prodrags, Formula II compounds, and other
compounds of the invention without an additional therapeutic agent.
PHARMACEUTICAL COMPOSITIONS AND DOSAGE FORMS
Pharmaceutical compositions and single unit dosage forms comprising a
Formula I prodrugs, Formula II compounds, and other compounds of the invention,
or a pharmaceutically acceptable salt, or hydrate thereof, are also encompassed by
the invention. Individual dosage forms of the invention may be suitable for oral, .
mucosal (including sublingual, buccal, rectal, nasal, or vaginal), parenteral
(including subcutaneous, intramuscular, bolus injection, intraarterial, or
intravenous), transdermal, or topical administration. Pharmaceutical compositions
and dosage forms of the invention typically also comprise one or more
pharmaceutically acceptable exdpients. Sterile dosage forms are also contemplated.
In an alternative embodiment, a pharmaceutical composition encompassed
by this embodiment includes a Formula I prodrug, Formula II compound, or other
compound of the invention, or a pharmaceutically acceptable salt, or hydrate thereof,
and at least one additional therapeutic agent Examples of additional therapeutic
agents include, but are not limited to, those listed above.
The composition, shape, and type of dosage forms of the invention will
typically vary depending on their use. For example, a dosage form used in the acute
treatment of a disease or a related disease may contain larger amounts of one or
38

WO 2006/066080 PCT/US2005/045589
more of the active ingredients it comprises than a dosage form used in the chronic
treatment of the same disease. Similarly, a parenteral dosage form may contain
smaller amounts of one or more of the active ingredients it comprises than an oral
dosage form used to treat the same disease or disorder. These and other ways in
which specific dosage forms encompassed by this invention will vary from one
another will be readily apparent to those skilled in the art. See, e.g., Remington's
Pharmaceutical Sciences, 18th ed, Mack Publishing, Easton PA (1990). Examples
of dosage forms include, but are not limited to: tablets; caplets; capsules, such as
soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories;
ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters;
solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms
suitable for oral or mucosal administration to a patient, including suspensions (e.g.,
aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or a water-in-oil
liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral
administration to a patient; and sterile solids (e.g., crystalline or amorphous solids)
that can be reconstituted to provide liquid dosage forms suitable for parenteral
administration to a patient.
Typical pharmaceutical compositions and dosage forms comprise one or
more carriers, excipients or diluents. Suitable excipients are well known to those
skilled in the art of pharmacy, and non-limiting examples of suitable excipients are
provided herein. Whether a particular excipient is suitable for incorporation into a
pharmaceutical composition or dosage form depends on a variety of factors well
known in the art including, but not limited to, the way in which the dosage form will
be administered to a patient For example, oral dosage forms such as tablets may
contain excipients not suited for use in parenteral dosage forms. The suitability of a
particular excipient may also depend on the specific active ingredients in the dosage
form.
This invention further encompasses anhydrous pharmaceutical compositions
and dosage forms comprising active ingredients, since water can facilitate the
degradation of some compounds. For example, the addition of water (e.g., 5%) is
widely accepted in the pharmaceutical arts as a means of simulating long-term
storage in order to determine characteristics such as shelf-life or the stability of
formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles &
Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80. In effect, water and
39

WO 2006/066080 PCT/US2005/04SS89
heat accelerate the decomposition of some compounds. Thus, the effect of water on
a formulation can be of great significance since moisture and/or humidity are
commonly encountered during manufacture, handling, packaging, storage, shipment,
and use of formulations.
Anhydrous pharmaceutical compositions and dosage forms of the invention
can be prepared using anhydrous or low moisture containing ingredients and low
moisture or low humidity conditions.
An anhydrous pharmaceutical composition should be prepared and stored
such that its anhydrous nature is maintained. Accordingly, anhydrous compositions
are preferably packaged using materials known to prevent exposure to water such
that they can be included in suitable formulary kits. Examples of suitable packaging
include, but are not limited to, hermetically sealed foils, plastics, unit dose
containers (g.g.( vials), blister packs, and strip packs.
The invention further encompasses pharmaceutical compositions and dosage
forms that comprise one or more compounds that reduce the rate by which an active
ingredient will decompose. Such compounds, which are referred to herein as
"stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH
buffers, or salt buffers.
Like the amounts and types of excipients, the amounts and specific types of
active ingredients in a dosage form may differ depending on factors such as, but not
limited to, the route by which it is to be administered to patients. However, typical.
dosage forms of the invention comprise compounds of the invention, or a
pharmaceutically acceptable salt or hydrate thereof comprise 0.1 mg to 1500 mg per
unit to provide doses of about 0.01 to 200 mg/kg per day.
Oral Dosage Forms
Pharmaceutical compositions of the invention that are suitable for oral
administration can be presented as discrete dosage forms, such as, but are not limited
to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored
syrups). Such dosage forms contain predetermined amounts of active ingredients,
and may be prepared by methods of pharmacy well known to those skilled in the art.
See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing,
EastonPA(1990).
Typical oral dosage forms of the invention are prepared by combining the
active ingredients) in an intimate admixture with at least one excipient according to
40

WO 2006/066080 PCT/US2005/045589
conventional pharmaceutical compounding techniques. Excipients can take a wide
variety of forms depending on the form of preparation desired for administration.
For example, excipients suitable for use in oral liquid or aerosol dosage forms
include, but are not limited to, water, glycols, oils, alcohols, flavoring agents,
preservatives, and coloring agents. Examples of excipients suitable for use in solid
oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not
limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents,
lubricants, binders, and disintegrating agents.
Because of their ease of administration, tablets and capsules represent the
most advantageous oral dosage unit forms, in which case solid excipients are
employed. If desired, tablets can be coated by standard aqueous or nonaqueous
techniques. Such dosage forms can be prepared by any of the methods of pharmacy.
In general, pharmaceutical compositions and dosage forms are prepared by
uniformly and intimately admixing the active ingredients with liquid carriers, finely
divided solid carriers, or both, and then shaping the product into the desired
presentation if necessary.
For example, a tablet can be prepared by compression or molding.
Compressed tablets can be prepared by compressing in a suitable machine the active
ingredients in a free-flowing form such as powder or granules, optionally mixed
with an excipient. Molded tablets can be made by molding in a suitable machine a
mixture of the powdered compound moistened with an inert liquid diluent.
Examples of excipients that can be used in oral dosage forms of the invention
include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders
suitable for use in pharmaceutical compositions and dosage forms include, but are
not limited to, corn starch, potato starch, or other starches,, gelatin, natural and
synthetic gums such as acacia, sodium alginate, alginic acid, other alginates,
powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose,
cellulose acetate, carboxymethyl cellulose calcium, sodium carbdxymethyl
cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch,
hydroxypropyl methyl cellulose, (e.g., Nos. 2208,2906,2910), microcrystalline
cellulose, and mixtures thereof.
Examples of fillers suitable for use in the pharmaceutical compositions and
dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate
(e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates,
41

WO 2006/066080 PCT/US2005/045589
kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures
thereof. The binder or filler in pharmaceutical compositions of the invention is
typically present in from about 50 to about 99 weight percent of the pharmaceutical
composition or dosage form.
Suitable forms of microcrystalline cellulose include, but are not limited to,
the materials sold as AVICEL-PH-101, AVICEL-PH-103 AVICEL RC-581,
AVICEL-PH-105 (available from FMC Corporation, American Viscose Division,
Avicel Sales, Marcus Hook, PA), and mixtures thereof. An specific binder is a
mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as
AVICEL RC-581. Suitable anhydrous or low moisture excipients or additives
include AVICEL-PH-103™ and Starch 1500 LM.
Disintegrants are used in the compositions of the invention to provide tablets
that disintegrate when exposed to an aqueous environment Tablets that contain too
much disintegrant may disintegrate in storage, while those that contain too little may
not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient
amount of disintegrant that is neither too much nor too little to detrimentally alter
the release of the active ingredients should be used to form solid oral dosage forms
of the invention. The amount of disintegrant used varies based upon the type of
formulation, and is readily discernible to those of ordinary skill in the art Typical
pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of
disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
Disintegrants that can be used in pharmaceutical compositions and dosage
forms of the invention include, but are not limited to, agar-agar, alginic acid,
calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone,
polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre-
gelatinized starch, other starches, clays, other algins, other celluloses, gums, and
mixtures thereof.
Lubricants that can be used in pharmaceutical compositions and dosage
forms of the invention include, but are not limited to, calcium stearate, magnesium
stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene
glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated
vegetable oil {e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil,
corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and
mixtures thereof. Additional lubricants include, for example, a syloid silica gel
42

WO 2006/066080 PCT/US2005/045589
(AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated
aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB-O-SIL (a
pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures
thereof. If used at all, lubricants are typically used in an amount of less than about 1
weight percent of the pharmaceutical compositions or dosage forms into which they
are incorporated.
Delayed Release Dosage Forms
Active ingredients of the invention can be administered by controlled release
means or by delivery devices that are well known to those of ordinary skill in the art.
Examples include, but are not limited to, those described in U.S. Patent Nos.:
3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, 5,674,533,5,059,595,
5,591,767,5,120,548, 5,073,543,5,639,476, 5,354,556, and 5,733,566, each of
which is incorporated herein by reference. Such dosage forms can be used to
provide slow or controlled-release of one or more active ingredients using, for
example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable
membranes, osmotic systems, multilayer coatings, microparticles, liposomes,
microspheres, or a combination thereof to provide the desired release profile in
varying proportions. Suitable controlled-release formulations known to those of
ordinary skill in the art, including those described herein, can be readily selected for
use with the active ingredients of the invention. The invention thus encompasses
single unit dosage forms suitable for oral administration such as, but not limited to,
tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
All controlled-release pharmaceutical products have a common goal of
improving drug therapy over that achieved by their non-controlled counterparts.
Ideally, the use of an optimally designed controlled-release preparation in medical
treatment is characterized by a minimum of drug substance being employed to cure
or control the condition in a minimum amount of time. Advantages of controlled-
release formulations include extended activity of the drug, reduced dosage
frequency, and increased patient compliance. In addition, controlled-release
formulations can be used to affect the time of onset of action or other characteristics,
such as blood levels of the drug, and can thus affect the occurrence of side (e.g.,
adverse) effects.
Most controlled-release formulations are designed to initially release an
amount of drug (active ingredient) that promptly produces the desired therapeutic
43

WO 2006/066080 PCT/US2005/045589
effect, and gradually and continually release of other amounts of drug to maintain
this level of therapeutic or prophylactic effect over an extended period of time. In
order to maintain this constant level of drug in the body, the drug must be released
from the dosage form at a rate that will replace the amount of drug being
metabolized and excreted from the body. Controlled-release of an active ingredient
can be stimulated by various conditions including, but not limited to, pH,
temperature, enzymes, water, or other physiological conditions or compounds.
Parenteral Dosage Forms
Parenterai dosage forms can be administered to patients by various routes
including, but not limited to, subcutaneous, intravenous (including bolus injection),
intramuscular, and intraarterial. Because their administration typically bypasses
patients' natural defenses against contaminants, parenteral dosage forms are
preferably sterile or capable of being sterilized prior to administration to a patient.
Examples of parenteral dosage forms include, but are not limited to, solutions ready
for injection, dry and/or lyophylized products ready to be dissolved or suspended in
a pharmaceutically acceptable vehicle for injection (reconstitutable powders),
suspensions ready for injection, and emulsions.
Suitable vehicles that can be used to provide parenteral dosage forms of the
invention are well known to those skilled in the art. Examples include, but are not
limited to: Water for Injection USP; aqueous vehicles such as, but not limited to,
Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and
Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles
such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene
glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil,
peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
Compounds that increase the solubility of one or more of the active ingredients
disclosed herein can also be incorporated into the parenteral dosage forms of the
invention.
Transdermal Dosage Forms
Transdermal dosage forms include "reservoir type" or "matrix type" patches,
which can be applied to the skin and worn for a specific period of time to permit the
penetration of a desired amount of active ingredients.
Suitable excipients (e.g., carriers and diluents) and other materials that can
be used to provide transdermal and topical dosage forms encompassed by this
44

WO 2006/066080 PCT/US2005/045589
invention are well known to those skilled in the pharmaceutical arts, and depend on
the particular tissue to which a given pharmaceutical composition or dosage form
will be applied. With that fact in mind, typical excipients include, but are not
limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane-1,3-
diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof.
Depending on the specific tissue to be treated, additional components may be used
prior to, in conjunction with, or subsequent to treatment with active ingredients of
the invention. For example, penetration enhancers can be used to assist in delivering
the active ingredients to the tissue. Suitable penetration enhancers include, but are
not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl;
alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl
formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone;
Kollidon grades (Povidone, Polyvidone); urea; and various water-soluble or
insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan
monostearate).
The pH of a pharmaceutical composition or dosage form, or of the tissue to
which the pharmaceutical composition or dosage form is applied, may also be
adjusted to improve delivery of one or more active ingredients. Similarly, the
polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve
delivery. Compounds such as stearates can also be added to pharmaceutical
compositions or dosage forms to advantageously alter the hydrophilicity or
lipophilicity of one or more active ingredients so as to improve delivery. In this
regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying
agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent
Different salts, hydrates or solvates of the active ingredients can be used to further
adjust the properties of the resulting composition.
Topical Dosage Forms
Topical dosage forms of the invention include, but are not limited to, creams,
lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to
one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 18th eds.,
Mack Publishing, Easton PA (1990); and Introduction to Pharmaceutical Dosage
Forms, 4th ed., Lea & Febiger, Philadelphia (1985).
Suitable excipients (e.g., carriers and diluents) and other materials that can
be used to provide transdermal and topical dosage forms encompassed by this
45

WO 2006/066080 PCT/US2005/045589
invention are well known to those skilled in the pharmaceutical arts, and depend on
the particular tissue to which a given pharmaceutical composition or dosage form
will be applied. With that fact in mind, typical excipients include, but are not
limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane-1,3-
diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof.
Depending on the specific tissue to be treated, additional components may be
used prior to, in conjunction with, or subsequent to treatment with active ingredients
of the invention. For example, penetration enhancers can be used to assist in
delivering the active ingredients to the tissue. Suitable penetration enhancers
include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and
tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide;
dimethyl formamide; polyethylene glycol; pyrrolidones such as
polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various
water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span
60 (sorbitan monostearate).
Mucosal Dosage Forms
Mucosal dosage forms of the invention include, but are not limited to,
ophthalmic solutions, sprays and aerosols, or other forms known to one of skill in
the art See, e.g., Remington's Pharmaceutical Sciences, 18th eds., Mack
Publishing, Easton PA (1990); and Introduction to Pharmaceutical Dosage Forms,
4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating
mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral
gels.. In one embodiment, the aerosol comprises a carrier. In another embodiment,
the aerosol is carrier free.
The compounds of the invention may also be administered directly to the
lung by inhalation. For administration by inhalation, a compound of the invehtionr
can be conveniently delivered to the lung by a number of different devices. For
example, a Metered Dose Inhaler ("MDI") which utilizes canisters that contain a
suitable low boiling propellant, e.g., dichlorodifluoromethane,
trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable
gas can be used to deliver a compound directly to the lung. MDI devices are
available from a number of suppliers such as 3M Corporation, Aventis, Boehringer
tngleheim, Forest Laboratories, Glaxo-Wellcome, Schering Plough and Vectura.
46

WO 2006/066080 PCT/US2005/045589
Alternatively, a Dry Powder Inhaler (DPI) device can be used to administer a
compound of the invention to the lung (see, e.g., Raleigh et ah, Proc. Amer. Assoc.
Cancer Research Annual Meeting, 1999,40,397, which is herein incorporated by
reference). DPI devices typically use a mechanism such as a burst of gas to create a
cloud of dry powder inside a container, which can then be inhaled by the patient.
DPI devices are also well known in the art and can be purchased from a number of
vendors which include, for example, Fisons, Glaxo-Wellcome, Inhale Therapeutic
Systems, ML Laboratories, Qdose and Vectura. A popular variation is the multiple
dose DPI ("MDDPI") system, which allows for the delivery of more than one
therapeutic dose. MDDPI devices are available from companies such as
AstraZcneca, GlaxoWellcome, IVAX, Schering Plough, SkyePharma and Vectura.
For example, capsules and cartridges of gelatin for use in an inhaler or insufflator
can be formulated containing a powder mix of the compound and a suitable powder
base such as lactose or starch for these systems.
Another type of device that can be used to deliver a compound of the
invention to the lung is a liquid spray device supplied, for example, by Aradigm
Coiporation. Liquid spray systems use extremely small nozzle holes to aerosolize
liquid drug formulations that can then be directly inhaled into the lung.
In a preferred embodiment, a nebulizer device is used to deliver a compound
of tlie invention to the lung. Nebulizers create aerosols from liquid drug
formulations by using, for example, ultrasonic energy to form fine particles that can
be readily inhaled (See e.g., Verschoyle et ah, British J. Cancer, 1999, 80, Suppl 2,
96, which is herein incorporated by reference). Examples of nebulizers include
devices supplied by Sheffield/Systemic Pulmonary Delivery Ltd. (See, Armer et ah,
U.S. Pat. No. 5,954,047; van der Linden et ah, U.S. Pat. No. 5,950,619; van der
Linden et al., U.S. Pat. No. 5,970,974, which are herein incorporated by reference),
Aventis and Batelle Pulmonary Therapeutics. .
Ma particularly preferred embodiment, an electrohydrodynamic ("EHD")
aerosol device is used to deliver compounds of the invention to the lung. EHD
aerosol devices use electrical energy to aerosolize liquid drug solutions or
suspensions (see, e.g., Noakes et ah, U.S. Pat. No. 4,765,539; Coffee, U.S. Pat. No.,
4,962,885; Coffee, PCT Application, WO 94/12285; Coffee, PCT Application, WO
94/14543; Coffee, PCT Application, WO 95/26234, Coffee, PCT Application, WO
95/26235, Coffee, PCT Application, WO 95/32807, which are herein incorporated
47

WO 2006/066080 PCT/US200S/045589
by reference). The electrochemical properties of the formulation may be important
parameters to optimize when delivering this drug to the lung with an EHD aerosol
device and such optimization is routinely performed by one of skill in the art. EHD
aerosol devices may more efficiently delivery drugs to the lung than existing
pulmonary delivery technologies. Other methods of intra-pulmonary delivery of the
compounds of the invention will be known to the skilled artisan and are within the
scope of the invention.
Liquid drug formulations suitable for use with nebulizers and liquid spray
devices and EHD aerosol devices will typically include a compound of the invention
with a pharmaceutically acceptable carrier. Preferably, the pharmaceutically
acceptable carrier is a liquid such as alcohol, water, polyethylene glycol or a
perfluorocarbon. Optionally, another material may be added to alter the aerosol
properties of the solution or suspension of the compound. Preferably, this material
is liquid such as an alcohol, glycol, polyglycol or a fatty acid. Other methods of
formulating liquid drug solutions or suspension suitable for use in aerosol devices
are known to those of skill in the art (see, e.g., Biesalski, U.S. Pat. Nos. 5,112,598;
Biesalski, 5,556,611, which are herein incorporated by reference) A compound can
also be formulated in rectal or vaginal compositions such as suppositories or
retention enemas, e.g.t containing conventional suppository bases such as cocoa
butter or other glycerides.
In addition to the formulations described previously, a compound of the
invention can also be formulated as a depot preparation. Such long acting
formulations can be administered by implantation (for example subcutaneously or
intramuscularly) or by intramuscular injection. Thus, for example, the compounds
can be formulated with suitable polymeric or hydrophobic materials (for example, as
an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble
derivatives, for example, as a sparingly soluble salt.
Alternatively, other pharmaceutical delivery systems can be employed.
Liposomes and emulsions are well known examples of delivery vehicles that can be
used to deliver the compounds of the invention. Certain organic solvents such as
dimethylsulfoxide can also be employed, although usually at the cost of greater
toxicity. The compounds of the invention can also be delivered in a controlled
release system, hi one embodiment, a pump can be used (Sefton, CRC Crit. Ref
BiomedEng., 1987,14,201; Buchwald et al., Surgery, 1980,88,507; Saudeke et al.,
48

WO 2006/066080 PCT/US2005/04S589
N. Engl J. Med., 1989,321,574). In another embodiment, polymeric materials can
be used (see Medical Applications of Controlled Release, Langer and Wise (eds.),
CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product
Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger
and Peppas, J. Macromol. Sci. Rev. Macromol. Chetn., 1983,23, 61; see also Levy
et al, Science, 1985,228,190; During et at, Ann. Neurol, 1989,25,351; Howard et
at, 1989, J Neurosurg. 71,105). In yet another embodiment, a controlled-release
system can be placed in proximity of the target of the compounds of the invention,
e.g., the lung, thus requiring only a fraction of the systemic dose (see, e,g,, Goodson,
in Medical Applications of Controlled Release, supra, voL 2, pp. 115 (1984)). Other .
controlled-release system can be used (see, e.g. Langer, Science, 1990,249,1527).
Suitable excipients (e.g., carriers and diluents) and other materials that can
be used to provide mucosal dosage forms encompassed by this invention are well
known to those skilled in the pharmaceutical arts, and depend on the particular site
or method which a given pharmaceutical composition or dosage form will be
administered. With that fact in mind, typical excipients include, but are not limited
to, water, ethanol, ethylene glycol, propylene glycol, butane-l,3-diol, isopropyl
myristate, isopropyl palmitate, mineral oil, and mixtures thereof, which are non-
toxic and pharmaceutically acceptable. Examples of such additional ingredients are
well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 18th eds.,
Mack Publishing, Easton PA (1990).
The pH of a pharmaceutical composition or dosage form, or of the tissue to
which the pharmaceutical composition or dosage form is applied, can also be
adjusted to improve delivery of one or more active ingredients. Similarly, the
polarity of a solvent carrier, its ionic strength, or tonicity can.be adjusted to improve
delivery. Compounds such as stearates can also be added to pharmaceutical
compositions or dosage forms to advantageously alter the hydrophilicity or
lipophilicity of one or more active ingredients so as to improve delivery. In this
regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying
agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent
Different salts, hydrates or solvates of the active ingredients can be used to further
adjust the properties of the resulting composition.
KITS
49

WO 2006/066080 PCT/US2005/045589
The invention provides a phannaceutical pack or kit comprising one or more
containers comprising a Formula I prodrug, Formula II compound, or other
compound of the invention useful for the treatment or prevention of a Hepatitis C
virus infection. In other embodiments, the invention provides a pharmaceutical pack
or kit comprising one or more containers comprising a compound of the invention
useful for the treatment or prevention of a Hepatitis C virus infection and one or
more containers comprising an additional therapeutic agent, including but not
limited to those listed above, in particular an antiviral agent, an interferon, an agent
which inhibits viral enzymes, or an agent which inhibits viral replication, preferably
the additional therapeutic agent is HCV specific or demonstrates anti-HCV activity.
The invention also provides a pharmaceutical pack or kit comprising one or
more containers comprising one or more of the ingredients of the pharmaceutical
compositions of the invention. Optionally associated with such container(s) can be a
notice in the form prescribed by a governmental agency regulating the manufacture,
use or sale of Pharmaceuticals or biological products, which notice reflects approval
by the agency of manufacture, use or sale for human administration.
The inventive agents may be prepared using the reaction routes and synthesis
schemes as described below, employing the general techniques known in the art
using starting materials mat are readily available. The synthesis of non-exemplified
compounds according to the invention may be successfully performed by
modifications apparent to those skilled in the art, e.g., by appropriately protecting
interfering groups, by changing to other suitable reagents known in the art, or by
making routine modifications of reaction conditions. Alternatively, other reactions
disclosed herein or generally known in the art will be recognized as having
applicability for preparing other compounds of the invention.
Preparation of Compounds
In the synthetic schemes described below, unless otherwise indicated all
temperatures are set forth in degrees Celsius and all parts and percentages are by
weight. Reagents were purchased from commercial suppliers such as Aldrich
Chemical Company or Lancaster Synthesis Ltd. and were used without further
purification unless otherwise indicated. Tetrahydrofuran (THF) and N, N-
dimethylforamide (DMF) were purchased from Aldrich in Sure Seal bottles and used
as received. Unless otherwise indicated, the following solvents and reagents were
50

WO 2006/066080 PCT/US2005/045589
distilled under a blanket of dry nitrogen. THF, and EtaO were distilled from Na-
benzophenone ketyi; CH2CI2, diisopropylamine, pyridine and Et3N were distilled
from CaH2; MeCN was distilled first fromPaOs, then from CaHb; MeOH was
distilled from Mg; PhMe, EtOAc and i-PrOAc were distilled from CaH2; TFAA was
purified via simple atmospheric distillation under dry argon.
The reactions set forth below were done generally under a positive pressure
of argon at an ambient temperature (unless otherwise stated) in anhydrous solvents,
and the reaction flasks were fitted with rubber septa for the introduction of substrates
and reagents via syringe. Glassware was oven dried and/or heat dried. The
reactions were assayed by TLC and terminated as judged by the consumption of
starting material. Analytical thin layer chromatography (TLC) was performed on
aluminum-backed silica gel 60 F254 0.2 mm plates (EM Science), and visualized
with UV light (254 nm) followed by heating with commercial ethanolic
phosphomolybdic acid. Preparative thin layer chromatography (TLC) was
performed on aluminum-backed silica gel 60 F2541.0 mm plates (EM Science) and
visualized with UV light (254 nm).
Work-ups were typically done by doubling the reaction volume with the
reaction solvent or extraction solvent and then washing with the indicated aqueous
solutions using 25% by volume of the extraction volume unless otherwise indicated.
Product solutions were dried over anhydrous Na2SO4 and/or Mg2SC>4 prior to
filtration and evaporation of the solvents under reduced pressure on a rotary
evaporator and noted as solvents removed in vacuo. Column chromatography was
completed under positive pressure using 230-400 mesh silica gel or 50-200 mesh
neutral alumina. Hydrogenolysis was done at the pressure indicated in the examples
or at ambient pressure.
-NMR spectra were recorded on a Varian Mercury-VX400 instrument
operating at 400 MHz and 13C-NMR spectra were recorded operating at 75 MHz.
NMR spectra were obtained as CDCI3 solutions (reported in ppm), using chloroform
as the reference standard (7.27 ppm and 77.00 ppm), CD3OD (3.4 and 4.8 ppm and
49.3 ppm), DMSO-d6, or internally tetramemylsilane (0.00 ppm) when appropriate.
Other NMR solvents were used as needed. When peak multiplicities are reported,
the following abbreviations are used: s (singlet), d (doublet), t (triplet), q (quartet),
51

WO 2006/066080 PCT/US2005/045589
m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets).
Coupling constants, when given, are reported in Hertz (Hz).
Infrared (IR) spectra were recorded on a FT-IR Spectrometer as neat oils, as
KBr pellets, or as CDCI3 solutions, and when given are reported in wave numbers
(cm"1). Mass spectra reported are (+)-ES LC/MS conducted by the Analytical
Chemistry Department of Anadys Pharmaceuticals, Inc. Elemental analyses were
conducted by the Atlantic Microlab, Inc. in Norcross, GA. Melting points (mp)
were determined on an open capillary apparatus, and are uncorrected.
The described synthetic pathways and experimental procedures utilize many
common chemical abbreviations, THF (tetrahydrofuran), DMF (N,N-
dimethylformamide), EtOAc (ethyl acetate), DMSO (di-methyl sulfoxide), DMAP
(4-dimethylaminopyridine), DBU (l,8-diazacyclo[5.4.0]undec-7-ene), DCM (4-
(dicyanomemylene)-2-memyl-6-(4-dimemylamino-styryl)-4 ff-pyran), MCPBA (3-
chloroperoxybenzoic acid), EDC (l-(3-dimethylaminopropyl)-3-ethylcarbodiirm'de
hydrochloride), HATU (0-(7-azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronium
hexafluorophosphate), HOBT (1-hydroxybenzotriazole hydrate), TFAA
(trifluoroacetic anhydride), pyBOP (benzotriazol-1-
yloxy)tripyrrolidinophosphonium hexafluorophosphate), DIEA
(diisopropylethylamine), BOC (tert-butoxycarbonyl), 2,2-DMP (2,2-
dimethoxypropane), IPA (isopropyl alcohol), TEA (triethylamine), DCE (1,2-
dichloroethane), PPTS (pyridinium p-toluenesulfonate), DEAD
(diethylazodicarboxylate), PS (polymer supported), HF (hydrogen fluoride), MeCN
(acetorritrile), MeOH (methanol), Val (valine), Phe (phenyl alanine), HPLC (high
pressure liquid chromatography), TLC (thin layer chromatography), Bz (benzoyl),
Ac (acetyl), Tol (toluoyl), Me (methyl), and the like.
52
Example 1
5-Amino-3-(2'-C-methyl-b-D-ribofiiranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one


WO 2006/066080 PCT/US2005/045589
Step 1) Preparation of 5-Amino-3-(2'-C-methyl-2',3',5'-tri-0-benzoyt-b-D~
ribofuranosyl)-3H-thiazolo[4,5~d]pyrimidin-2-one(S)

a. BSA, MeCN, it; TMSOTf, 80°C, 67%
To a heterogeneous mixture of heterocycle 1 (168 nag, 1.00 mmol) and
perbenzoyl ribose 2 [prepared according to the method of Wolfe et al. J. Org. Chem.
1997,62,1754-1759] (522 mg, 0.90 mmol) in anhydrous MeCN (10 mL) was added
BSA (742 JJL, 3.00 mmol). The resultant mixture was stirred 15 min whereupon
TMSOTf (333 pL, 1.50 mmol) was added. The reaction mixture was stirred for 4 h
at 65° C, Ihen 3 h at 90° C. The mixture was then cooled to rt, diluted with DCM
(150 mL) and partitioned with pH 7 buffer (100 mL). The aqueous phase was further
extracted with DCM (3 x 50 mL), and the combined organic phases were dried over
Na2SO4 and filtered. The clear filtrate was diluted with EtOAc (200 mL), filtered
through a short pad of SiO2, concentrated and submitted to flash chromatography
(10-40% EtOAc-DCM), affording 380 mg (67%) of nucleoside 3 as a white solid:
*H (400 MHz, DMSO-d6) 5 8.43 (s, 1H), 7.85-8.02 (m, 6H), 7.46-7.97 (m, 7H), 7.35
(t, J= 8.06,2H), 6.96 (br s, 2H), 6.77 (br s, 1H), 4.54-4.82 (m, 4H), 1.77 (s, 3H);
[M+H]+m/z 627.
Step 2) Preparation of 5-Amino-3-(2'~C-methyl-b~D-ribofijranosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-one(4)

b.K2CO3, MeOH,10%.
To a suspension of nucleoside 3 (380 mg, 0.606 mmol) in MeOH (20 mL)
was added K2CO3 (17 mg, 0.12 mmol) at rt. The resulting mixture was stirred 18 h
at rt whereupon it was treated with HO Ac (15 uL, 0.25 mmol), concentrated and
53

WO 2006/066080 PCT/US2005/045589
submitted to HPLC purification (MeCN-H2O), affording 20 mg (10%) of the title
compound 4 as a white solid after lyophilization: !H (400 MHz, DMSO-d6) 8 8.33
(s, 1H), 6.86 (br s, 2H), 6.09 (br s, 1H), 5.19 (br s, 1H), 4.88 (br s, 1H), 4.55 (t, J=
5.87,1H), 3.97 (br s, 1H), 3.76-3.81 (m, 1H), 3.61 (br s, 2H), 1.04 (s, 3H); [M+H]+
m/z 315. Analysis calc'd for C11H14O5SH2O: C, 39.75; H, 4.85; N, 16.86; S, 9.65.
Found: C, 40.23; H, 4.76; N, 16.64; S, 9.45.
Example 2
5-Atnino-3-(2'-deoxy-b-ribofurmosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one

Step 1) Preparation of N'-(7-Chloro-2-oxo-2,3-dihydro~thiazolo[4,5-d]pyrimidin-5-
yl)-N,N-dimethyl-formamidine (7)

To a suspension of 5-arnino-7-hydroxy-3H-thiazolo[4,5-d]pyrimidin-2-one
(10.0 g, 53.7 mmol) in MeCN (200 mL) at 0° C was added SOC12 (20.0 mL, 274
mmol) dropwise via addition funnel over 20 min. The resulting mixture was slowly
warmed to rt then immersed into a 60° C oil bath where it was stirred for 48 h. The
reaction mixture was cooled to rt and slowly poured into 300 g of cracked ice in 300
mL of water containing NaHCO3 (46 g, 548 mmol). The aqueous mixture was
extracted with 20% IPA-DCM (3 x 500 mL), and the combined organic phases were
dried over Na2SO4 and concentrated to a residue that was triturated with EtOAc to
afford 6.33 g (46%) of chloroamidine 7 as a tan solid: lH (400 MHz, DMSO-d6) 5
12.60 (s, 1H), 8.69 (s, 1H), 3.25 (s, 3H), 3.11 (s, 3H); [M+H]+ m/z 258.
54

WO 2006/066080 PCT/US2005/045589
Step 2) Preparation of N'-(7-Chloro-2-oxo-3-[2'-deoxy-3',5'-di-O-(p-toluoyl)-b-D-
ribofuranosyl]-2,3-dihydro-thiazolo[4,5-d]pyrimidin-5-yl)-N,N-diinethyl-
formamidine (8)

a. 6, NaH, MeCN, 90%.
To a suspension of heterocycle 7 (1.79 g, 6.94 mmol) in anhydrous MeCN
(90 mL) at rt was added 95% NaH (183 mg, 7.63 mmol). The resulting mixture was
stirred 30 min whereupon chlorosugar 5 (2.70 g, 6.94 mmol) [purchased from Berry
& Associates, Inc., Dexter, MI] was added. The reaction mixture was heated to 55°
C, stirred lh, cooled, concentrated, and then submitted to flash chromatography
(SiO2, 5-10% EtOAc-DCM) affording 3.8 g (90%) of nucleoside 8 as a solid
material that may be further purified via trituration in MeOH: *H (400 MHz,
DMSO-d6) 5 8.64 (s, 1H), 7.83 (ABq, JAB = 8.19, AVAB = 38.53,4H), 7.28 (ABq, JAB
= 8.19, AVAB = 36.13,4H), 6.56 (dd, J= 8.19,5.07,1H), 5.76-5.80 (m, 1H), 4.56-
4.60 (m, 1H), 4.45-4.50 (m, 2H), 3.27-3.34 (m, 1H), 3.15 (s, 3H), 3.03 (s, 3H), 2.57-
2.64 (m, 1 H), 2.35 (s, 3H), 2.39 (s, 3H).
Step 3) Preparation of5-Amino-3-(2',3'-di-0-(p-toluoyl)-p-D-ribofnranosyl)-
3H-thiazolo[4,5-d]pyrimidin-2-one(9)

b. Zn-Cu, HOAo, 67%.
To a solution of chloroaryl nucleoside 8 (924 mg, 1.47 mmol) in acetic acid
(10.4 mL) at rt was added Zn-Cu couple (1.54 g, 11.9 mmol). The resulting
suspension was stirred vigorously at ambient temperature for 3.5 h, filtered through
celite, and then concentrated to a solid material that was submitted to flash
55

WO 2006/066080 PCT/US2005/045S89
chromatography (SiO2, 0-10% EtOAc-CHCl3), yielding 520mg (67%) of compound
9 as a tan solid: 1'H NMR (400 MHz, de-DUSO) 8 8.34 (s, 1H), 7.85 (ABq, JAB =
8.4, AVAB = 17.6,4H), 7.30 (ABq, JAM = 8.4, AVAB = 27.1,4H), 6.87 (s, 2H), 6.47(m,
1H), 5.78 (m, 1H), 4.62 (m, 1H), 4.47 (m, 2H), 3.35 (m, 2H), 2.38 (s, 3H), 2.35 (s,
3H).
Step 4) Preparation of 5-Amino-3-(2'-deoxy-p-D-ribofuranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (10)

c. K2CO3, MeOH, 46%.
To a suspension of diester 9 (300 mg, 0.577 ramol) from Step 3 (above) in
MeOH (20 mL) at rt was added K2CO3 (188 mg, 1.36 mmol). The resulting mixture
was stirred 8 h whereupon it was quenched with HO Ac (164 {XL, 2.86 mmol), then
concentrated and submitted to HPLC (MeCN-H2O, TFA) to afford 75 mg (46%) of
the title compound 10 as a white solid (TFA salt) after lyophilization: 1H (400 MHz,
DMSO-D6) 5 8.34 (s, 1H), 7.15 (br s, 2H), 6.33 (t, J = 7.0,1H), 4.33 (br s, 1H), 3.72-
3.73 (m, 1H), 3.39-3.56 (m, 2H), 2.89-2.96 (m, 1H), 1.99-2.05 (m, 1H); [M+H]+»vfe
285. Analysis calc'd for C11H12NS-CaHFaOz: C, 36.18; H, 3.29; N, 14.31; S,
8.05. Found: C, 36.28; H, 3.35; N, 13.96; S, 8.05.
Example 3
5-Amino-3-(2'-O-acetyl-3'-deoxy-j3-D-ribojuranosyl)-3H-thiazolo[4,5-d]pyrimidin-

Step 1) Preparation cf5-Aniino-3-(25'-di-0-benzoyl-3'-deoxy-frD-ribofuinnosyl)~
3H-thiazolo[4,5-d]pyrimidin-2-one (12)
56

WO 2006/066080 PCT/US2005/045589

. a. 11a, BSA, MeCN, rt; TMSOTf, 80°C, 78%.
To a heterogeneous mixture of heterocycle 1 (25.0 g, 0.149 mol) and
deoxyribofuranose 11a (47.0 g, 0.122 mol) [may be prepared from the
corresponding methyl ribofuranoside (Walton, et, al. J. Med. Cliem. 1965,8,659-
663) via the method of Valdivia, et. al. Tetrahedron Lett. 2005,46,6511-6514] in
anhydrous MeCN (640 mL) at RT was added dropwise via addition funnel BSA
(113 mL, 0.458 mol) over 20 min. The resultant suspension was treated dropwise
with TMSOTf (41.5 mL, 0.229 mol) at rt over 20 min, whereupon it became nearly
homogeneous. The mixture was heated to reflux (internal T 83° C) and stirred for 8
h, then cooled to rt and concentrated to an oily residue via rotary evaporation. The
residue was dissolved in EtOAc (500 mL) and cooled to 10° C where it was slowly
treated with 1M pH 7 phosphate buffer (400 mL), keeping the internal temperature
below 35° C. Upon completed addition of buffer, the pH of the mixture was adjusted
to 7.0 with solid K2HPO4 and vigorous stirring was continued for 1 h. Celite (25 g)
was added, and the mixture stirred an additional 30 min. Filtration of the triphasic
mixture through a short pad of celite provided two clear phases. The aqueous phase
was saturated with solid NaCl then extracted with EtOAc (4 x 250 mL). The
combined organic phases were washed with brine (400 mL), dried over Na2SO4 and
charcoal (1 g), and then filtered through a short pad of S1O2. The clear amber filtrate
was concentrated to dryness, whereupon solid heterocycle had precipitated. The
residue was taken up in DCM, treated with a small amount of MgSO4, and then
filtered through celite. The clear filtrate was concentrated and further dried under
high vacuum at 35° C to provide a tan, crispy foam (69.5 g). Submission of this
. solid foam to flash chromatography (SiCh, 5-40% EtOAc-hexanes) afforded 46.3 g
(77%) of nucleoside 12 as a light beige solid foam: *H NMR (DMSO-4) 5 8.35 (s,
1H), 7.93-8.01 (m, 4H), 7.61-7.69 (m, 2H), 7.47-7.56 (m, 4H), 6.94 (s, 2H), 6.09 (d,
J = 1.9,1H), 6.00 (d, / = 7.4,1H), 4.64-4.69 (m, 1H), 4.57 (dd, / = 12.1,2.7,1H),
57

WO 2006/066080 PCT/US2005/045589
4.36 (dd, J = 12.1,5.8,1H), 2.92-3.00 (m, 1H), 2.32 (dd, /= 14.0,5.8,1H); [M+H]+
m/z 493.
Step 2) Preparation of 5-Amino-3-(3'~deoxy-j3-D-ribofuranosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-one(13)

b. K2CO3, MeOH, 78%.
To a heterogeneous mixture of dibenzoate 12 (46.3 g, 94.0 mmoJ) and
anhydrous MeOH (1.0 L) was added K2CO3 (2.59 g, 18.8 mmol) at rt. The mixture
became homogeneous within 30 min, then heterogeneous again within 3 h.
Additional MeOH (100 mL) was added to increase fluidity, and the reaction mixture
was stirred for a total of 24 h. The suspension was treated with HOAc (2.26 mL,
39.5 mmol) and then concentrated at 45° C whereupon it was cooled, then triturated
with EtOH (200 mL) and ether (1800 mL) for 1 h. The solid material was filtered,
washed with ether (3 x 250 mL), air dried and then washed with water (2 x 250 mL),
affording 19.47 g (78%) of diol 13 as a white solid that was dried in vaccuo and
recrystallized from water: *H NMR (DMSO-d6) 8 8.31 (s, 1H), 6.82 (s, 2H), 5.82 (d,
1H), 5.41 (d, 1H), 4.79-4.83 (m, 1H), 4.65 (t, J - 5.8,1H), 4.13-4.20 (m, 1H), 3.40-
3.49 (m, 2H), 2.31 (ddd, J= 16.0,9.4,7.0,1H), 1.81 (ddd, /= 12.5,5.8,2.3,1H);
[M+Hf m/z-285. Analysis calc'd for C10H12N4O4S: C, 42.25; H, 4.25; N, 19.71; S,
11.28. Found: C, 42.36; H, 4.32; N, 19.72; S, 11.23.
Step 3) Preparation of5-Amino-3-(2',5'-di-O-acetyl-3'-deoxy-j3-D-
ribofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (14)

c. Ac2O, Et3N, DMAP, MeCN. t. 1, BSA, MeCN, rt; TMSOTf, 80°C.
To a suspension of diol 13 (8.00 g, 28.1 mmol), Et3N (11.8 mL, 84.4 mmol),
and DMAP (344 mg, 2.81 mmol) in anhydrous MeCN (190 mL) at 0° C was added
58

WO 2006/066080 PCT/US2005/045589
dropwise AC2O (5.44 mL, 57.7 mmol). The resultant mixture, that became
homogeneous within 1.5 h, was slowly warmed to it and stirred for 18 h whereupon
it was concentrated to a residue that was submitted to flash chromatography (SiO2,
0-100% EtOAc-DCM) to afford 8.34 g (80%) of diacetate 14 as a white solid foam
that may be further purified via trituration with ether-hexanes: !H (400 MHz,
DMSO-40 6 8.34 (s, 1H), 6.91 (s, 2H), 5.90 (d, J = 1.9,1H), 5.65 (d, J = 7.4,1H),
4.33-4.39 (m, 1H), 4.25 (dd, J= 12.1,3.1,1H), 4.01 (dd,/= 11.7, 6.6,1H), 2.65-
2.73 (m, 1H), 2.06 (dd, J= 13.6,6.2,1H), 2.05 (s, 3H), 1.98 (s, 3H); [M+H]+ m/z
369. Analysis calc'd for C14RAS: C, 45.65; H, 4.38; N, 15.21; S, 8.70. Found:
C, 45.69; H, 4.52; N, 15.02; S, 8.64.
Alternatively diacetate 14 may be prepared from heterocycle 1 and
deoxyribo-furanose 11b [may be prepared via the method of Valdivia, et. al.
Tetrahedron Lett. 2005, 46,659-663] in a manner similar to Step 1 above with a
yield of 63%.
Step 4) Preparation of 5-Amino-3-(2'-0~acetyl-3'-deoxy-fi-D-ribofuranosyl)-3H-
thiazolo[4,5-djpyrimidin-2'one(15)

d. Candida Aretha, Acetone, pH 7 buffer 91 %.
To a slowly stirring suspension of diacetate 14 (5.08 g, 13.8 mmol) and
Candida Arctica lipase acrylic resin (2.50 g) [purchased from Sigma] in acetone (50
mL) was added 50 mM pH 7 phosphate buffer (250 mL) at rt The resulting mixture
was stirred slowly for 18 h whereupon it was filtered, concentrated and extracted
with EtOAc (4 x 250 mL). The combined organic phases were dried over Na2SO4,
concentrated and then submitted to flash chromatography (SiO2, 0-15% 1PA-DCM)
to afford 4.11 g (91%) of the title compound 15 as a white solid that may be further
purified via trituration with ether-hexanes: lH (400 MHz, DMSO-d6) 5 8.33 (s, 1H),
6.87 (s, 2H), 5.88 (d, / = 2.3,1H), 5.66 (d, / = 7.8,1H), 4.76 (t, J = 5.8,1H), 4.11-
4.18 (m, 1H), 3.43-3.53 (m, 2H), 2.50-2.57 (m, 1H), 2.05 (s, 3H), 1.98 (dd, J = 13.6,
5.8,1H); [M+H]+ m/z 327. Analysis calc'd for CHOsS: C, 44.17; H, 4.32; N,
17.17; S, 9.83. Found: C, 44.12; H, 4.45; N, 16.88; S, 9.69;
59

WO 2006/066080 PCT/US2005/045589
Example 4
5-Amino-3-(2'-0-acetyl-5'-0-benzoyl-3'-deoxy-b-D-ribofuratiosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-one(16}



(t 11c, BSA, MeCN, rt; 80°C.
To a heterogeneous mixture of heterocycle 1 (106 mg, 0.633 mmol) and
deoxyribofuranose 11c [purchased from Berry & Associates, Inc., Dexter, MI] (183
mg, 0.57 mmol) m anhydrous MeCN (8 mL) was added BSA (464 uL, 1.89 mmol)
at it The resultant mixture was immersed into a 60° C oil bath and stirred for 4 h.
The mixture was concentrated via rotary evaporation and partitioned between
EtOAc (100 mL) and saturated NaHCO3 (50 mL). The organic phase was dried over
Na2SO4 and then concentrated. The crude material was triturated with Et2O-EtOAc
to afford 132 mg (54%) of an off-white solid that was submitted to HPLC (MeCN-
H2O) to provide an analytical sample: 1H (400 MHz, DMSO-d6) 8 8.33 (s, 1H),
7.91-7.94 (m, 2 H), 7.60-7.65 (m, 1H), 7.47-7.50 (m, 2H), 6.94 (s, 2H), 5.93 (d, J=
1.8,1H), 5.72 (dd, J= 5.9,1.5,1H),4.50-4.53 (m, 2H), 4.31 (q, J=7.0,1H), 2.80-
2.88 (m, 1H), 2.14 (dd, J= 13.2,5.1,1H), 2.07 (s, 3H); [M+Hf m/z 431.
Example 5
5-Ammo-3-benzyl-3H-thiazolo[4,5-d]pyrimidin-2-one(lS)

a. POCI3, DMF, 90°C. b. Guanidine HCI, NaOMe, MaOH, 19% over 2 steps
To POCI3 (8.09 mL, 86.8 mmol) at 0° C was added consecutively solid 3-
benzyl-thiazolidine-2,4-dione [prepared according to the method of Lo, et al. /.
Org. Chem. 1957,999-1001] and DMF. The reaction mixture was stirred 5 min, then
transferred to a 90° C oil bath where it was stirred 3 h. The dark mixture was poured
60


WO 2006/066080 PCM/US2005/045589
into ice (100 g) and water (100 mL), and then extracted with EtOAc (3 x 100 mL).
The combined organic phases were dried over MgSO4 then filtered through celite,
concentrated, taken up in DCM, filtered through a short pad of SiO2 and finally
concentrated to a dark oil that was used without further purification.
Guanidine hydrochloride (8.87 & 92.8 mmol) was added as solid to 25%
NaOMe-MeOH (18 mL, 79.6 mmol) in MeOH (48 mL) at -5° C. The resulting
mixture was stirred 5 min and then treated with erode chloroaldehyde [from above]
as a solution in MeOH (20 mL). The reaction mixture was placed into a 90° C oil
bath, concentrated by distillation of the MeOH over 2 h, and then heated an
additional 30 min. The residue was taken up in EtOAc (200 mL) and partitioned
with 1 N HC1 (100 mL). The aqueous phase was treated with solid NaHCO3 and
partitioned with EtOAc (3 x 100 mL). The combined organic phases from the
alkaline extraction were dried over Na2SO4 and then concentrated to a residue that
was submitted to chromatography (SiOz, 50-70% EA-DCM), affording 1.4 g (19%)
of the title compound 18 as a white solid: XH (400 MHz, DMSO- 7.24-7.34 (m, 5H), 6.83 (s, 2H), 5.01 (s, 2H); [M+H]+ m/z 259.
Example 6
5-Amim-3~(3\3'-C,0-dimethyl-ftD-ribojuranosyl)-3H-thiazolo[4,5~d]pyrimidin-2

Step 1) Preparation of l,2-O-hopropylidene-3-methyl-3-.O-methyl-5~O-trityl-(a-D-
ribofuranose (20)

a KH (xs), dioxane, DMF (3:1); Mel, it, 72 h, 87%
To a mixture of tertiary alcohol 19 (716 mg, 1.60 mmol) [prepared according
25 to the method of Just et al. Tetrahedron Lett. 2000,41,9223-9227] in anhydrous
dioxane (6 mL) was added an excess of KH (30% dispersion in mineral oil) at rt.
61

WO 2006/066080 PCT/US2005/045589
The resulting mixture was stirred 1 h and was then treated with Mel (2 mL, 32
mmol), producing a copious precipitate. DMF (2 mL) was added to keep the reaction
mixture fluid enough for stirring over 72 h. The mixture was diluted with EtOAc
(100 mL) and partitioned with saturate aqueous NaHCO3 (50 mL). The organic
phase was dried over Na2SO4, concentrated and submitted to flash chromatography
(SiO2,10-60% EtOAc-hexanes), affording 640 mg of methyl ether 20 as a white
solid: 1H (400 MHz, DMSO-d6) 5 7.22-7.35 (m, 15 H), 5.72 (d, J = 2.9,1H), 4.30
(d, J = 2.9,1H), 4.07 (dd, J = 7.7,2.6,1H), 3.14 (s, 3H), 2.92-3.05 (m, 2H); 1.51 (s,
3H), 1.28 (s, 3H), 0.82 (s , 3H).
Step 2) Preparation of l,2,5-tetra-0-Acetyl~3,3~C,0-dimethyl-j3-D-ribofu.ranose (21)

b. HOAC, Ac2O, H2SO4, rt, 33%.
To a solution of HOAc (50.0 mL) and Ac2O (3.83 mL, 40.6 mmol) was
added furanose 20 (3.62 g, 8.11 mmol). To this colorless solution was added 1 M
H2SO4 in HOAc (0.41 mL, 0,41 mmol), resulting in an intensely yellow colored
solution. The solution was stirred at rt for 16 lir, and then concentrated via rotary
evaporation. The excess HOAc was azeotroped via several volumes of toluene. The
residue was dissolved in DCM (100 mL) and washed with saturated NaHCO3 (30
mL). The organic layer was dried over Na2SO4, decanted, and concentrated to an
oily heterogenous mixture. This mixture was subjected to flash chromatography
(SiO2,5-35% EtOAc-hexanes) affording 0.78 g (33%) of triacetate 21 as a pale
yellow oil: 1H (400 MHz, DMSO-4) 8 5.91 (d, /= 2.0,1H), 5.07 (d, /= 2.4,1H),
4.29 (dd,/ = 3.2,12.0,1H), 4.15 (dd, J - 3.2,7.2,1H), 3.96 (dd, 7 =6.8,12.0,1H),
3.17 (s, 3H), 2.10 (s, 3H), 2.04 (s, 6H), 1.33 (s, 3H).
Step 3) Preparation of 5-Amino-3-(2',5'di-O-acetyl-3',3'-C,O-dimethyl-b-D-
ribojuranosyl)-3H~thiazolo[4,5-d]pynmidin-2-one (22)
62

WO 2006/066080 PCT/US2005/045589



c. BSA, MeCN, rt; TMSOTf, 80°C, 65%.
To a mixture of heterocycle 1 in anhydrous MeCN (5.0 mL) was added
dropwise BSA (0.52 mL, 2.11 mmol) at rt. The resulting mixture was immersed in a
40° C oil bath, and stirred for 90 min whereupon it became homogenous. Furanose
21 (0.20 g, 0.73 mmol) was added followed by TMSOTf (158.4 uL, 0.88 mmol).
The reaction mixture was then immersed in an 80° C oil bath and heated for 2hr.
The reaction was cooled to rt, and partitioned between 1 M pH 7 phosphate buffer
(15 mL) and EtOAc (30 mL). The resulting emulsion was filtered through a pad of
celite yielding two distinct layers. The organic layer was separated and dried over
Na2SO4, filtered, and concentrated in vacuo to a dark orange brown residue. This
residue was submitted to flash chromatography (S1O2,5-50% EtOAc-hexanes)
purification affording 0.30g (59%) of nucleoside 22 as a finely divided pale yellow
solid: *H (400MHz, DMSO-4) § 8.34 (s, 1H), 6.90 (br s, 2H), 6.11 (dd, / = 8.0,
27.6,2H), 4.3-4.17 (m, 3H), 2.75 (s, 3H), 2.02 (s, 3H), 2.01 (s, 3H), 1.35 (s, 3H);
[M+H]+m/z413.

Step 4) Preparation of 5-Amino-3-(3',3'-C,0-dimethyl-j3-D-ribofuranosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-om(2$)
d. K2CO3, MeOH, rt.
Nucleoside 22 (230 mg, 0.56 mmol) was dissolved in 5.6 mL MeOH. Solid
K2CO3 (15.4 mg, O.11 mmol) was added and the reaction stirred at rt for 16 h. HOAc
(16.0 ul, 0.28mmol) was added and the reaction stirred for 30 min, and then
concentrated mixture to dryness in vacuo. The residual yellow solids were triturated
63

WO 2006/066080 PCT/US2005/045589
with Et2O (3x10 mL) carefully decanting filtrates via pipette. The solid material
was then washed with H2O (3 x 5mL), rinsed with Et2O (2x5 mL) and dried on
house vacuum 24 h to obtain 76.4 mg (42%) of a finely divided white solid: 1H
(400MHz, DUSO-d6) § 8.35 (s, 1H), 6.83 (br s, 2H), 5.93 (d, 7 = 8.58,1H), 5.24 (d,
7 = 7.02,1H), 4.95 (t, 7 = 7.4,1H), 4.57 (t, 7 = 5.46,1H), 3.95 (t, 7 = 5.07,1H),
3.46-3.58 (m, 2H), 3.26 (s, 3H), 1.29 (s, 3H); [M+H]+ m/z 329.
Example 7
5-Amino-3-(5'-0-Acetyl-2'-0-[2"-0-acetylpropyl]-3-methyl-frD-ribofuranosyl)-
3H'thiazob[4,5-dJpyrinudin-2-one (27)

Step 1) Preparation of l,2-0-Isopropylidine-3-methyl-5-0-trityl~os-D-ribofuranose
(25)
a. H2,5% Pd/C, EtOAc, rt, 48hr, 6:1 (o/p), 78%.
To a solution of 24 (2.40g, 5.60 mmol) [prepared according to Bera, et al.
Helvetica ChimicaActa 2000,83(7), 1398-1407] dissolved in EtOAc (50 mL),
under a blanket of N2, was added 5% Pd/C (240 mg). The flask was charged with H2
at 1 arm, and stirred at rt for 72 h. The reaction mixture was filtered through celite,
and concentrated invacuo to a clear colorless oil. Flash chromatography purification
(SiO2,0-40% EtOAc-hexanes) afforded 1.88g of 25 (78%) as a 6:1 (ot/p) mixture of
isomers: JH (400MHz, DMSO-40 8 7.22-7.38 (m, 15H), 7.63 (d, 7 = 3.2,1H), 4.53
(t, /=4.0,1H), 3.67 (dq, 7= 2.8,1.6,1H), 3.18 (dd, 7=3.2,10.4,1H), 2.99 (dd, 7 =
5.2,10.8,1H), 1.89-1.98 (m, 1H), 1.38 (s, 3H), 1.24 (s, 3H), 0.81 (d, 7 = 6.8,3H).
Step 2) Preparation ofl,5~di-0~Acetyl-2-O-(2'-0-acetylpropyl)-3-methyl-fi-D~
ribofuranpse (26)
64

WO 2006/066080 PCT/US2005/045589

b. HOAc, Ac2O, H2SO4, rt, 19%.
In. a manner similar to Step 3 of Example 6,25 was converted to 26 in a 19%
yield. The crude residue was submitted to flash chromatography (S1O2,2-30%
EtOAc-hexanes), yielding a 5:1 (p/oc) mixture of anomers (tainted with
triphenylmethane) by !H NMR (DMSO-Js) that was used without further
purification in the next step: !H (400MHz, DMSO-d6) 5 6.07 (d, / = 2.8,1H), 5.02
(ddd, /= 6.8,6.8,2.8,1H), 4.25 (dd, /= 12.0,3.2,1H), 4.06-4.21 (m, 2H), 2.14-
2.23 (m, 1H), 2.03 (s, 3H), 2.00 (s, 3H), 1.99 (s, 3H), 1.41 (s, 3H), 1.39 (s, 3H), 0.91
(d,/=6.8,3H).
Step 3) Preparation of5-Amino-3-(5'-0-Acetyl-2'-0-[2"-0-acetylpropyl]-3-methyl-
0-D-ribofiiranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-om (27)

0. BSA, MeCN, rt; TMSOTf, 60°C, 11%.
To a mixture of the heterocycle 1 in anhydrous MeCN (5.0 mL) at rt was
added dropwise BSA (0.52 mL, 2.11 mmol). The resulting mixture stirred for 30
min, and then furanose 26 (0.20 g, 0.73 mmol) was added followed by TMSOTf
(158.4 uL, 0.88 mmol). The reaction mixture was immersed in a 60° C oil bath,
whereupon it became a homogenous solution. Stirring was continued for 2.5 h. The
reaction mixture was cooled to rt, then partitioned between 1M pH 7 phosphate
buffer and EtOAc. The mixture was filtered through a pad of celite and the distinct
layers were separated. The organic phase was dried over Na2SO4, filtered,
concentrated and submitted to flash chromatography (SiO2,5-50% EtOAc-hexanes),
affording 30.0 mg of the title compound 27 as a white solid: JH (400MHz, DMSO-
d6) § 8.36 (s, 1H), 6.90 (s, 2H), 6.09 (d, / = 7.2,1H), 5.16 (t, j = 7.2,1H), 4.88 (dt, /
65

WO 2006/066080 PCT/US2005/045589
= 6.4,3.2 1H), 4.20 (dd, J= 3.2,12.0,1H), 4.08 (dd, J= 6.4,12.0,1H), 2.28 (dq, J=
6.8,14,1H), 1.97 (s, 3H), 1.87 (s, 3H), 1.54 (s, 3H), 1.41 (s, 3H), 0.86 (d, / = 6.8,
3H);[M+H]+ m/z 441.
Example 8
5 5-Amino-3-(3 '-methyl-P-D-ribofuransyl)-3H-thiazolo[4,5-d]pyiimidin-2-one (30)

Step 1) Preparation of5-Amino-3-(3'-0-acetyl-2',5'-di-0~benzoyl-3'-meikyl-$-D-
ribofuranosyl)-3H-thiazolo[4,5-d]pyrimidm-2-one (29)

a. Heterocycle 1, BSA, TMSOTf, GH3CN, 88%.
5-Amino-3H-thiazolo[4,5-d]pyrimidin.-2-one (123 mg, 0.733 mmol), 3'-C-
methyl-ribofuranose 28 [prepared according to the method of Wang et al. / Med.
Chem. 2000, 43, 3704-3713] (302 mg, 0.66 mmol), BSA (447 mg, 2.2 mmol) and
MeCN (8 mL) were mixed vigorously for 30 min until a homogeneous solution was
obtained. The reaction was then charged with TMSOTf (0.186 mL, 1.1 mmol) and
placed into a preheated oil bath at 65° C. After 3 h the reaction was cooled to rt and
the solvent was removed by rotary evaporation. The resultant solid was dissolved in
EtOAc (200 mL) and extracted by saturated aqueous NaHCO3 (2 x 100 mL). The
organic phase was dried with Na2SO4 and concentrated. The crude product was
submitted to flash chromatography (SiO2, 0 to 40% EtOAc-CHCl3), yielding 365 mg
(88%) of a tan solid: *H NMR (400 MHz, d6-DMSO) 8 8.38 (s, 1H), 7.92 (d, / = 6.4,
4H), 7.69 (m, 2H), 7.53 (m, 4H), 6.93 (br s, 2H), 6.90 (s, 1H), 6.43 (d, /= 7.2,1H),
6.10 (t, /= 9.2, 1H), 4.80 (br s, 1H), 4.59 (br, s, 1H), 2.09 (s, 3H), 1.97 (s, 3H);
[M+H]+ m/z 565.
Step 2) Preparation of'5-Amino-3-(3'-methyl-P-D-ribofnransyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (30)
66

WO 2006/066080 PCT/US2005/045589

b. K2CO3, MeOH, 66%.
5-Anuno-3-(2,'5'-di-0-benzoyl-3'-0-acetyl-3'-C-methyl-p-D-riboftiransyI)-
3H-thiazolo[4,5-d]pyrimidin-2-one (365 mg, 0.647 mmol) was dissolved in MeOH
(10 rnL). K2CO3 (17.9 mg, 0.129 mmol) was added, and the reaction was stirred for
16 h at rt Acetic acid (15.5 mg, 0.258 mmol) was added, and the reaction was
concentrated via rotary evaporation. The crude product was then submitted to HPLC
purification (MeCN-H2O), yielding 135 mg (66%) of a solid material: *H NMR (400
MHz, 4-DMSO) 5 8.35 (s, 1H), 6.82 (br s, 2H), 5.91 (d, /= 8.0,1H), 5.38 (d, /=
6.4,1H),4.81 (t, 7=6.0, lH),4.68(s, 1H),4.51 (t,/=5.6,1H),3.78(t,7=5.6,
1H), 3.48-3.53 (m, 2H), 1.21 (s, 3H); [M+H]+ m/z 315. Analysis calc'd for
CiiHi4N4OsS«0.5 H2O: C, 40.86; H, 4.68; N, 17.33; S, 9.92. Found: C, 40.78; H,
4.90; N, 16.94; S, 9.87.
Example 9
Preparation of 5-amino-2,3'dihydro-2-thioxo-3-j8-D-ribofaranosyl-thiazolo[4,5-
d]pyrimidin~7(6H)-one (33)

Step 1) Preparation of5-amino-2,3-dihydro-2-thioxo-3-(2',3',5'-tri-0-acetyl-p-D-
ribofuranosyl)thiazolo[4,5-d]pyrimidin-7~(6H)-one(32)
67

WO 2006/066080 PCT/US2005/045589

a. BSA, TAR, MeCN, 60° C; TMSOTf, 60° C, 80%.
Heterocycle 31 [prepared according to the method of Robins et. ah J. Med.
Chem. 1990,33,407-415] (150 mg, 0.75 mmol), TAR (214 mg, 0.675 mmol),
MeCN (10 mL) and BSA (0.55 mL, 2.25 mmol) were combined and heated for 1 h
at 60°C. The reaction was then charged with TMSOTf (250 mg, 1.13 mmol) and
stirred for 16 h at 60 C. The mixture was concentrated via rotary evaporation, and
the crude solid was dissolved in EtOAc (20 mL). This organic phase was then
extracted with saturated aqueous NaHCO3 (2 x 10 mL), and concentrated to dryness
by rotary evaporation. Trituration of the residue with Et2O (10 mL) yielded 200 mg
(80%) of a solid material that was further purified via HPLC (MeCN-H2O) to
generate an analytically pure sample: *H NMR (400 MHz, 4-DMSO) 5 11.54 (s, 1
H), 7.04 (br s, 2H), 6.59 (m, 1H), 6.10 (m, 1H), 5.70 (t, /=7.2,1H), 4.42 (dd, / =
12.0, 3.2,1H), 4.27 (m, 1H), 4.18 (m, 1H), 2.08 (s, 3H), 2.05 (s, 3H), 1.99 (s, 3H);
[M+H]+ m/z 459. Analysis calc'd for CHOgSz: C, 41.92; H, 3.96; N, 12.22; S,
13.99. Found: C, 41.78; H, 3.99; N, 12.02; S, 13.72.
Step 2) Preparation of 5-amino-2,3-ditydro-2-thioxo-3-/3-D-ribofuranosyl-
b. K2CO3, MeOH, it, 66%.
68
thiazolo[4,5-d]pyrinudiri'7(6H)-one (33)


WO 2006/066080 PCT/US2005/045589
Nucleoside triester 32 (100 mg, 0.21 mmol) and K2CO3 (42.7 mg, 0.31
mmol) were dissolved in MeOH (5 mL) and stirred for 16 h at ambient temperature.
To (his mixture was added HOAc (37 mg, 0.62 mmol) and the solvent was removed
via rotary evaporation. The residue were then submitted to HPLC purification
(MeCN-H2O) yielding 47 mg (66%) of a solid: *H NMR (400 MHz, 4-DMSO) 8
11.66 (s, 1H), 6.91 (br s, 2H), 6.47 (s, 1H), 5.31 (d, 7= 5.2,1H), 4.94 (s, 1H), 4.78
(s, 2H), 4.27 (d, J= 8.0,1H), 3.78 (m, 1H), 3.69 (m, 1H), 3.52 (m, 1H); [M+H]+ m/z
333. Analysis calc'd for C10H12N405S2 • 0.5 TFA • 0.75 H2O • 0.25 MeCN: C, 33.43;
H, 3.60; N, 14.41. Found: C, 33.11; H, 3.73; N, 14.80.
Example 10

Preparation of 5-amino-2,3-dihydro-2-thioxo~3-(2',3\5')~tri-O-acetyl-fi-D-
xylofuranosyl)thiazolo[4,5-d]pyrimidin-7-(6H)-one (34)

5-amino-2,3-dihydro-2-thioxo-3-(2 ,',5'-tri-O-acetyl-b-D-
xylofuranosyl)thiazolo[4,5-d]pyrimidin-7'(6H)-one (34)
a. BSA, tetraacetylxylofuranose, MeCN, 60° C, 30 min; TMSOtf, 4 h, 15%.
Heterocycle 31 (265.3mg, 1.33mmol), tetraacetylxylofuranose (380 mg, 1.19
mmol), BSA (1.26 mL, 5.32 mmol) and MeCN (10 ml) is heated to 60°C for 30
minutes. Then the reaction was charged with TMSOTf (0.36 mL, 2.0 mmol). After 4
h the reaction was worked up by removing the solvent by rotary vacuum and talcing
up the crude solid in ethyl acetate (15 mL). This organic phase was then extracted
with saturated sodium bicarbonate (2 x lOmL). The organic phase was concentrated
69

WO 2006/066080 PCT/US2005/045589
and the crude solid was triturated in 1:1 EtOAc-hexanes. The solid was collected
and submitted to HPLC purification (MeCN-H2O) yielding 40 mg (15%): 1H NMR
(400 MHz, 4-DMSO) 8 11.45 (s, 1H), 6.90 (br s, 2H), 6.49-6.58 (m, 1H), 6.35-6.49
(m, 1H), 5.58 (d, J= 5.6,1H), 4.55 (s, 1H), 4.31 (m, 2H), 2.11 (m, 3H), 1.99 (m,
3H), 1.98 (m, 3H); [M+H]+ m/z 459. Analysis calc'd for C16H18N4Sa: C, 41.92; H,
3.96; N, 12.22; S, 13.99. Found: C, 42.14; H, 3.99; N, 12.11; S, 14.01.
Example 11
5-Amino-3-b-D-ribofuranosyl-3H'thiazolo-[4,5-d]pyrimidin-2-thione (39)

Step 1) Preparation of 5-Amino-3H-thiazolo[4,5-d]pyrimidfte-2-thione (37)

b. O-Ethylxanthic acid potassium salt, DMF, 30%.
5-Bromo-pyrimidine-2,4-diamine (2.0 g, 10.58 mmol) [prepared in a manner
similar to English et. al. J. Am. Chem. Soc. 1946,68,453-458] and O-ethylxanthic
acid potassium salt (3.39 g, 21.16 mmol) was heated in DMF (25 mL) to 140° C.
After 5 h the reaction was cooled to ambient temperature and 25 mL of water was
added. The pH was then adjusted to 5.0 using 1N HC1. A red precipitate formed
and was collected by filtration, yielding 900 mg (30%) of a solid: 1H NMR (400
MHz, de-DMSO) 8 13.85 (s, 1H), 8.33 (br s, 1H), 6.90 (s 2H).
Step 2) Preparation of 5-Amino-3-(2'3')5'-tri-0-acetyl-P-D-ribofuranosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-thione (38)
70

WO 2006/066080 PCT/US2005/045589



o. TAR, BSA, CH3CN, TMSOTf, 57%.
5-Amino-3H-thiazolo[4,5-d]pyrimidine-2-thione (250 mg, 1.36 mmol), TAR
(389 mg, 1.22 mmol) and BSA (1.0 mL, 4.08 ramol) was heated to 60° C in
acetontrile (10 mL). After 30 minutes the reaction was charged with TMSOTf (0.37
mL, 2.04 mmol) and the reaction was allowed to progress for 16 h. The solvent was
then removed via rotary evaporation, and the crude product re-dissolved in EtOAc
(15 mL). The organic phase was extracted with concentrated aqueous NaHCO3 (2 x
l0mL). The organic phase was then concentrated again and submitted to flash
chromatography (SiO2,5% MeOH-EtOAc) yielding 301 mg (57%) of white solid:
1H NMR (400 MHz, 4*-DMSO) 5 8.49 (s, 1H), 7.08 (br s, 2H), 6.65 (s 1H), 6.12 (m,
1H), 5.79 (t, J = 8.0,1H), 4.43 (dd, /= 12.0,3.6,1H), 4.28-4.34 (m, 1H), 4.17 (dd, /
= 11.6, 6.8,1H), 2.09 (s, 3H), 2.05 (s, 3H), 1.97 (s, 3H); [M+H]+ m/z 443.

Step 3) Preparation of 5-Amino-3-b-D-ribofaranosyl-3H-thicizolo-[4,5-d]pynmidin-
2-thione (39)
d K2CO3, MeOH, 74%.
5-Amino-3-(2' ,3' ,5'-tri-O-acetyl-b-D-ribofuranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-thione (202 mg, 0.46 mmol) was dissolved in MeOH (5 mL), and
K2CO3 (18.9 mg, 0.14 mmol) was added. After 1 h acetic acid (21 mg, 0.28 mmol)
was added, and the reaction was concentrated via rotary evaporation. The crude
solid was then submitted to HPLC purification (MeCN-H2O), yielding 108 mg
(74%) of white solid: !H NMR (400 MHz, d6-DMSO) 5 8.45 (s, 1H), 6.96 (br s, 2H),
6.53 (d, J=4.4,1H), 5.33 (d, /= 6.0,1H), 5.03 (m, 1H), 4.86 (d, = 6.4,1H), 4.67
(t, /= 6.0,1H), 4.33 (m, 1H), 3.79 (m, 1H), 3.70 (m, 1H), 3.53 (m, 1H). Analysis
71

WO 2006/066080 PCT/US2005/045589
calc'd for C10H12N4O4S2U35 H2O: C, 37.22; H, 3.97; N, 17.36; S, 19.87. Found: C,
37.64; H, 3.87; N, 17.02; S, 19.39.
Example 12
5-Amino-3~3-D-xylofuranosyl-3H-thiazolo-[4,5-d]pyrimidin-2-thione (41)

Stepl) Preparation of 5-Amino-3-(2',3',5'-tri-0-acetyl-j3-D-xylofuranosyl)-3H~
thiazolo[4,5-d]pyrimidin-2-thione(40)

0. Tetraacetylxylofuranose, BSA, MeCN.TMSOTf, 13%.
5-Amino-3H-thiazolo[4,5-d]pyrimidine-2-thione (237 mg, 1.28 mmol), tetra-
acetylxylose (370 mg, 1.16 mmol) and BSA (1,25 mL, 5.12 mmol) were heated in
MeCN (10 mL) to 60° C for 30 min. To this was added TMSOTf (347 pL, 1.92
mmol), and the reaction mixture was stirred at 60° C for 16 h, whereupon the solvent
was removed by rotary evaporation, and the crude solid was re-dissolved in EtOAc
(15 mL). This organic phase was then extracted with concentrated NaHCO3 (2 x 10
mL), and then concentrated to a solid residue that was submitted to flash
chromatography (0-100% EtOAc-CHCl3) yielding 67 mg (13%) of tan solid: !H
NMR (400 MHz, ds-DMSO) 8 8.51 (s, 1H), 6.90 (br s, 2H), 6.67 (d, / = 4.0,1H),
6.49 (t, =2.0, 1H), 5.62 (m, 1H), 5.63 (m, 1H), 4.37 (m, 1H), 4.21 (br m, lH), 2.18
(s, 3H), 1.97 (s, 3H), 1.94 (s, 3H); [M+H]+ m/z 443.
Step 2) Preparation of 5-Amitto-3-3-D-xylqfuranosyl-3H-thiazolo-[4,5-d]pyrimidin-
2-thione (41)
72

WO 2006/066080 PCT/US2005/045589



f. K2CO3, MeOH, 62%.
5-Amino-23-dihydro-2-thioxo-3-(2,3,5-tri-0-acetyl-P-D-xylofuranosyl)thia-
zolo[4,5-d]pyrimidra-7-thione (65 mg, 0.14 mmol) is dissolved in MeOH (5 mL).
To this was added K2CO3 (19 mg, 0.137 mmol), and the resultant mixture was
stirred for 3 In whereupon it was quenched with HO Ac (140 JLLL, 2.4 mmol), and the
solvent was removed by rotary evaporation. The crude product is submitted to
HPLC purification (MeCN-H2O) yielding 30 mg (62%) of white solid: !H NMR
(400 MHz, d6-DMSO) 8 8.50 (s, 1H), 6.90 (br s, 2H), 6.45 (d, J = 5.2,1H), 5.67 (d, J
= 8.0,1H), 5.49 (d, J = 8.4,1H), 5.03 (m, 1H), 4.49 (t, J = 5.2,1H), 4.02 (m, 2H),
3.72 (m, 2H). Analysis calc'dfor HaNSa'OH2O: C, 37.12; H, 3.99; N,
1.7.32; S, 19.82. Found: C, 37.53; H, 3.80; N, 17.04; S, 19.42.
Example 13
5-Amino-7-ethoxy-3-(2 5'-di-O-acetyl-3 '~deoxy-b-D-ribofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidine-2-one (43)

Stfp 1) Preparation of5-Amino-7-hydroxy-3-(2',5'-di-O-acetyl-3'-deoxy-j3-D-
ribofara]iosyl)-3H,6H-thiazolo[4,5-d]pyrimidine-2-one{42)
73

WO 2006/066080 PCT/US2005/045589

a. 1,2,5-tri-O-acetyl-b-D-ribofuranosa, BSA, MeCN, 40°C; TMSOTf, 80"C.
To a mixture of heterocycle 6 (4.60 g, 25.00 mmol) in anhydrous MeCN
(83.0 mL) was added dropwise BSA (15.28 raL, 62.49 mmol). The reaction was
then immersed in a 40° C oil bath and stirred for 90 min, and l,2,5-tri-O-acetyl-{3-D-
ribofuranose (5.42 g, 20.80 mmol) was added followed by TMSOTf (5.65 mL, 31.24
mmol). The resulting thick mixture was immersed in an 80° C oil bath whereupon
the mixture clarified to a homogenous solution after 15 min. The reaction was stirred
for 2 h at 80° C, cooled to rt, and then partitioned between 1M pH 7 phosphate
buffer (50 mL) and EtOAc (100 mL). The resulting emulsion was filtered through a
pad of celite yielding two distinct layers that were separated. The organic layer was
dried over Na2SO4, filtered, and concentrated in vacuo to a residue. This residue was
submitted to flash chromatography (S1O2,0-6% MeOH-DCM) affording 3.41 g
(43%) of nucleoside 42 as a finely divided pale yellow solid: 1H (400MHZ, DMSO-
d6) 11.22 (s, 1H), 6.95 (br s, 2H), 5.79 (d, J = 2.0,1H), 5.59 (d, 7 « 7.2,1H), 4.20-
4.34 (m, 1H), 4.22 (dd, / = 3.2,12.0,1H), 3.99 (dd, J = 6.4,11.6,1H), 2.57-2.67(m,
1H), 2.05 (s, 3H), 1.99 (s, 3H), 1.97-2.03 (m, 1H); [M+H]+ m/z 385. Analysis calc'd
for C14H16N4O7S: C, 43.75; H, 4.20; N, 14.58; S, 8.34. Found: C, 43.64; H, 4.31; N,
14.37; S, 8.19.
74
Step 1) Preparation of5-Amino-7-ethoxy-3-(2',5'-di-O~acetyl-3'-deoxy-j3-D-
ribofuranosyl)-3H,6H'thiazolo[4,5-d]pyrimidine-2-one (43)


WO 2006/066080 PCT/US2005/045589
To a solution of 42 (99 mg, 0.26 mrnol) dissolved in anhydrous THF (5.5
mL) was added S-TPP PPh3 resin (0.36 g, 0.77 mmol, 2.15 mmol/g). The mixture
was chilled to 0° C, and EtOH (30.1 pL, 0.52 mmol) was added followed by DEAD
(176.0 uL, 0.39 mmol). The reaction mixture was removed from the ice bath and
warmed to rt whereupon it stirred for 16 h. The mixture was concentrated in vacuo
to a residue that was subjected to several passes of flash chromatography
purification (SiO2, elution with 2% MeOH / 0-40% EtOAc in hexanes) affording
0.22 mg of 43 (20%): ]H (400MHz, DMSO-d6) $ 6.95 (br s, 2H), 5.87 (d, J = 2.4,
IK), 5.64 (d, J = 7.2,1H), 4.39 (q, J = 6.8,2H), 4.32-4.4 (m,lH), 4.25 (dd, ./ = 3.2,
11.6,1H), 3.96-4.03 (m, 1H), 2.63-2.71 (m, 1H), 2.05 (s, 3H), 2.03-2.08 (m, 1H),
1.99 (s, 3H), 1.30 (t, / = 6.8,3H); [M+Hf mfz 413.
Example 14
5-Amino~7~ethoxy-3-(fcD-ribofuranosyl)-2,3-dihydro-2-thioxo-thiazoh[4,5-

Step 1) Preparation of5-Amino-7-ethoxy-3-(2',3'f5'-tri-0-acetyl-j3-D~
ribofuranosyl)~2,3-dihydro-2-thioxo-thiazolo[4,5-d]pyrimidin-7(6H)-one{4,A)

d]pyrimidin-7(6H)-one (45)
c. S-TPP, Ethanot, DEAD, THF, 65%.
5-Amino-2,3-d&ydro-2-thioxo-3-(2,3,5-tri-0-acetyl-(3-D-ribofurariosyl)thia--
zolo[4,5-d]pyrimidin-7(6H)-one (250 mg, 0.54 mmol) and S-TPP Ph3P resin (753
mg, 1.62 mmol) were suspended in THF (15 mL) and cooled to 0° C. Ethyl alcohol
75

WO 2006/066080 PCT/US2005/045589
(50 uL, 1.08nunol) and DEAD (148 \\L, 0.82 mmol) were added sequentially. After
1 h the reaction mixture was warmed to ambient temperature and stirred for 16 h.
The reaction mixture was then filtered, concentrated and submitted to flash
chromatography (SiO2,15% EtOAc-CHCl3), affording 200 mg (65%) of a white
foam: 3H NMR (400 MHz, d6-DMSO) 8 6.91 (s, 1H), 6.47 (br s, 2H), 6.29 (m, 1H),
6.18 (s, 1H), 4.62-4.31 (m, 5H), 1.42 (t, 7 = 4.2. 3H), 1.38 (s, 3H), 1.35 (s, 3H), 1.32
(s,3H);[M+H]+ m/z 487.
Step 2) Preparation of 5-Amino-7-ethoxy-3-j3-D-ribofuranosyl-2,3-dihydro-2-thioxo-
thiazolo[4,5-d]pyrimidin-7(6H)-one (45)

d.K2CO3lMeOH,a3%.
5-Amino-2,3-dihydro-2-thioxo-3"(2,3,5-tri-O-acetyl-P-D-
ribofuranosyl)tlnazolo-[4,5-d]pyrimidin-7(6H)-ethyl ether (180 mg, 0.37 mmol) and
K2CO3 (12.8 mg, 0.01 mmol) were suspended in MeOH (5 mL). After 1 h acetic
acid was added and the solvent removed rotary evaporation. The crude product was
then submitted to HPLC purification (MeCN-H20) yielding 105 mg (83%) of a
solid: *HNMR (400 MHz, d6-DMSO) 5 6.98 (s, 2H), 6.50 (d, 7 = 4.8,1H), 5.32 (d,
7= 5.2,1H), 5.01 (s, 1H), 4.85 (d, 7= 5.6,1H), 4.68 (t, 7 = 5.6,1H), 4.43 (dd, 7=
13.6,6.8, 2H), 4.30 (m, 1H), 3.80 (m, 1H), 3.71 (m, 1H), 3.66 (m, 1H), 1.31 (t, 7=
6.8,3H); [M+HJ+ m/z 361.
Preparation of 5-Amino-3H-oxazolo[4,5-d]pyrimidin-2-one (50)

d NaH, ImgC^O, OMF.
2,4-Diamino-pyrimidin-5-ol (500 mg, 3.97 mmol) [prepared according to the
method of Hull. 7. Chem. Soc. 1956,2033-2035] was suspended in DMF (10 mL).
76

WO 2006/066080 PCT/US2005/045589
To this was added consecutively NaH (86.7 mg, 3.77 mmol) and CDI (707 mg, 4.36
mmol), and the reaction mixture was heated with vigorous stirring at 60° C for 3 h.
The mixture was cooled to ambient temperature, and then quenched with water (25
mL). The solvent and water were removed via rotary evaporation, and the residue
was then triturated in water (5 mL). The solid was then collected by filtration and
dried, affording 230 mg (38%): 1H NMR (400 MHz, -DMSO) 511.90 (br s, 1H),
7.72 (br s, 1H), 6.72 (s, 2H); Analysis calc'd for C5H4N4CVO.2 H2O: C, 38.56; H,
2.85; N, 35.98. Found: C, 39.01; H, 2.71; N, 35.58; [M+Hf m/z 153.

a. NaH, lm2C=S, DMF. b. BSA, appropriate p-D-ribofuranose, MeCN, rt; TMSOTf,
80°C. c. K2CO3, MeOH, it d. NaH, lm2C=0, DMF.
The diaminohydroxypyrimidine 46 was reacted with NaH and CDI in DMF
to afford heterocycle 50, or with NaH and TCDI in DMF to provide heterocycle 47.
Both arrunopyrimidines 47 and 50 can be independently submitted to BSA-TMSOTf
mediated coupling reactions with an appropriately selected j5-D-ribofuranose
(wherein R1, R2 and R3 may be independently acetyl or benzoyl) to give nucleosides
77

WO 2006/066080 PCT/US2005/045589
48 and 51 respectively. Alkaline raethanolysis of 48 and 51 should afford
deprotected nucleosides 49 and 52 respectively.

Preparation of 5-Amino-3-pyridin-3-ylmethyl-3H-thiazolo[4,5-d]pyrimidin~2~
one (54)
Step 1: Preparation of 5-Amino-3-pyridin-3-ylmetIryl-3H-thiazolo[4,5-
d]pyrimidin-2-one (54)
5-Arnino-3H-thiazolo[4,5-d]pvrirnidin-2-one (107 mg, 0.64 mmol) was
dissolved in DMF (4 mL) at ambient temperature. Sodium hydride (30 rag, 1.32
mmol) was added and the mixture was heated to 30 °C. Stirring was continued
for 0.5 h before 3-bromomethyl-pyridine hydrobromide (179 mg, 0.71 mmol)
was added. The mixture was then heated to 75 °C and allowed to stir for 4 h.
Upon completion, the reaction was allowed to cool to room temperature, then
concentrated. Water (12 mL) was added. The resulting mixture was diluted with
H2O (12 mL), then extracted with ethyl acetate (3x5 mL). The combined
organic layers were washed with brine, dried over Na2SO4, filtered, and
concentrated. The crude material was purified by column chromatography
(SiO2,20-50% EtOAc-CH2Cl2) to generate 90 mg (54%) of 54 as a white solid:
5H NMR (400 MHz, DMSO-d*) 8 8.59 (s, 1H), 8.48 (d, /= 3.6,1H), 8.32 (s,
1H), 7.71 (d, /= 8.4,1H), 7.36 (m, 1H), 6.86 (s, 2H), 5.04 (s, 2H); [M+H]+
260.1.
78

WO 2006/066080 PCT/US2005/045589
Example 16
Preparation of 5-Amino~3-(6-chloro-pyridin-3-ylmethyl)-3H-thiazolo[4,5-
dJpyrimidin-2-one (55)
Step 1: Preparation of5-Amino-3-(6-chloro-pyridin-3-yhnethyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (55)
In a manner similar to Example 15, Step 1,111 mg of the title compound 55
was generated in 54% yield as an orange solid: *H NMR (400 MHz, DMSO-d6)
5 8.42 (s, 1H), 8.30 (s, 1H), 7.77 (d, J = 8.8,1H), 7.47 (d, 7= 8.0,1H), 6.85 (s, 2H),
5.11 (s, 2H); [M+Hf 294.1.
Example 17
Preparation of (Z)-5-Amino-3-(4~chlom-2-buten-l-yl)-3H-thiazolo[4,5-d]pyrimidin-
2-one (56)
Step 1: Preparation of(Z)~5-Amino-3-(4-chloro-2-buten-l-yl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (56)
In a manner similar to Example 15, Step 1, 74 mg of the title compound 56
was generated in 46% yield as a yellow solid: *H NMR (400 MHz, DMSO-^j)
8 8.29 (s, 1H), 6.80 (s, 2H), 5.81 (m, 1H), 5.66 (m, 1H), 4.53 (d,/ = 6.0,2H), 4.27
(d, / = 8.0,2H); [M+H]+ 257.2.
Example 18
Preparation of5-Amino-3-hexyl-3R~thiazolo[4,5-d]pyrimidin-2-one (57)


WO 2006/066080 PCT/US2005/045589
Step 1: Preparation of 5-Amino-3-hexyl-3H-thiazolo[4,5-d]pyrimidin-2-one (57)
In a manner similar to Example 15, Step 1, 51 mg of the title compound 57
was generated in 15% yield as an off-white solid: 'H NMR (400MHZ, DMSO-d6) 8
8.28 (s, 1H), 6.78 (s, 1H), 3.82 (t, /= 7.2, 2H), 1.64 (m, 2H), 1.27 (m, 6H), 0.85 (t, J
= 6.8,3H);[M+H+] 253.1.
Example 19
Preparation of (±)-5-Amino-3-cyclopentyl-3H-thiazolo[4,5-dJpyrimidin-2-one (58)

Step 1: Preparation of(±)-5-Amino-3~cyclopentyl-3H-thiazolo[4,5-d]pyrimidin-2-
one (58)
In a manner similar to Example 15, Step 1,21 mg the title compound 58 was
generated in 5% yield as a light yellow solid: 1H NMR (400 MHz, CDCl3) 8 8.09 (s,
1H), 5.23 (s, 2H), 2.24 (m, 1H), 2.00 (m, 4H), 1.66 (m, 4H); [M+H]+ 237.0.
Example 20
Preparation of 5-Anuno~3-(4-nitro-phenyI)-3H-thiazolo[4.5-d]pyrimidin-2-one (59)

Step 1: Preparation of 5-Amino~3-(4'nitro-phenyl)-3H-thiazolo[4,5-d]pyrimidin-2-
one (59)
In a manner similar to Example 15, Step 1,45 mg of the title compound 59
was generated in 10% yield as an orange solid: '1H NMR (400 MHz, DMSO-4)
8 8.59 (s, 1H), 8.18 (d, J = 9.2,2H), 7.99 (d, J= 9.2,2H), 5.74 (s, 2H); [M+H]+
290.2.
Example 21
5-Amino-3-(2,3,5,6-tetrafluoro-pyridin-4-yl)-3H-tHazolo[4,5-d]pyrimidin-2'one
(60)
80

WO 2006/066080 PCT/US2005/045589

Step 1: Preparation of5-Amino-3-(2,3,5,6~tetrafluoro-pyridin-4-yl)-3H-
thiazoh[4,5-d]pyrimidin-2-one (60)
In a manner to Example 15, Step 1,35 mg of the title compound 60 was
generated in 5% yield as an orange solid: *H NMR (400 MHz, DMSO-d6) § 8.16(s,
1H), 4.01(s, 2H); [M+Hf 318.4.

a) (E)-1,4-dichloro-2-butene, NaH, DMF
b)O.1MHCI
Example 22
Preparation of (E)-5-Amino-3-(4-chloro-2-buten-l-yl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (62)
Step 1: Preparation of(E)-5-Amino-3-(4-chloro-2-buten-l-yt)-3H-thiazolo[4,5-
dJpyrimidin-2-one (61).
The title compound 61 can be synthesized by treating 5-Amino-3#~
thiazolo[4,5-rf|pyrimidin-2-one (1) in DMF with sodium hydride and (E)-l,4-
dichloro-2-butene under various conditions.
Step 2: Preparation of(E)~5-Amino-3-(4-hydroxy-2-buten-l-yl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (62)
The title compound 62 could be synthesized by treating (E)-5-Amino-3-(4-
chloro-2-buten-l-yl)-3/7-thiazolo[4,5-rf]pyrimidin-2-one (61) with 0.1 M HC1 under
various conditions.
Scheme 3
81

WO 2006/066080 PCT/US2005/045589

a) BSA, TMSOTf, CH3CN, 80ºC, 3-4 h
b) K2CO3, DMF, rt, overnight
Example 23
Preparation of (3 'S)-5-Amino-3-(3 '-deoxy-3 '-hydroxymethyl-b-D-ribofuranosyl)-
3H-thiazolo[4,5-d]pyrimidin-2-one (65)
Step 1: Preparation of(3'S)-5-Amino-3-(3'-acetoxymethyl-2',5'-di-0-acetyl-3'-
deoxy~J3-D-ribofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (64)

(3S)-3-0-Acetoxymethyl-l, 2,5-tri-0-acetyl-3-deoxy-c,p-D-ribofaranose
(63) [prepared according to the method of Cooperwood et al, Nucleosides,
Nucleotides, and Nucleic Acids 2000,19,219-236 in which the enantiomer of the
same compound was made] (176 mg, 0.53 mmol) was dissolved in acetonitrile (7
mL) at ambient temperature. 5-Ammo-3H-thiazolo[4,5-rf]pyriinid.in-2-one (1) (89
mg, 0.53 mmol) was added, the mixture was then stirred for 0.5 h before it was
heated to 40 °C. After 5 min at 40 °C, BSA (0.39 mL, 1.59 mmol) was added and
the mixture was stirred for another 0.5 h. The mixture was then heated to 80 °C.
TMSOTf (0.14 mL, 0.80 mmol) was added and the reaction was stirred for 3-4 hours
at 80 °C. Upon completion, the reaction was allowed to cool to room temperature
and then quenched by a pH 7.0 buffer (1.0 M K2HPO4 and 1.0 M NaH2PO4,2 ml).
The mixture was extracted with CH2CI2 (3 x 10 mL). The combined organic layers
were washed with brine, dried with Na2SO4, and concentrated in vacuo. The crude
product was purified by column chromatography (S1O2,0-10% MeOH-CH2Cl2 to
82

WO 2006/066080 PCT/US2005/045589
afford 77 mg (33%) of 64 as a powdery light yellow solid: !H NMR (400 MHz,
CDCk) 5 8.14 (s, 1H), 6.04 (d, /= 1.6,1H), 5.90 (dd, 7= 6.8,1.6,1H), 5.24 (s, 2H),
4.52 (dd, /= 12.0,2.8,1H), 4.36 (m, 2H), 4.17 (m, 2H), 3.54 (m, 1H), 2.18 (s, 9H);
[M+H]+ 441.2; Elemental analysis for CHzoNgS-O.eHaO: calc'd: C, 45.25; H,
4.74; N, 12.42; S, 7.11; found: C, 45.24; H, 4.66; N, 12.02; S, 7.24.
Step 2: Preparationof(3'S)-5-Ammo-3-(3'-deoxy-3'-hydroxymethyl-fi-D-
ribofura.nosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (65)
(3'S)-5-Amino-3-(3'-acetoxymethyl-2',5'-di-0-acetyl-3'-deoxy-p-D-
ribofuranosyl)-3H-thiazoio[4,5-3Tipyrimiuin-2-one 64 (114 ing, 0.28 mtnol) was
dissolved in methanol (2 mL) at ambient temperature. Potassium carbonate (2 mg,
cat.) was added and the mixture was stirred at room temperature overnight. Upon
completion, acetic acid was added (2UL) and the mixture was stirred another 30
minutes at room temperature. The mixture was concentrated, purified by HPLC,
then triturated by EtOAc to afford 79 mg (90%) of 65 as a white solid: H NMR
(400 MHz, D2O) 8 8.28 (s, 1H), 6.10 (m, 1H), 5.18 (m, 1H), 4.20 (m, 1H), 3.95 (m,
2H), 3.78 (m, 2H), 3.00 (m, 1H); [M+H]+ 315.2; Elemental analysis for
CnHi4N4O5S-0.3H2O-0.15iPrOH: calc'd: C, 41.83; H, 4.84; N, 17.04; S, 9.75;
found: C, 41.92; H, 4.61; N, 16.89; S, 9.78.
Example 24
Preparation of 5-Ammo-3-(5'-deoxy-5'-hydroxymethyl-b-D-ribofuranosyl)~3H-
thiazolo[4,5-d]pynmidin-2-one (68)
Step 1: Preparation of5-Amino-3~(5'~0-acetoxymethyl-2',3'-di-0-acetyl-5'-deoxy-
P-D-ribofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one(67)

83


WO 2006/066080 PCT/US2005/045589
In a manner similar to Example 23, Step 1,113 mg of the title compound 67
was generated from 5-O-acetoxymethyl-l, 2,3-tri-O-acetyl-5-deoxy-a,p-D-
ribofuranose (66) [prepared according to the method of Pakulski et al. Polish J.
Chem. 1995, 69,912-917] in 53% yield as a sticky yellow solid: 'H NMR (400
MHz, CDCl3) S 8.15 (s, 1H), 6.34 (m, 1H), 6.25 (d, J = 6.0,1H), 6.11 (d, J - 4.0,
1H), 6.04 (m, 1H), 5.76 (t, 7= 6.0,1H), 5.42 (s, 1H), 4.93 (m, 1H), 4.35 (m, 1H),
4.21 (q, /= 5.6,1H), 2.20 (s, 9H); [M+H]+441.2.
Step2: Preparation of 5-Amino-3-(5'-deoxy-5'-hydroxymethyl-^-D-ribofuranosyl)-
3H-thiazolo[4,5-d]pyrimidin-2-one(6S)
In a manner similar to Example 23, Step 2,43 mg of the title compound
68 was generated in 71% yield as a white solid: 1H NMR (400 MHz, &-DMSO)
5 8.33 (s, 1H), 6.84 (s, 2H), 5.86 (d, / = 4.4,1H), 5.26 (d, / = 5.2,1H), 4.93 (m,
1H), 4.74 (q, /= 10.0,4.4,2H), 4.40 (m, 1H), 3.82 (m, 2H), 1.76 (m, 3H);
[M+H]+ 315.2; Elemental Analysis for CH4N4OS-0.4H2O-0.2PrOH: calc'd:
C, 41.77; H, 4.96; N, 16.80; S, 9.61; found: C, 41.61; H, 4.85; N, 16.68; S, 9.58.
Example 25
Preparation of 5-Amino-3-(3 '-deoxy-3 '-O-p-toluenesulfonyl-b-D-xylofuranosyl)-
3H-thiazolo-[4,5-d]pyrimidine-2-one (73)

a) TsCI, py, it, 24 h
b) Ac2O, AcOH, H2SO4, rt, 24 h
Step 1: Preparation of 1,2-O-Isopropyliene-3-O-p-toluenesulfonyl-5-O-trityl-/}-D-
xylojuranose (70)
l,2-0-Isopropylidene-5-0-trityl-P-D-xylofuranose (69) [prepared according
to the method of Johnston etal Tetrahedron Lett. 1995, 36, 4341-4344] (4.25 g,
9.83 rnmol) was dissolved in pyridine (60 mL) at ambient temperature. P-
84


WO 2006/066080 PCT/US2005/045589
In a manner similar to Example 23, Step 1,113 mg of the title compound 67
was generated from 5-O-acetoxymethyl-l, 2,3-tri-O-acetyl-5-deoxy-a,p-D-
ribofuranose (66) [prepared according to the method of Pakulski et al. Polish J.
Chem. 1995, 69,912-917] in 53% yield as a sticky yellow solid: 'H NMR (400
MHz, CDCl3) 8 8.15 (s, 1H), 6.34 (m, 1H), 6.25 (d, J = 6.0,1H), 6.11 (d, J = 4.0,
1H), 6.04 (m, 1H), 5.76 (t, = 6.0,1H), 5.42 (s, 1H), 4.93 (m, 1H), 4.35 (m, 1H),
4.21 (q, /= 5.6,1H), 2.20 (s, 9H); [M+H]+441.2.
Step2: Preparation of 5-Amino-3-(5'-deoxy-5'-hydroxymethyl-fi-D-ribofuranosyl)-
3H-thiazolo[4,5-d]pyrimidin-2-one(68)
In a manner similar to Example 23, Step 2,43 mg of the title compound
68 was generated in 71% yield as a white solid: H NMR (400 MHz, d6-DMSO)
5 8.33 (s, 1H), 6.84 (s, 2H), 5.86 (d, / = 4.4,1H), 5.26 (d, / = 5.2,1H), 4.93 (m,
1H), 4.74 (q, = 10.0,4.4,2H), 4.40 (m, 1H), 3.82 (m, 2H), 1.76 (m, 3H);
[M+H]+ 315.2; Elemental Analysis for C11HN4O5S-0.4H2O-0.2PrOH: calc'd:
C, 41.77; H, 4.96; N, 16.80; S, 9.61; found: C, 41.61; H, 4.85; N, 16.68; S, 9.58.
Example 25
Preparation of 5-Amino-3-(3 '-deoxy-3 '-O-p-toluenesulfonyl-b-D-xylofuranosyl)-
3H-thiazolo-[4,5-d]pyrimidine-2~one (73)

a) TsCI, py, rt, 24 h
b) Ac2O, AoOH, H2SO4, rt, 24 h
Step 1: Preparation of l,2-O-Isopropyliene-3-O-p-toluenesulfonyl-5-O-trityl-j3-D-
xylojuranose (70)
l,2-0-Isopropylidene-5-0-trityl-P-D-xylofuranose (69) [prepared according
to the method of Johnston et al. Tetrahedron Lett. 1995,36, 4341-4344] (4.25 g,
9.83 mmol) was dissolved in pyridine (60 mL) at ambient temperature. P-
84

WO 2006/066080 PCT/US2005/045589
Toluenesulfonyl chloride (2.81 g, 14.74 mmol) was added to the solution. After 24 h
the reaction had gone to completion, the crude mixture was concentrated. The
residue was dissolved in EtOAc (50 mL), washed with saturated aqueous NH4CI (25
mL), saturated aqueous NaHC(>3 (25 mL) and brine (25 mL). The organic phase
was dried over MgSO4, filtered, then concentrated. The mixture was then purified by
ISCO chromatography (SiO2,2-15% EtOAc-Hexane), affording 5.20 g (90%) of 70
as a white solid: H (400 MHz, CDCl3) 5 7.61 (m, 2H), 7.32-7.34 (m, 6H), 7.23-7.32
(m, ?H), 5.92 (d, J=4.4, IE), 4.74 (dd, /= 11.2, 3.6, 2H), 4.19-4.22 (m, 1H), 3.45
(dd, J= 10.4,6.4,1H), 3.05 (q, J- 5.2,1H), 2.40 (s, 3H), 1.49 (s, 3H), 1.31 (s, 3H).
Step 2: Preparation of l,2,5-Tri-0-acetyl-3-0-p-toluenesulfonyl -a, B-D-
xylofuranose (71)
l,2-O-isopropyliene-3-O-p-toluenesuIfonyl-5-O-trityl-P-D-xylofuranose(70)
(5.20 g, 8.86 mmol) was dissolved in AcOH (60 mL) at ambient temperature. Acetic
anhydride (4.23 mL, 44.71 mmol) was added dropwise to the solution. The resulting
mixture was cooled to 0°C, followed by slow addition of 1M H2SO4 (9.75 mL, 9.75
mmol). After 24 h the reaction had gone to completion, the crude mixture was
concentrated, then azeotroped with toluene (2 x 20 mL). The residue was dissolved
in CH2CI2 (50 mL), washed with saturated aqueous NaHCQ} (20 mL). The Organic
phase was dried over MgSCU, filtered, then concentrated. The mixture was then
purified by ISCO chromatography (SiO2,2-40% EtOAc-Hexane), affording 3.09 g
(81%) of 71 as a colorless oil: 'H (400 MHz, CDCI3) 6 (a mixture of a and p
isomers) 7.80-7.85 (m), 7.37-7.39 (m), 6.36 (d, / = 4.4), 6.06 (s), 5.20-5.30 (m),
4.56-4.62 (m), 4.26-4.29 (m), 2.50 (s), 2.06-2.08 (m).
Step 3: Preparation of5~Amino-3-[2'5'-di-0-acetyl-3'-deoxy-3'~0-p-
toluenesulfmyl-P-D-xyhfu.ranosyl]-3H-thiazolo-[4t5-d]pyrimidine-2-one (72)

In a manner similar to Example 23, Step 1,161 mg of the title compound 72
was generated in 54% yield as a fluffy yellow solid: 1H NMR (400 MHz, CDCl3)
5 8.12 (s, 1H), 7.85 (d, J~ 8.8,2H), 7.39 (d, / = 8.8,2H), 6.18 (d, /= 2.8,1H), 5.90
85

WO 2006/066080 PCT/US2005/045589
(br s, 2H), 5.77 (d, 7 = 4.4,1H), 5.01 (dd, 7 = 6.4, 5.2,1H), 4.34 (m, 1H), 4.27 (m,
2H), 2.48 (s,3H), 2.04 (s,6H);[M+H]+ 539.3.
Step 4: Preparation of 5-Amino-3-[3'-deoxy-3'-O-p-toluenesulfonyl-0-D-
xylofuranosyl]-3H-thiazolo-[4,5-d]pyrimidine-2-one (73)
In a manner similar to Example 23, Step 2,68 mg of the title compound 73
was generated in 61% yield as a white solid; *H NMR (400 MHz, DMSO-ds) 8 8.36
(s, IE), 7.83 (d, 7= 8.0, 2H), 7.48 (d, 7= 8.0, 2H), 6.80 (s, 2H), 5.92 (d, 7=6.0,
IB), 5.71 (d, 7= 6.4,1H), 5.20 (m, 1H), 4.89 (q, 7 = 5.6,3.6,1H), 4.73 (s, 2H), 4.10
(m, 2H), 2.43 (s, 3H); [M+Hf 455.2; Elemental analysis for (C17H18N4O7S2
0.4H2O): calc'd: C, 44.22; H, 4.10; N, 12.14; S, 13.89; found: C, 44.45; H, 4.15; N,
12.07; S, 13.71.
Example 26
Preparation of '5-Amino-3-(3 '-deoxy-3 '-methylidene-b-D-ribojuranosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-one (76)

Stepl: Preparation of 5-Amino-3-(2'-O-acetyl-5'-O-benwyl-3'-deoxy-3'-
7nethylidene-^-D-ribofiiranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one(7S)

In a manner similar to Example 23, Step 1,107 mg of the title compound 75
was generated from l,2-di-O-acetyl-5-O-benzoyl-3-deoxy-3-methylidene-a,(3-D-
ribofiiranose (74) [prepared according to the method of Girardet et al. J. Med. Chem.
2000,43, 3704-3713] in 85 % yield as a yellow solid: 1H NMR (400 MHz, CDCl3)
8 8.13 (s, 1H), 8.05 (dd, = 8.4,1.2,2H), 7.57 (tt, =7.2,1.2,1H), 7.44 (t, J= 7.2,
2H), 6.51 (m, 1H), 6.17 (d, = 4.4,1H), 5.30 (s, 2H), 5.11 (m, 2H), 4.82 (dd,J =
11.6,4.8,2H), 4.52 (dd, 7= 11.6,6.8,1H), 2.14 (s, 3H); [M+H]+ 443.2.
86

WO 2006/066080 PCT/US2005/045589
Step 2: Preparation of 5-Amino-3-(3'-deoxy-3'-methylidene-fi-D-ribofiiranosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-one (76)
In a manner similar to Example 23, Step 2, 35 mg of the title compound 16
was generated in 35 % yield as a gray-white solid: 1H NMR (400 MHz, DMSO-d6)
5 8.37 (s, 1H), 5.83 (d, / = 5.6,1H), 5.74 (d, 7=7.6,1H), 5.51 (m, 2H), 5.19 (d, 7 =
11.2,2H), 4.72 (t, 7= 6.0,1H), 4.54 (br s, 2H), 3.85 (s, 2H); [M+H]+ 297,2;
Elemental analysis for (CnHi2N4O4S-0.2H2O-0.25iPrOH): calc'd: C, 44.81; H, 4.61;
N, 17.79; S, 10.18; found: C, 44.84; H, 4.33; N, 17.76; S, 10.22.
Example 27
Preparation of (3'R)-5-Amino-3-(3'-deoxy-3'-fludro-p-D-xylofumnosyl)-3H-
thiazolo[4,5-d]pyrimidin-2-one (79)

Step 1: Preparation of(3'R)-5-Amino-3-(2'-O-acetyl-5'-O-benzoyl-3'-deoxy-3'-
flMoro-,8-D-xylofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (78)

In a manner similar to Example 23, Step 1,148 mg of the title compound 78
was generated from 1,2-Di-0-acetyl-5-0-benzoyl-3-deoxy-3-(R)-fluoro-a,J5-D-
xylofuranose (77) [prepared according to the method of Gosselin et al.
Carbohydrate Research 1993,249,1-17] in 56% yield as a light yellow solid: !H
NMR (400 MHz, DMSO-&) 5 8.16 (s, 1H), 8.05 (d, = 8.8,2H), 7.57 (t, J = 7.6,
1H), 7.44 (t, = 8.0,2H), 6.29 (ddd, = 21.2,4.8,1.2,1H), 5.98 (d, = 4.8,1H),
5.32 (ddd, = 52.0,4.0,1.2,1H), 5.20 (s, 1H),4.83 (dd,/= 11.2,4.8,1H), 4.61 (m,
1H), 2.00 (s, 3H); [M+H]+ 449.3.
Step 2; (3 'R)-5-amino-3-(3 '-deoxy-3 '-fluoro-P-D-xylofitranosyl)-3H-thiazolo[4,5-
dJpyrimidin-2'One (79)
87

WO 2006/066080 PCT/US2005/045589
In a manner similar to Example 23, Step 2,43 mg of the title compound 79
was generated in 56 % yield as yellow solid: 1H NMR (400 MHz, DMSO-ck) 8 8.36
(s, 1H), 6.87 (s, 2H), 5.97 (d, 7=4.8,1H), 5.73 (d, J = 5.6,1H), 5.22 (dtd, /=24.4,
5.6,2.0,1H), 5.02 (ddd, J= 52.8,4.4,1.6,1H), 4.07 (m, 2H), 3.62 (m, 2H); [M+H]+
303.6.
Example 28
Preparation of (3'S)-5-amino-3-(2',5 '-di-O-acetyl-3 '-azido-3 '-deoxy-fl-D-
ribofuranosyl)-3H-thiazolo[4,5-d]pyrimidin~2-one($3)

a) Tf2O, py, CH2CI2l -10 °C, 0.5 h
b) NaN3, py, DMF, it, 5 d
c) AcaO, AcOH, H2SO4, rt, 24 h
Stepl: Preparation of (3R)-3-Azido-3-deoxy-l,2-0-isopropyliene-5-0-trityl-j3-D~
ribofuranose (81)
l,2-0-Isopropylidene-5-0-trityl-p-D-xylofuranose (69) [prepared according
to the method of Johnston et al. Tetrahedron Lett. 1995,36, 4341-4344] (3.28 g,
7.58 mmol) was dissolved in CH2CI2 (75 mL) at ambient temperature before it was
cooled to -10 °C. Pyridine (0.86 mL, 10.61 mmol) was added to the solution,
followed by slow addition of trifluorometlianesulfonic anhydride (1.53 mL, 9.10
mmol). After stirring at-10 °C for 1 h the reaction was quenched by slow addition
of 5% NaHSO3 (150 mL) before it was warmed up to room temperature. The layers
were then separated, and the aqueous phase was further extracted with CH2CI2 (2 x
75 mL). The organic layers were combined, dried with MgSCU, then filtered and
concentrated. The residue was azeotroped with toluene (2 x 10 mL), then dried on
high vacuum to afford triflate 80.
88

WO 2006/066080 PCT/US2005/045589
Triflate 80 was dissolved in DMF (100 mL) at ambient temperature. Pyridine
(0.92 mL, 11.37 mmol) was added to the solution, followed by addition of sodium
azide (1.97 g, 30.32 mmol). After 5 d the reaction had gone to completion, the crude
mixture was concentrated. The residue was dissolved in EtOAc (60 mL), washed
with saturated aqueous NH4CI (40 mL). The organic layer was dried over MgSC>4,
filtered and concentrated. The mixture was then purified by ISCO chromatography
(SiO2, 2-15% EtOAc-Hexane), affording 1.80 g (52% for 2 steps) of 81 as a white
solid: *H (400MHz, CDCl3) 8 7.44-7.47 (m, 6H), 7.26-7.32 (m, 6H), 7.24-7.25 (m,
3H), 5.90 (d, = 3.6,1H), 4.77 (t,J=4.4,1H), 4,18-4.22 (m, 1H 3.66 (q, 7=6.0,
1H), 3.52 (dd, = 10.4, 3.2, 1H), 3.20 (dd, / = 10.8,4.0,1H), 1.59 (s, 3H), 1.40 (s,
3H).
Step 2: Preparation of(3R)-l,2,5-Tri-0-acetyl-3-azido-3-deoxy-a,b-D-
ribofuranose (82)
(3R)-3-Azido-3-deoxy-l,2-0-isopropyliene-5-0-trityl-P-D-ribofuranose(81)
(1.20 g, 2.62 mmol) was dissolved in AcOH (30 mL) at ambient temperature. Acetic
anhydride (1.24 mL, 13.10 mmol) was added dropwise to the solution. The resulting
mixture was cooled to 0 °C, followed by slow addition of 1MII2SO4 (2.88 mL, 2.88
mmol). After 24 h the reaction had gone to completion, the crude mixture was
concentrated, then azeotroped with toluene (2 x 10 mL). The residue was dissolved
in CH2CI2 (30 mL), washed with saturated aqueous NaHCO3 (20 mL). The Organic
phase was dried over MgSO4, filtered, and concentrated. The mixture was then
purified by ISCO chromatography (SiO2,2-40% EtOAc-Hexane), affording 0.66 g
(83%) of 82 as a colorless oil: 1H (400 MHz, CDCl3) 8 (a mixture of a and P
isomers) 6.43 (d, J = 4.4), 6.14 (s), 5.34 (d, /= 4.8), 5.21 (dd, 7= 7.6), 4.20-4.37
(m), 4.04-4.10 (m), 2.10-2.20 (m).
Step 3: Preparation of (3'R)-5-amino-3-(2',5'-di-0-acetyl-3'-azido-3'-deoxy-p-
D-ribofuranosyl)-3H-thlazolo[4,5-d]pyrimidin-2-one(S5)
In a manner similar to Example 23, Step 1,288 mg of the title compound 83
was generated in 85% yield as a orange solid: 'H NMR (400 MHz, CDCI3) 8 8.17 (s,
1H), 6.18 (d, /« 2.4,1H), 5.95 (dd, /= 6.4, 2.8,1H), 5.14 (s, 2H), 4.61 (m, 1H),
4.24 (dd, /= 12.0,5.2,1H), 4.17 (m, 2H), 2.12 (s, 6H); [M+H]+ 410.4.
Scheme 6
89

WO 2006/066080 PCT/US2005/045589

a) Tf2O, py. CH2CI2, 0 °C, 0.5 h; Chloroamidine bass, NaH, CH3CN, rt, 50 °c, 12 h
b) NalO4, OsO4, CH3OH/H2O; NaBH4, CH3OH
c) Zn-Cu, AcOH
d) 2M HCI, CH3OH
Example 29
Preparation of (l %2 'S,3'R,4'R)- N'-[7-chloro-2-oxo-3-(2',3'-0-
isopropylidme-4'-vinyl~cyclopentan~r-yl)-2,3-dihydro-thiozolo[4,5-
d]pynmidin-5-yl]-N,N-dimethyl~formamidine (88)

Step 1: Preparation of {I %2'S,3'R,4'R)- N'-[7-chloro-2-oxo-3-( 2',3'-
0-isopropylidene-4'-vinyl-cyclopentan-r-yl)-2,3-dihydro-ihiozolo[4,5-
d]pyrimidin-5-yl]-N.,N-dimethyl~formamidine (85)
(lR,2S,3R,4R)2,3-0-isopropylidene-4-vinyl-cyclopentan-l-ol(84)
[prepared according to the method of Yang et al. J. Org. Chem. 2004, 69, 3993-
3996] (96 mg, 0.52 mmol) was dissolved in CH2C12 (2 mL) and pyridine (10 mL) at
ambient temperature. The solution was cooled to 0 °C, followed by slow addition of
trifluoromethanesulfonic anhydride (115 uL, 0.68 mmol). After 0.5 h, the reaction
had gone to completion, the reation was quenched with H2O (10 mL), then further
diluted with CH2CI2 (10 mL). After the layers were separated, the aqueous phase
was further washed with CH2CI2 (2 x 10 mL). The Organic fractions were combined,
90

WO 2006/066080 PCT/US2005/045589
dried over MgSCU, filtered, then concentrated. The resulting yellowish oil was used
for next step directly.
The above triflate (131 mg, 0.51 mmol) was suspended in CH3CN (10 mL)
at ambient temperature. Sodium hydride (15 mg, 0.62 mmol) was added to the
solution, followed by the addition of a solution of N'-[7-chloro-2-oxo-2,3-dihydro-
thozolo[4,5-flpyrimidm-5-yl)-iV,Ar-dimethyl-formamidine (Chloroamidine base,
160 mg, 0.62 mmol) in CH3CN (8 mL). The reaction was stirred at 50 °C for 12 h
before it was quenched by addition of H2O (5 mL). The resulting mixture was
extracted with EtOAc (3 x 20 mL). The organic fractions were combined, dried over
MgSCU, filtered, then concentrated. The.mixture was then purified by column
chromatography (SiO2, 2-20% EtOAc-Hexane), affording 94.8 mg (43%) of 85 as a
white solid: 1H (400MHz, CDCl3) 5 8.63(s, 1H), 5.92 (m, 1H), 5.09-5.29 (m, 4H),
4.55-4.61 (m, 1H), 3.23 (s, 3H), 3.21 (s, 3H), 2.65-2.77 (m, 1H), 2.42-2.51 (m, 1H),
2.18-2.22 (m, 1H), 1.56 (s, 3H), 1.29 (s, 3H); [M+H]+ 424.1.
Step 2: Preparation of(l 'R,2'S,3'R,4'R)-5-Aminor7-chloro-3-(2',3'-0-
isopropyHdene-4'-hydroxymethyl-cyclopentan-r-yl)-3H-thiazolo[4,5-d]pyrimidin-
2-one (86)
The title compound 86 can be synthesized by first treating
(l'R,2'S3'R,4'R)-N'-i;7-chloro-2-oxo-3-(2',3'-O-isopropylidene-4'-vinyl-
cyclopentan-1' -yl)-2,3-dihydro-thiozolo[4,5-d]pyrimidin-5-yl]-N,N-dimethyl-
formamidine (85) in CH3OH and H2O with sodium periodate and Osmium tetroxide.
The crude product can then be treated with sodium borohydride in CH3OH to afford
86.
Step 3: Preparation of(l 'R,2'S,3'R,4'R)-5-Anuno-3-( T ,y-O-isopropylidene-4'-
hydroxymethyl-cyclopentan-r-yl)-3H-thiazolo[4,5-d]pyrimidin-2-one (87)
The title compound 87 can be synthesized by treating (l'R,2'S,3'R,4'R)-5-
Amino-7-chloro-3 -(2' ,3' -O-isopropylidene-4' -hydroxymethyl-cyclopentan-1' -yl)-
3H-thiazolo[4,5-d]pyrimidin-2-one (86) in AcOH with Zinc copper couple under
various conditions.
Step 4: Preparation of(l 'R,2'S,3'R,4'Ry5-Amino~3-(2',3'-dioxy-4'-hydroxymethyl-
cyclopentan-1 '-yl)-3H-thiazolo[4,5-d]pyritnidin-2-one (88)
The title compound 88 can be synthesized by treating (1 'R,2'S,3'R,4'R)-5-
Amino-3-(2' ,3' -O-isopropylidene-4' -hydroxymethyl-cyclopentan-1' -y\)-3H-
91

WO 2006/066080 PCT/US200S/045589
thiazolo[4,5-d]pyrimidin-2-one (87) in CH3OH with 2M HC1 under various
conditions.
Example 30
Preparation of 5-Amino-3-(3 '-(R)-methoxy~fi-D-xylojuranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (89)
The required sugar, acetic acid l,2,5-tri-'O-acetyl-3)-methoxy-D-
xylofuranose a and /? mixture (91) was prepared as follows:

a. NaR THF, CH3I b. H2SO4, AcOH-Ac2O
Step 1: Preparation of l,2-0-isopropylidene-3-methoxy-5-0-trityl-D-xylojuranose
(90)
Trityl alcohol 69 (5 g, 11.57mmol) was mixed with meihyl iodide (2.5 ml,
34.7 tnmol) in THF (40 ml). Tetrabutylammonium iodide (427 mg) was added and
the mixture cooled in an ice bath. Under a slow stream of nitrogen solid sodium
hydride-oil mixture (1.33g, 60% NaH, 34.7 mmol) was added in small portions. The
reaction was stirred overnight while warming slowly to ambient temperature. The
reaction was carefully poured into a mixture of saturated ammonium chloride and
ice and extracted three times with ethyl ether. The ether portions were combined,
washed with brine, dried (MgSO4), filtered and the solvent evaporated to yield 90 as
a cloudy oil. 1H NMR (400 MHz, CDC13) 5 7.42 7.42 (tn, 6H), 7.26 (m, 9H), 5.85
(d, J=3.6Hz, 1H), 4.54 (d, J=3.6Hz, 1H), 4.38 (m, 1H), 3.785 (d, J=3.2Hz, 1H), 3.42
(m, 1H), 3.35 (m, 4H), 1.53 (s, 3H), 1.336 (s, 3H)
Step 2: l,2,5-tri-O-acetyl-3-methoxy-D-xylofuranose a and ftmixture (91)
The trityl compound 90 (6.1g, 11.57mmol) was dissolved in a mixture of
acetic acid (20 ml) and acetic anhydride (10 ml) and cooled in a cool water bath. A
92

WO 2006/066080 PCT/US2005/045589
mixture of sulfuric acid in acetic anhydride and acetic acid was added (0.5 ml
sulfuric acid, 2.5 ml acetic acid, 2.5 ml acetic anhydride, pre-cooled in an ice bath
before addition) and the mixture stirred at ambient temperature over night. The
reaction was poured onto 400 g of ice water and extracted three times with ethyl
acetate. The organic portions were combined, washed with saturated sodium
bicarbonate, dried (MgSO4), filtered and evaporated to yield a semisolid. This was
purified using flash chromatography on a 120 g silica gel column eluting with a
gradient of ethyl acetate in hexane (10-100%) to give 91 (1.26 g, 4.34 mmol, 38%)
as an oily mixture of anorners. 'H NMR (400 MHz, CDC13) 5 6.39 (d, J=4.4Hz),
6.17 (s), 4.4-4.52 (m), 4.3-4.39 (m), 4.1-4.24 (m), 3.86 (d, /=5.6Hz), 3.45 (s), 3.41
(s), 2.07-2.16 (m).
Step 3: Preparation of 5-Amino-3-(3'--methoxy-fi-D-xylofuranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (89)
hi a manner similar to Example 23, stepl, using l,2,5-tri-O-acetyl-3-
methoxy-D-xylofuranose a and |3 mixture (91), afforded 43 mg (6 %) of 89 as a
white solid: 1H NMR (DMSO-4) 8 2.01 (d, J= 9.2 Hz, 6H), 3.36 (s, 3H), 4.17-4.24
(m, 2H), 4.31-4.37 (m, 2H), 5.86 (d, J = 6 Hz, 1H), 6.14 (dd, J = 4.4,1.6 Hz, 1H),
6.85 (br s, 2H), 8.35 (s, 1H); MS (ESI) [(M + H)1 399.96, Elemental analysis for
(C5H18N4O7S.O.5 H2O): calc'd: C, 44.22; H, 4.70; N, 13.75. Found C, 44.27; H,
4.54; N, 13.60.
Example 31
Preparation of 5-Amino-3-(3 '-octyloxy -fi-D-xylofuranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (92)
The required sugar acetic acid l,2,5-tri-O-acetyl-3(S)-octyloxy-D-
xylofuranose a and b mixture (94) was prepared as follows:
93

WO 2006/066080 PCT/US2005/045589

Step 1: Preparation of l,2-0-isopropylidene-3~octyloxy-5-0-trityl-D-xylofuranose
(5)
Trityl alcohol 69 (5 g, 11.57 mmol) was mixed with octylbromide (3.99 ml,
23.14 mmol) in THF (40 ml). Tetrabutylaramonium iodide (427mg) was added and
the mixture cooled in an ice bath. Under a slow stream of nitrogen solid sodium
hydride-oil mixture (1.33g, 60% NaH, 34.7 mmol) was added in small portions. The
reaction mixture was stirred overnight while warming slowly to ambient
temperature. The reaction was carefully poured into a mixture of saturated
ammonium chloride and ice and extracted three times with ethyl ether. The ether
portions were combined, washed with brine, dried (MgSO4), filtered and the solvent
evaporated to yield a cloudy oil. The oil was purified by flash chromatography on a
120-gram silica gel column using a gradient of ethyl acetate in hexane (1-30%) to
give 93 as a clear oil (2.37 g, 4.72 mmol, 41%). JH NMR (400 MHz, CDC13)
5 7.42 7.42 (m, 6H), 7.26 (m, 9H), 5.85 (d, J=3.6Hz, 1H), 4.50 (d, 7=3.6 Hz, 1H),
4.343 (m, 1H), 3.86 (d, 7=3.6Hz, 1H), 3.45 (m, 2H), 3.32 (m, 2H), 1.54 (m, 3H),
1.41 (m 2H), 1.33 (s, 3H), 1.22 (m, 10H), 0.889 (t, 7=6.8Hz, 3H)
Step 2: Acetic acid l,2,5-tri-0-acetyl-3-octyloxy-D-xylofuranose
a and ft mixture (94)
The trityl compound 93 (4.12 g, 7.57 mmol) was dissolved in a mixture of
acetic acid (35 ml) and acetic anhydride (15 ml) and cooled in a cool water bath. A
mixture of sulfuric acid in acetic anhydride and acetic acid was added (0.5 ml
sulfuric acid, 2 ml acetic acid, 2 ml acetic anhydride, pre-cooled in an ice bath
before addition) and the mixture stirred at ambient temperature over night. The
reaction was poured onto 400 g of ice water and extracted three times with ethyl
acetate. The organic portions were combined, washed with saturated sodium
bicarbonate, dried (MgSO4), filtered and evaporated to yield a semisolid. This was
purified using flash chromatography on a 120 g silica gel column eluting with, a
94

WO 2006/066080 PCT/US2005/04S589
gradient of ethyl acetate in hexane (5-60%) to give 94 (1.12 grams, 2.88mmol, 38%)
as an oily mixture of anomers. 1H NMR (400 MHz, CDC13) 8 6.39 (d, =3.6Hz), 6.1
(s,), 5.19 (m) 4.46-4.52 (m), 4.31-4.43 (m), 4.11-4.25 (m), 3.93 (m), 3.45-3.68 (m),
3.4-3.46 (m), 2.07-2.1 (m), 1.540 (m), 1.27 (m), 0.882 (t, =6.8Hz)
5 Step 3: Preparation of 5-Amino-3-(3'-octyloxy -fi-D-xylofiiranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one(92)
In a manner similar to Example 23, step 1, using Acetic acid 1,2,5-tri-O-
acetyl-3-octyloxy-D-xylofuranose a and (5 mixture (94), afforded 80 mg (11 %) of
92 as a fluffy white solid: 1H NMR (400 MHz DMSO-4) 5 0.84-0.88 (m, 3H), 1.23-
10 1.30 (m, 10H), 1.49-1.52 (m, 2H), 2.00 (s, 3H), 2.02 (s, 3H), 3.41-3.44 (m, 1H),
3.57-3.59 (m, 1H), 4.16-4.21 (m, 1H), 4.30-4.37 (m, 3H), 5.87 (d, J= 5.6, IK), 6.12
(dd, J= 4.4,1.2 Hz, 1H), 6.85 (br s, 2H), 8.35 (s, 1H); MS (ESI) [(M + H)+] found
497.40. Elemental analysis for (C22H32N4O7S): C, 53.21; H, 6.50; N, 11.28. Found
C, 53.52; H, 6.49; N, 11.21.
Example 32
Preparation of 5-Amino-3-(3'-(R)-(2-methoxy-ethoxy), 2',5'-di-O-acetyl-fi-D-
xylofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (95)

The required sugar, acetic acid l,2(5-tri-O-acetyl-3-(2-methoxy-e1hoxy)-
D-xylofuranose a and b mixture (98) as follows:
95

WO 2006/066080 PCT/US2005/045589

a. NaH, THF, 2-bromoethylmethyl ether b. Acetylbromide, Ac2O, c. H2SO4, AcOH-Ac20
Step 1: l,2-0-isopropylidene-3-(2-methoxy-ethoxy)-5-0-trityl-D-xylofuranose (96)
Trityl alcohol 69 (5 grams, 11.57 mmol) was mixed with 2-
bromoethylmethyl ether (2.17 ml, 23.14 ramol) in THF (40 ml).
Tetrabutylammonium iodide (427 mg) was added and the mixture cooled in an ice
bath. Under a slow stream of nitrogen solid sodium hydride-oil mixture (1.33 g,
60% NaH, 34.7 mmol) was added in small portions. The reaction was stirred
overnight while warming slowly to ambient temperature. The reaction was
carefully poured into a mixture of saturated ammonium chloride and ice and
extracted three times with ethyl ether. The ether portions were combined, washed
with brine, dried (MgSO4), filtered and the solvent evaporated to yield a cloudy oil.
The oil was purified by flash chromatography on a 120 g silica gel column using a
gradient of ethyl acetate in hexane (3-30%). The ether product 96 was isolated as a
thick oil (4.54 g, 9.26 mmol, 80%). *H NMR (400 MHz, CDC13) 5 7.42 (m, 6H),
7.26 (m, 9H), 5.86 (d, 7= 3.6Hz, 1H), 4.54 (d, /=4Hz, 1H), 4.37 (m, 1H), 4.35 (m,
1H), 3.95 (d, /=2.8Hz, 1H), 3.64-3.68 (m, 1H), 3.47-3.52 (m, 2H)3.29-3.33 (m,
2H), 3.24 (s, 3H)1.53 (s, 3H), 1.326 (s, 3H)
Step 2: l,2-0-isopropylidene-3-(2-methoxy-ethoxy)-5-0-acetyl-D-xylofuranose (97)
The trityl ether 96 (5.5 g, 11.22 mmol) was dissolved in acetic anhydride (30
ml) and acetyl bromide (2.0 ml, 22.4 mmol) was added. After one hour the reaction
was filtered and the filtrate evaporated to dryness. The residue was purified using
flash chromatography on a 120 g silica gel column using a gradient of ethyl acetate
96

WO 2006/066080 PCTAJS2005/045589
in hexane (10-100%) to yield 1.54 g (5.31 mmol, 47%) of acetate 97. *H NMR (400
MHz, CDC13) 8 5.925 (d, 7=3.6Hz, 1H), 4.58(d, 7=3.6 Hz, 1H), 4.38 (m, 2H), 4.23
(m, 1H), 3.92 (d, 7=3.6Hz, 1H), 3.72 (m, 1H), 3.60 (m, 1H), 3.50 (m, 2H), 3.35 (s,
3H), 2.08 (s, 3H), 1.49 (s, 3H), 1.32 (s, 3H).
Step 3: Acetic acid l,2,5-tri-0-acetyl-3-(2-met}ioxy-ethoxy)-D-xylofuranose
a and f5 mixture (98)
The acetate 97 (1.71 grains, 5.89 mmol) was dissolved in a mixture of acetic
anhydride and acetic acid (1:4,30ml) and cooled in an ice bath. A solution of
suifuric acid in acetic acid (125 uL H2SO4 in 1.0ml acetic anhydride) was added and
the mixture maintained at -10 degrees overnight The cold solution was poured onto
80 g of ice, let stand for 20 minutes and then extracted three times with ethyl acetate.
The organic portions were combined, washed with brine, dried (MgSCU), filtered
and the solvent removed to get 1.94 g of crude product The crude product was
purified using flash chromatography on a 120 g silica gel column eluting with a
gradient of ethyl acetate in hexane (10-75%) to yield 760mg (2.27 mmol, 38%) of
98 as a mixture of anomers. Hi NMR (400 MHz, CDCI3) 8 6.40 (d, J= 4.4 Hz), 6.10
(s), 5.21 (m), 4.47-4.54 (m), 4.33-4.45 (m), 4.16-4.27 (m), 4.03 (m), 3.72-3.85 (m),
3.6-3.7 (m), 3.48-3.54 (m), 3.35-3.48 (m), 2.06-2.11 (m)
Step 4: Preparation of 5-Amino-3-(3'-(2-methoxy-ethoxy), 2',5'-di-O-acetyl-ft'D-
xylofumnosyl)-3H-thiazolo[4,5-d]pyrimidin~2-one (95)
In a manner similar to Example 23, step 1, using Acetic acid 1,2,5-tri-O-
acetyl-3-(2-methoxy-eilwxy)-D-xylofuranose (X and fl mixture (98), afforded 220
mg (42 %) of 95 as a fluffy white solid: JH NMR (400 MHz DMSO-d6) 5 2.04 (d, /
= 8.4 Hz, 6H), 3.29 (s, 3H), 3.49-3.52 (m, 2H), 3.58-3.63 (m, 1H), 3.76-3.81 (m, •
1H), 4.19-4.24 (m, 1H), 4.36-4.43 (m, 3H), 5.88 (d, /= 6 Hz, 1H), 6.18 (dd, /= 3.6,
2 Hz, 1H), 6.87 (br s, 2H), 8.38 (s, 1H); MS (ESI) [(M + H)*] found 443.31.
Elemental analysis for (CnHad^OgS-O.l H2O-0.2 EtOAc): C, 46.29; H, 5.19; N,
12.13. Found C, 46.09; H, 5.25; N, 11.72.
Example 33
Preparation of5-Amino-3-(3'-Butoxy-a-D-xylofuranosyl)-3H-thiazolo[4,5-
d]pyrimidin-2-one (99)
97

WO 2006/066080 PCT/US2005/045589
The required sugar acetic acid l,2,5-tri-O-acetyl-3-butoxy-D-xylofuranose
a and b mixture (101) was prepared as follows:

a. NaH, THF, nbutyliodide b. H2SO4, AcOH-Ac2O
Step 1: Preparation of l,2-0-isopropylidene-3-butoxy-5-0-acetyl~D-xylofarafiose
(100)
Trityl alcohol 69 (5 grams, 11.57 mmol) was mixed with n-butyliodide (2.6
ml, 23.14 mmol) in THF (40ml). Tetrabutylammonium iodide (427 mg) was added
and the mixture cooled in an ice bath. Under a slow stream of nitrogen solid sodium
hydride-oil mixture (1.33g, 60% NaH, 34.7 mmol) was added in small portions. The
reaction was stirred overnight while wanning slowly to ambient temperature. The
reaction was carefully poured into a mixture of saturated ammonium chloride and
ice and extracted three times with ethyl ether. The ether portions were combined,
washed with brine, dried (MgSCU), filtered and the solvent evaporated to yield a
cloudy oil. The oil was purified by flash chromatography on a 120 g silica gel
column using a gradient of ethyl acetate in hexane (1-30%) to give 100 as a clear oil
(2.32 g, 4.75 mmol, 41%). JH NMR (400 MHz, CDC13) 5 7.42 (m, 6H), 7.26 (m,
9H), 5.86 (d, i=3.6Hz, IE), 4.51 (d, 7=3.6Hz, 1H), 4.35 (m, 1H), 3.86 (d, J=3.6Hz,
1H), 3.46 (m, 2H), 3.29 (m, 2H), 1.54 (m, 3H), L38 (m, 2H), 1.33 (s, 3H), 1.23 (m,
2H),0.83(t,J=7.6Hz,3H)
Step 2: Preparation Acetic acid l,2,5-tri-O-acetyl-3-butoxy-D-xylofuranose
aand fi'mixture (101)
The trityl compound 100 (2.32 g, 4.75 mmol) was dissolved in 5% acetic
anhydride in acetic acid (50ml), cooled in a cool water bath and 0.02 ml of sulfuric
98

WO 2006/066080 PCT/US2005/045589
acid was added and the mixture stirred overnight at ambient temperature. The
reaction mixture was poured onto 150grams of ice and extracted three times with
methylene chloride. The organic portions were dried (MgSO/t), filtered and taken to
dryness with toluene to give 3.19 g of a semi-solid. This was purified using flash
chromatography on a 50 g silica gel column eluting with a gradient of ethyl acetate
in hexane (5-75%) to give 101 (0.760 g, 2.29 mmol, 48%) as an oily mixture of
anomers. 1H NMR (400 MHz, CDC13) 5 6.38 (d, /=3.6Hz), 6.11 (s), 5.2 (m), 4.50
(m), 4.31-4.42 (m), 4.13-4.25 (m), 3.93 (d, /=3.6Hz), 3.5-3.7 (m), 3.4-3.47 (m),
2.06-2.15 (m), 1.51-1.55 (m), 1.3-1.4 (m), 0.89-0.94 (m)
Step 3: Preparation of 5-Amino-3~(3 ')-Butoxy-2', 5 '-di-O-acetyl - a-D-
xyloJuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (102)
In a manner similar to Example 23, step 1, using Acetic acid 1,2,5-tri-O-
acetyl-3-butoxy-D-xylofuranose a and p mixture (101), afforded 40 mg (8 %) of
102 as a white solid. Taken crude on to step 2.
Step 2: Preparation of5-kmino-3-{3'-Butoxy-fcD-xylofuranosyl)-31i-thiazolo[4,5-
d]pyrimidin-2-one (99)
In a manner similar to Example 23, step 2,102 afforded 5 mg (15 %) of 99
as a white solid: JH NMR (400 MHz, (CDC13) 8 0.84 (t, J = 7.2 Hz, 3H), 1.27-1.32
. (m, 2H), 1.51-1.54 (m, 2H), 3.20 (t, J= 9.2 Hz, 1H), 3.35 (t, /= 10.8 Hz, 1H), 3.72-
3.84 (m, 3H), 3.97-4.01 (m, 1H), 4.74 (t, i=9.2 Hz, 1H), 5.17 (br s, 2H), 5.38 (d, /
= 9.2 Hz, 1H), 7.96 (s, 1H); MS (ESI) [(M + H)"*] found 356.80.
Example 34
Preparation of 5-Amno-3-(3'-metltyl, 2',3\5'-tri-0-acetyl-P-D-xylofuranosyl)-3H-
thiazolo[4,5~d]pyrimidin-2-one (103)

The preparation of the required sugar, 1,2,3,5 tetra-O-acetyl-3(S)-methyl D-
xylofuranose a and/? mixture (105), was prepared as follows:
99

WO 2006/066080 PCT/US2005/045589

a. i. Ac2O, pyridine, ii.H2SO4, ACaO-AcOH
Step 1: 1,2,3,5 tetra-O-acetyl-3-methyl D-xylofuranose aand fi mixture (105)
The diol 104 [prepared as described by Lu and Just; Tetrahedron Letters 41
(2000) 9223-9227] (1.69 g, 8.28 mmol), was dissolved in tnethylene chloride (25
ml) and pyridine (4.7 ml) was added. Acetic anhydride (3.9 ml, 41 mmol) was
added along with DMAP (50 rag) and this mixture was stirred overnight at ambient
temperature. The reaction was diluted with methylene chloride and washed with
saturated ammonium chloride. The aqueous phase was extracted twice more with
methylene chloride, the organic portions combined, dried (MgSCU), filtered and
evaporated to give a colorless oil. The oil was dissolved in 5% acetic anhydride in
acetic acid (68 ml) and sulfuric acid (0.02 ml) was added and this was stirred
overnight at ambient temperature. The reaction was poured onto 150 g of ice,
extracted three times with methylene chloride, the organic phases combined, washed
twice with saturated sodium bicarbonate, dried (MgSO4), filtered and evaporated to
get 2.84 g of an oil. The residue was purified using flash chromatography on a 120
g silica gel column using a gradient of ethyl acetate in hexane (5-75%) to yield 1.2 g
(3.61 mmol, 44%) of 105 as a clear oil whose spectra are consistent with a mixture
of anomers. 'H NMR (400 MHz, CDC13) 8 6.41 (d, 7=4.8 Hz), 6.03 (d, 7=1.2 Hz),
5.75 (d, 7=0.8Hz), 5.49 (d, 7=5.2 Hz), 4.37-4.45 (m), 4.2-4.29 (m), 2.03-2.135 (m),
1.637 S), 1.624 (s)
Step 2: Preparation of5-Amino-3-(3'-methyl, 2,'3',5'-tri~O-acetyl-fi-D-
xylofumnosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (103)
In a manner similar to Example 23, step 1, using 1,2,3,5 tetra-O-acetyl-3-
methyl D-xylofuranose a and (3 mixture (105), afforded 170 mg (28 %) of 103 as a
white solid: 1H NMR (400 MHz DMSO-d6) 51.57 (s, 3H), 2.03 (s, 6H), 2.07 (s,
3H), 4.04 (dd, 7 = 8.0,2.8 Hz, 1H), 4.24 (m, 1H), 4.41 (dd, 7 = 12.0,2.8 Hz, 1H),
5.73 (d, 7=4.8 Hz, 1H), 6.24 (d, 7 = 4.4 Hz, 1H), 6.89 (br s, 2H), 8.36 (s, 1H); MS
100

WO 2006/066080 PCT/US2005/045589
(ESI) [(M + H)4] found 441.08. Elemental analysis for (Ci7H2oN4OsS-0.3 H2O): C,
45.80; H, 4.66; N, 12.57. Found C, 45.84; H, 4.50; N, 12.47.
Example 35
Preparation of 5-Amino-3-(5' -(1,2-diacetoxy-ethyl), 2',3'-di-O-acetyl-b-D-
glucofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2~one(106)

The required sugar, penta-O-acetylglucofuranose (108) was prepared as
described below.

Stepl; Penta-O-acetylglucofuranose (108)
l,2-O-Isopropylidine-a-D-glucofranose (107) (5 g, 22.7 mmol) was
dissolved in acetic acid (180 ml) and acetic anhydride (2L5 ml), cooled in a cool
water bath and sulfuric acid (0.02 ml, 98%) was added and the mixture stirred 24
hours at ambient temperature. The mixture was poured onto 500 g of ice, water was
added and this was extracted four times with methylene chloride. The organic
portions were combined, washed twice with saturated sodium bicarbonate, dried
(MgSO4), and filtered to yield an oily residue. This was purified on a 120 g silica
gel column using a gradient of ethyl acetate in hexane (20-100%) to give 5.33g
(13.66 mmol, 60%) of 18 as an oil whose spectra are consistent with a mixture of
anomers. 1H NMR (400 MHz, CDC13) 5 6.41 (d, 7=3.6Hz), 6.12 (s), 5.58 (m), 5.41
(d,/=3.6Hz), 5.20-5.37 (m), 4.56-4.62 (m), 4.02-4.18 (m), 2.00-2.13 (m).
Step 2: Preparation of 5-Amino-3-(5'-(1,2-diacetoxy-ethyl), 2',3'-di-O-acetyl-j3-D-
glucofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (106).
101

WO 2006/066080 PCT/US2005/045589
In a manner similar to Example 23, step 1, using penta-O-
acetylglucofuranose (108), afforded 80 mg (9 %) of 106 as a white solid: lH NMR
(400 MHz CDC13) 8 2.07 (s, 6H), 2.09 (s, 3H), 2.14 (s, 3H), 4.03-4.07 (m, 1H), 4.47
(dd, J= 6.4,2 Hz, 1H), 4.67 (dd, = 12.4,2.0 Hz, 1H), 5.31 (br s, 2H), 5.58-5.6 (m,
1H), 5.68 (m 1H), 5.97 (d, = 5.6 Hz, 1H), 6.10 (dd, / = 3.6,2 Hz, 1H), 8.16 (s,
1H); MS (ESI [(M + H)+] found 499.40. Elemental analysis for (CNS-O.l
IPA): C, 45.95; H, 4.56; N, 11.11. Found C, 45.92; H, 4.76; N, 10.80.
Example 36
Preparation of 5-Amino-3-(3'-acetoxymethyl-, 2 ',3',5'-tn-O-acetyl-j3-D-
xylofuranosyl)-3H-thiazolo[4,5-d]pyrimidin-2-one (109)

The required sugar, tetra-O-acetyl-3-acetoxymethyl-D-xylofuranos
a and /? mixture (113) was prepared as follows:

a. NaOH, dioxane b. Acetic Anhydride, Pyldine, c. H2SO4, AC2O-ACOH
Step 1: l,2-0-isopropylidfene-3(S)-(hydwxy-hydroxymethyl)-D-xylofuranose (111)
The epoxide 110 [prepared as described by Lu and Just; Tetrahedron Letters
41 (2000) 9223-9227] (1.68 g, 8.3 mmol) was dissolved in dioxane (9 ml) and 1.0M
NaOH was added (16.6 ml, 16.6 mmol) and the reaction heated to 50 degrees for 30
minutes. The reaction was cooled to ambient temperature, 16.6 ml of 1.0 M HC1
102

WO 2006/066080 PCT/US2005/045589
and 100 ml of absolute ethanol were added, stirred 5 minutes and the mixture
evaporated under vacuum to give a solid. The solid was suspended in 200 ml of
CH2CI2 and sonicated to give a very fine suspension of solid. This was dried
(MgSO4), filtered through Celite and evaporated to give 111 as a thick oil (1.81
grams, 8.22 mmol, 99%). H NMR (400 MHz, CDC13) 8 5.94 (d, =3.6Hz, 1H),
4.4 (d, = 4Hz, 2H), 4.03 (m, 2H), 3.83 (d, = 11.6,1H), 3.78 (d, J=12Hz, 1H),
2.66 (bs, 2H, OH), 1.7 (bs, 1H, OH), 1.52 (s, 3H), 1.33 (s, 3H)
Step 2: l,2-0-isopropylidene-3-(acetoxy-methylacetoxy)-5-0-acelyl-D'xylofuranose
(112)
The triol 111 (l,81g, 8.22 mmol) was dissolved in pyridine (30 ml), acetic
anhydride (7.75 ml, 82 mmol) was added followed by DMAF (50 rng) and the
mixture stirred for 72 hours. The volatiles were evaporated under vacuum and the
residue partioned between methylene chloride and saturated ammonium chloride.
The aqueous phase was extracted twice methylene chloride and the organic portions
combined, dried (MgSO4), filtered and the solvent evaporated to get 2.83 g of an oil.
The residue was purified using flash chromatography on a 120 g silica gel column
using a gradient of ethyl acetate in hexane (10-80%) to yield 1.82 g (5.26 mmol,
64%) of 112 as a clear oil. %). H NMR (400 MHz, CDCI3) o 5.92 (d, S=3.6Hz,
. 1H), 5.02 (m, 2H), 4.51-4.58 (m, 2H), 4.35 (dd, J=SAEz, 7=2.8Hz, 1H), 4.16-4.22
(in, 1H), 2.10 (s, 3H), 2.09 (s, 3H), 2.08 (s, 3H), 1.53 (s, 3H), 1.32 (s, 3H)
Step 3: Tetra-O-acetyl-3-(acetoxymetJtyl)-D-xylofuranose, a and J3 mixture (113)
The triacetate 112 (1.74 g, 5.01 mmol) was dissolved in acetic acid (45 ml),
acetic anhydride was added (2.37 ml, 25 mmol) followed by sulfuric acid in acetic
acid (0.5 ml of a 1.0M solution, 0.5 mmol) and this mixture was stirred overnight at
ambient temperature. The reaction was diluted with methylene chloride (70 ml) and
washed with water. The water layer was extracted twice with methylene chloride.
The organic portions were combined, transferred to a large beaker and saturated
sodium bicarbonate was added. To this was added solid sodium bicarbonate until no
more bubbling is observed. Separate the organic phase, extract the aqueous phase
with methylene chloride, combine the organic phases and dry (MgSO4), filter and
evaporate to get an oil that was further taken to dryness with toluene to yield 113 as
a clear oil (1.91 g, 3.89mmol, 97%) whose NMR is consistent with a mixture of
anomers. 1H NMR (400 MHz, CDC13) 8 6.42 (d, =4.4Hz), 6.05 (d, 7=1.6Hz), 5.79
103

WO 2006/066080 PCT/US2005/045589
(d, y=1.6Hz), 5.5 (d, 7=4.4Hz), 4.89-4.93 (m), 4.12-4.58 (m), 2.04-2.2 (many
singlets).
Step 4: Preparation of 5-Amino-3-(3 '-acetoxymethyl-, 2',3 ',5 '-tri-0-acetyl-/3-D-
xylofuranosyl)-3H-ihiazolo[4,5-d]pyrimidin-2-one (109)
In a manner similar to Example 23, step 1, using tetra-O-acetyl-3-
acetoxymethyl-D-xylofuranos a and (3 mixture (113) afforded 272 nig (37 %) of 109
as a white solid: lE NMR (400 MHz (DMSO-&) 8 2.03 (s, 3H), 2.05 (s, 3H), 2.07
(s, 3H), 2.09 (s, 3H), 4.20 (m, 1H), 4.44-4.57 (m, 3H), 4.79 (d, J = 12.4 Hz, 1H),
5.84 (d, J = 5.6 Hz, 1H), 6.38 (d, J = 6 Hz, 1H), 6.87 (br s, 2H), 8.37 (s, 1H); MS
(ESI) [(M + H)+] found 499.12. Elemental analysis for (C-O.l H2O): C,
45.61; H, 4.47; N, 11.20. Found C, 45.93; H, 4.44; N, 10.83.
Example 37
Preparation of 5-Ambw-3-(2 ',3',5 '-tri-0-acetyl-fi-D-xylofumnosyl)~3H-
thiazolo[4,5-d]pyrimidin-2-one (114)

Step 1: Preparation of5-Amino-3-(2,'3,'5'-tn-0-acetyl-b-D-xylofuranosyl)~3H-
thiazolo[4,5-d]pyrimidin-2-one (114)
In a manner similar to Example 23, step 1, using commercially available
tetta-O-acetylxylofuranose, afforded 110 mg (14 %) of 114 as a white solid: !H
NMR (400 MHz (CDC13) 5 2.08 (s, 3H), 2.10 (s, 3H), 2.18 (s, 3H), 4.42-4.45 (m,
2H), 4.52-4.56 (m, 1H), 5.13 (br s, 2H), 5.49 (dd, /=3.6, 2.4 Hz, 1H), 6.00 (d, 7.=
5.2 Hz, 1H), 6.22 (dd, = 4,1.6 Hz, 1H), 8.15 (s, 1H); MS (ESI) [(M + JXf] found
426.93. Elemental analysis for (CHOSS): C, 45.07; H, 4.25; N, 13.14. Found
C, 44.86; H, 4.17; N, 13.05.
Example 38
5-Amino-3-(3 '-C-methyl-p-D-ribofuranosyl)thiazolo[4,5-dJpyrimidine-2,7(3H, 6H)-
dione (117)
104

WO 2006/066080 PCT/US2005/045589

Stepl) Preparation of5-Amino-3-(3'-O-acetyl-2',5'-di-O-benzoyl -3'-C-methyl-fi-D-
ribofuranosyl)thiazolo[4,5-d]pyrmidine-2,7(3H,6H)-dione (116)

a. BSA, MeCN, rt, 1 h; + sugar, TMSOTf, 60" C, 1 h, 45%.
5-Amino-3H,6H-thiazolo[4,5-d]pyrimidine (116 mg, 0.631 mtnol), 3'-C-
methyl-ribfuranose 11 (272 mg, 0.598 mmol) [prepared according to the method of
Giradet, et. al. J Med Chan. 2000,43,3704-3713], BSA (0.462 ml, 1.89 mmol)
and acetonitrile (5 mL) were mixed vigorously at ambient temperature for 40 min.
Once a homogeneous solution was obtained, the reaction was charged with TMSOTf
(0.171 mL, 210 mg). Then the reaction was heated to 60° C. After 1 h the solvent
was removed by rotary evaporation. The resultant solid was dissolved in ethyl
acetate (10 mL) and extracted with saturated sodium bicarbonate (2x5 mL). The
aqueous phase was then back extracted with ethyl acetate (5 mL) and the organic
layers were combined. A solid impurity promptly precipitated out of the organic
phase, this was filtered off and discarded. The organic phase was concentrated and
the resultant solid then triturated in ether (5 mL) yielding 167 mg (45%) of tan solid:
]H NMR (400 MHz, 4-DMSO) 812.2 (br s, 1H), 8.01 (m, 4H), 7.83 (m, 2H), 7.53
(m, 4H), 7.28 (br s, 2H), 6.4 (m, 1H), 6.02 (m, 1H), 4.82-4.67 (m, 2H), 3.38 (s, 1H),
2.01(s, 3H), 1.98 (s, 3H); [M+Hf m/z 581.
Step 2) Preparation of 5-Amino-3-(3'-C-methyl-P-D-ribofuranosyl)thiazolo[4,5-
d]pyrimidine-2,7(3H,6H)-dione (117)
105

WO 2006/066080 PCT/US2005/045589

b. K2CO3, MeOH, rt, 74%.
Nucleoside triester 116 (100 mg, 0.172 mmol) was dissolved in methanol (5
mL) and K2CO3 (28.6 mg, 0.207 mmol) was added. The reaction progressed for 16
h. The reaction was neutralized with acetic acid (24.8 mg, 0.412 mmol). Then the
solvent removed by rotary vacuum and the solid submitted to HPLC purification
(MeCN-H2O) yielding 42 mg (74%) of white solid: H NMR (400 MHz, d 8 11.20 (s, 1H), 6.89 (br s, 2H), 5.80 (d, = 8.0,1H), 5.36 (d, J = 6.0, III), 4.77 (t, J
= 8.0,1H), 4.62 (s, 1H), 4.48 (m, 1H), 3.75 (m, 1H), 3.58-3.44 (m, 2H), 1.19 (s,
3H); Analysis calc'd for C14H14N4O6S 0.125 H2O • 0.125 HCO2H: C, 39.33; H,
4.34; N, 16.43; S, 9.40. Found: C, 39.77; H, 4.81; N, 15.02; S, 9.69; [M+H]+ m/z
331.
Example 39
5~AminO'3-(2'-C-methyl-b-D-ribofuranosyl)-3H,6H-thiazolo[4,5~d]pyrimidine-
2,7-dione (120)

120
Step 1) Preparation of5'Amino-3-(2,'3',5'~tn-0-benzoyl-2'-C-methyl-P-D-
ribofuranosyt)-3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-dione (119)
106

WO 2006/066080 PCT/US2005/045589

a. BSA, MeCN, 80° C, 2.5 h; + sugar, SnCI4,80" C, 1.5 h, 42%.
To a suspension of heterocycle 4 (268 mg, 1.44 mmol) in anhydrous MeCN
(8 inL) at rt was added BSA (971 uL, 3.93 mmol). The resultant mixture was heated
to 80° C for 2.5 h whereupon 2-C-methyl-P-D-ribofiiranose 118 [prepared according
to Wolfe et al. Org. Chem. 1997,62,1754-1759] (760 mg, 1.31 mmol) was added
as a solution in MeCN (6 mL). To this mixture was added SnCU (276 uL, 2.35
mmol), and stirring at 80° C was continued for an additional 1.5 h. TLC analysis
with 10% MeOH-CHCl3 indicated that the reaction was complete. The mixture was
cooled to rt, diluted with EtOAc (150 mL), and partitioned with a 1:1 mixture (100
mL) of brine-NaHCO3. The aqueous phase was further extracted with EtOAc (50
mL), and the combined organic phases were dried over Na2SO4, filtered and
concentrated. The residue was submitted to HPLC (SiO2,0-4% MeOH-DCM) to
afford 353 mg (42%) of a white solid: JH (400 MHz, DMSO-^) 8 11.3 (br s, 1H),
7.93-8.08 (br m, 3 H), 7.85-7.87 (m, 2 H), 7.33-7.66 (m, 10 H), 6.97 (br s, 2H), 6.64
(s, 1H), 6.16-6.26 (br m, 1H), 4.56-4.79 (br m, 3H), 1.79 (s, 3H); M4 m/z 642.
Step 2) Preparation of 5-Amino-3-(2'-C-methyl-ft-D-ribofuranosyl)-3H,6H-
thiazolo[4,5-d]pyrimidine-2,7-dione (120)

A mixture of nucleoside triester 119 (209 mg, 0.325 mmol) and MeOH (10
mL) saturated with NH3 (g) at -30° C was stirred in a sealed tube for 48 h. The
mixture was warmed to rt, depressurized, concentrated then submitted to HPLC
purification (MeCN-H20), affording 36 mg (34%) of the title compound as a white
107

WO 2006/066080 PCT/US2005/045589
solid after lyophilization: ]H (400 MHz, DMSO-d6) 8 11.18 (s, 1H), 6.92 (br s, 2H),
5.95 (s, 1H), 5.18-5.28 (br m, 1H), 4.75 (br s, 1H), 4.53 (dd, / = 11.3, 5.46, 1H),
3.95 (br s, 1H), 3.73-3.78 (m, 1H), 3.29 (br s, 2H), 1.04 (s, 3H); [M+H]+ m/z 331.
Example 40
5-Amino-3-(3 '-deoxy-3-D-ribofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-
dione (122)

Step 1) Preparation of5-Amino-3-(2',5'-di-0-acetyl-3'-deoxy~8-D-ribofuranosyl)-
3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione (121)

a. BSA, MeCN, rt, 1 h; + sugar, TMSOTf, 60" C, 1 h, 45%.
To a suspension of heterocycle 6 (4.60 g, 25.0 nrmol) and deoxyribofuranose
11B (5.42 g, 20.8 mmol) in MeCN (83 mL) at rt was added BSA (15.3 mL, 62.5
mmol). The resultant mixture was immersed into a 40° C oil bath for 1.5 h, and
TMSOTf (5.65 mL, 31.2 mmol) was added dropwise. The thick reaction mixture
was immersed into an 80° C oil bath and stirred for 2.5 h whereupon it was
concentrated via rotary evaporation to a residue that was partitioned between EtOAc
(300 mL) and pH 7 buffer (100 mL). The organic phase was dried over Na2SO4 and
concentrated to a residue that was triturated with EtO Ac and then filtered to yield
2.31 g (29%) of fine white solid. The filtrate was concentrated to a residue that was
submitted to flash chromatography (SiO2,0-6% MeOH-DCM) to afford 1.12 g
(14%) of very fine pale yellow solid. Altogether, there was a 43% combined yield of
nucleoside 121: 1H (400 MHz, DMSO-40 811.23 (s, 1H), 6.95 (br s, 2H), 5.79 (d, /
= 2.0,1H), 5.59 (d, /a 7.2,1H), 4.28-4.34 (m, 1H), 4.22 (dd, /= 3.2,12.0,1H),
108

WO 2006/066080 PCT/US2005/045589
3.99 (dd, J = 6.4,11.6,1H), 2.56-2.65 (m, 1H), 2.04 (s, 3H), 1.98 (s, 3H), 1.97-2.04
(m, 1H); [M+H]+ m/z 384.8. Analysis cal'd for: Ci4Hi6N4O7S "0.5 H2O: C, 42.74; H,
4.36; N, 14.24; S, 8.15. Found: C, 42.72; H, 4.22; N, 14.15; S, 8.19.
Step 2) Preparation of5-Amino-3-(3'-deoxy-/S-D-ribofifranosyl)-3H,6H-
thiazolo[4,5-d]pyrimidine-2,7-dione (122)

b. KaCO3, MeOH, rt, 74%.
To a suspension of nucleoside diester 121 (1.91 g, 4.97 mmol) in MeOH (50
mL) at rt was added K2CO3 (820 mg, 5.97 mmol). The reaction mixture was stirred
for 18 h, then quenched with HO Ac (0.68 mL, 12 mmol), stirred 30 min, and finally
concentrated via rotary evaporation. The residue was azeotroped with toluene (3 x
50 mL), and then triturated with water (250 mL). The solid material was filtered,
washed with water (2 x 250 mL), air dried, triturated with ether (250 mL), and
filtered to provide 1.17 g (62%) of nucleoside 122 as an off-white solid: H (400
MHz, DMSO-4) 511.26 (br s, 1H), 6.93 (br s, 1H) 7.14 (d, = 2.4,1H), 5,39 (d,
= 4.4,1H) 4.74-4.79 (m, 1H), 4.65 (t, = 5.6,1H), 4.09-4.16 (m, 1H), 3.41-3.43
(m, 2H), 2.23-2.30 (m, 1H), 1.78 (ddd, J = 2.4,6.4, 8.4,1H) 1.76-1.81 (m, 1H);
[M+H]+ @ m/z 301.5. Analysis cal'd for: Q0H12N4O5S' 1.25 H2O: C, 37.20; H,
4.53; N, 17.36; S, 9.93. Found: C, 37.06; H, 4.27; N, 17.14; S, 9.84.
Example 41
5-Amino-3-(2 '~deoxy-]3-D-ribofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-

109

WO 2006/066080 PCT/US2005/045589
Step 1) Preparation of 5-N-Acetyl-amino-3-3-D-ribofuranosyl)-3H,6H-
thiazolo[4,5-d]pyrimidine-2,7-dione

To a suspension of isatoribioe tetraacetate 1,23 [CAS # 533897-42-6,
prepared according to Webber et. al US Patent 6,924,271] (12.3 g, 25.4 rnmol) in
MeOH (180 raL) was added concentrated NH4OH (180 mL). The resultant mixture
was stirred 1 h whereupon it was concentrated and submitted to flash
chromatography (SiO2,15-30% IPA-CHCb) to afford 2.50 g (27%) of acetamide
124 as a white solid: [M+H]+ m/z 359.
Step 2) Preparation of5-N-Acetyl-aniino-3-(5'-0-tert~butyldimethylsilyl-p-D-
ribofiiranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidine~2,7-dione

To a solution of triol 124 (2.48 g, 6.93 mmol) in DMF (15 mL) was added
sequentially imidazole (943 mg, 13.9 mmol) and TBSC1 (1.04 g, 6.93 mmol) at rt.
The resultant mixture was stirred 1 h, then diluted with EtOAc (300 mL) and
extracted with water (2 x 100 mL) then brine (100 mL). The organic phase was dried
over Na2SO4, concentrated and triturated with ether to afford 2.18 g (67%) of
siloxane 125 as an off-white solid: (400 MHz, DMSO-^) 512.16 (br s, 1H),
11.81 (br s, 1H), 5.81 (d,=4.77,1H), 5.34 (d, J = 5.13,1H), 5.03 (d,= 5.50,
1H), 4.79 (dd, J = 10.3,5.13,1H), 4.13 (dd, = 10.6,5.5,1H), 3.71-3.78 (m, 2H),
3.40-3.64 (m, 1H), 2.18 (s, 3H), 0.84 (s, 9H), 0.00 (s, 6H); [M+H]+ m/z 473.
Step 3) Preparation of5-N-Acetyl-amino-3-(5'-O-tert-butyldimeihylsilyl-2',3'-
thioxo-3-D-ribofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-dione
110

WO 2006/066080 PCT/US2005/045589

To a solution of diol 125 (1.00 g, 2.12 nunol) in MeCN (50 mL) was added
TCDI (754 mg, 4.23 mmol) atrt. The resultant mixture was stirred for 18 h
whereupon it was concentrated, submitted to flash chromatography (SiOo, 40%
EtOAc~CHCl3), and triturated with ether to afford 730 mg (67%) of a white solid: 1H
(400 MHz, DMSO-dd) o 12.18 (br s, 1H), 11.75 Cor s, 1H), 6.21-6.24 (m, 211), 5.79
(br s, 1H), 4.35 (br s, 1H), 3.72 (br d, /= 6.6,2H), 2.21 (s, 3H), 0.84 (s, 9H), 0.01 (s,
6H);[M+H]+m£515.
Step 4) Preparation of 5-N-Acetyl-amino-3-(5'-O-tert-butyldimethyl$ilyl-2'-deoxy-b-
D-ribofuranosyl)-3H,6H-ihiazolo[4,5-d]pyrimidine-2,7-dione

To a suspension of thiocarbonate 126 (712 mg, 1.38 mmol) and BU3S11H
(2.66 mL, 10.0 mmol) in anhydrous toluene (140 mL) was added AIBN (30 mg,
0.18 mmol) at rt. The mixture was immersed into a 130° C oil bath for 15 min then
removed, cooled, concentrated and submitted to flash chromatography (SiCh, 80-
100% EtOAc~CHCl3) to afford 450 mg (71%) of a mixture (2:1) of 2'-deoxy and 3'-
deoxy regioisomers (major isomer reported): 1H (400 MHz, JMSO-di) 5 12.12 (br
s, 1H), 11.82 (br s, 1H), 6.26 (t,/ = 7.0,1H), 5.22 (d, 7=4.0,1H), 4.31-4.34 (m,
1H), 3.69-3.75 (m, 2H), 3.57-3.62 (m, 1H), 2.93-2.99 (m, 1H), 2.18 (s, 3H), 2.00-
2.18 (m, 1H), 0.84 (s, 9H), 0.00 (s, 6H); [M+Hf m/z 457.
111

WO 2006/066080 PCT/US200S/045589
Step 5) Preparation of 5-N-Acetyl-amino-3-(2'-deoxy~b-D-ribofiiranosyl)-3Ht6H-
thiazolo[4,5-d]pyrimidine-2,7-dione

To a suspension of the regioisomers (744 mg, 1.60 mmol) from Step 4
(above) in MeCN (30 mL) at rt was added 48% aqueous HF (1.67 mL). The reaction
mixture was stirred 1 h whereupon it was concentrated to a purple residue that was
submitted to flash chromatography (SiO2,1.5-15% MeOH-DCM), affording 503 mg
(92%) of a mixture of regioisomers that was further purified via HPLC (MeCN-
H2O) to provide 169 mg (31%) of nucleoside 129 as a white solid after
lyophilization: 'H (400 MHz, DMSO-d6) 5 12.14 (s, 1H), 11.85 (s, 1H), 6.26 (t, J =
7.0,1H), 4.30-4.32 (m, 1H), 3.69-3.71 (m, 1H), 3.38-3.53 (m, 4H), 2.92-2.98 (m,.
1H), 2.19 (s, 3H), 1.97-2.03 (s, 1H); [M+H]+ ni/z 343.
Step 6) Preparation of5-Amino-3-(2'-deoxy-fl-D-ribofuranosyl)-3H,6H-
thiazolo[4,5-d]pyrimidine-2,7-dione

To a solution of acetamide 129 (169 mg, 0.494 mmol) in MeOH (10 mL)
was added K2CO3 (158 mg, 1.14 mmol) at rt. The resulting mixture was stirred for 8
h whereupon it was quenched with HOAc (137 uL, 2.40 mmol), concentrated and
submitted to HPLC (MeCN-H2O) to afford 125 mg (84%) of the title compound 130
as a white solid after lyophilization: !H (400 MHz, TMSO-d6) 511.16 (s, 1H), 6.90
(br s, 2H), 6.22 (t,= 7.0,1H), 4.27-4.31 (m, 1H), 3.67-3.71 (m, 1H), 3.52 (dd, / =
112

WO 2006/066080 PCT/US2005/045589
11.3, 5.5,1H), 3.40 (dd, = 11.7,6.2,1H), 2.86-2.93 (m, 1H), 1.97 (ddd,J = 12.9,
7.0,3.5,1H); [M+H]+ m/z 301. Analysis calc'd for C10H2N4O5S H2O: C, 37.73; H,
4.43; N, 17.60; S, 10.07. Found: C, 38.13; H, 4.27; N, 17.40; S, 9.89.

a) BSA, TMSOTf, CH3CN, 80 °C, 3-4 h
b) K2CO3, DMF, rt, overnight
Example 42
Preparatwn of 5-Amim-3~(3t~deoxy-3'-methylidene-fi-D~ribofuranosyl)-3H,6H-
thiazolo[4,5-dJpyrimidine-2,7-dione (132)

Step 1: Preparation of5~Amino-3-(2'~0-acetyl-5'-0-benzoyl-3'-deoxy-3'-
methylidene-/3-D-ribojurcindsyl)-3Ht6H-thiazolo[4,5-d]pvrimidine-2,7-dione (131)

l,2-di-0-acetyl-5-0-benzoyl-3-deoxy-3-methylidene-a,p-D-ribofuranose
(127) (132 mg, 0.39 mmol) [prepared according to the method of Girardet et al..
Med. Citem. 2000,43,3704-3713] was dissolved in acetonitrile (5 mL) at ambient
temperature. 5-Amino-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione (1) (73 mg, 0.39
mmol) was added, the mixture was then stirred for 0.5 h before it was heated to 40
°C. After 5 min at 40 °C, BSA (0.29 mL, 1.18 mmol) was added and the mixture
was stirred for another 0.5 h. The mixture was then heated to 80 °C. TMSOTf
113

WO 2006/066080 PCT/US2005/045589
(0.107 mL, 0.59 mmol) was added and the reaction was stirred for 3-4 hours at 80
°C. Upon completion, the reaction was allowed to cool to room temperature and
then quenched by a pH 7.0 buffer (1.0 M K2HPO4 and 1.0 M NaH2PO4, 2 ml). The
mixture was extracted with CH2CI2 (3 x 10 mL). The combined organic layers were
washed with brine, dried with Na2S04, and concentrated in vacuo. The crude
product was purified by column chromatography (SiO2,0-10% MeOH-CHaCk to
afford 23 mg (13%) of 131 as a light yellow solid: 1H NMR (400 MHz, CDCI3)
8 9.85 (s, 1H), 8.03 (d, 7=8,2H), 7.54 (m, 1H), 7.417 (t,1 = 8,2H), 6.50 (s, 2H),
6.07 (d, J=4.8,1H), 5.73 (m, 1H), 5.37 (d, J=32,2H), 4.83 (m, 2H), 4.46 (m, 1H),
2.00 (s, 3H); [M+H]+ 459.3; Elemental analysis for C1H1N407S-0.7EtOAc: calc'd:
C, 52.65; H, 4.57; N, 10.77; S, 6.16; found: C, 53.59; H, 4.57; N, 10.83; S, 6.17.
Step 2: Preparation of(3'S)-5-Amino-3-(3'-deoxy-3'-methylide)te-fi-D-
ribofuranosyl)-3H,6H-thiazolo[4,5-d]pyrimidin-2,7-dione (132)
(3'S)-5-Amino-3-(3'-acetoxymethyl-2',5'-di-0-acetyl-3'-deoxy-p-D-
ribofuranosyl)-3J7-thiazolo[4,5-rf]pyrimidin-2-one 131 (113 mg, 0.25 mmol) was
dissolved in methanol (5 mL) at ambient temperature. Potassium carbonate (38 mg,
0.27 mmol) was added and the mixture was stirred at room temperature overnight.
Upon completion, acetic acid was added (34pL) and the mixture was stirred another
30 minutes at room temperature. The mixture was concentrated, purified by HPLC,
then triturated by EtOAc to afford 49 mg (64%) of 132 as a white solid: 1H NMR
(400 MHz, DMSO-d6) 8 11.563 (s, 1H), 6.82 (s, 2H), 5.80 (m, 1H), 5.62 (m, 1H),
5.16 (d, J=14.4,2H), 4.51 (m, 1H), 3.53 (m, 2H), 1.89 (s, 2H); [M+H]+ 313.07.
Example 43
Preparation of 5-Amino-3-(2',3',5'-tri-hydroxy-fi-D-xylofiiranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (134)

Step 1: Preparation of 5-Amino-3-(2',3 5'-tri-0-acetyl-p-D-xylofuranosyl)-
3H,6H-thiazolo[4,5-d]pyrimidin-2-one (133)
114

WO 2006/066080 PCT/US2005/045589



In a manner similar to Example 42, Step 1, using commercially available
tetra-O-acetylxylofumnose, 740 mg of the title compound 133 was generated in 20%
yield as a white solid: 1H NMR (400 MHz, 4-DMSO) 5 11.32 (s, 1H), 6.98 (br s,
2H), 6.09 (dd, = 8.6,2.3 Hz, 1H), 5.76 (d, /=5.5 Hz, 1H), 5.38 (dd, J = 8.6,2.3
Hz, 1H), 4.14 (m, 1H), 4.26 (m, 1H), 4.16 (m, 1H), 2.08 (s, 3H), 2.04 (s, 3H), 2.00
(s, 3H); [M+Hf 442.8; Elemental Analysis for Ci6Hi8N4O9S-1.0H2O: calc'd: C,
41.74; H, 4.38; N, 12.17; found: C, 41.92; H, 4.23; N, 11.71.
Step 2: Preparation of5-Amino-3-(2',3',5'-tri-hydroxy-b-D-xylofuranosyl)-3H, 6H~
thiazolo[4,5-d]pyrimidin-2-one (134)
In a manner similar to Example 42, Step 2,43 mg of the title compound
134 was generated in 67% yield as a white solid: 1H NMR (400 MHz, d6-
DMSO) 5 11.24 (br s, 1H), 6.86 (br s, 2H), 5.60 (d,=4.68 Hz, 1H), 5.57 (d, /
= 4.68 Hz, 1H), 5.04 (d, J = 7.8 Hz, 1H), 4.64 (m, 1H), 4.41 (m, 1H), 3.87 (m,
2H), 3.54 (m, 2H); [M+H]+ 316.9; Elemental Analysis for CnHi4N4O5S-1.3H2O:
calc'd: C, 35.35; H, 4.33; N, 16.49; found: C, 35.73; H, 4.21; N, 16.15.
Example 44
Preparation of 5-Amino-3-(3'-(R)-octyloxy ~{!-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (135)

The required sugar acetic acid l,2,5-tri-O-acetyl-3(S)-octyloxy-D-
xylofuranose or and b mixture (94) was prepared as reported in Example 31.
115

WO 2006/066080 PCT/US2005/045589
Step 1: Preparation of 5-Amino-3-(3'-(R)-octyloxy -b-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (135)
In a manner similar to Example 42, Step 1,18.8 mg of the title compound
135 was generated from acetic acid l,2,5-tri-0-acetyl-3(S)-octyloxy-D-xylofuranose
or and b mixture (94) in 2% yield as a white solid: *H NMR (400 MHz, J^-DMSO)
8 11.21 (s, 1H), 6.94 (br s, 2H), 6.12 (m, 1H), 5.74 (d, J= 62 Hz, 1H), 4.30 (m,
3H), 4.16 (m, 1H), 3.57 (m, 1H), 3.41 (m, 1H), 2.02 (d, /= 8.6 Hz, 6H), 1.50 (m,
2H), 1.26 (m, 10H), 0.86 (m, 3H); [M+H]+ 512.9; Elemental Analysis for
C22H32NAS: calc'd: C, 51.55; H, 6.29; N, 10.93; found: C, 51.47; H, 6.37; N,
10.77.
Example 45
Preparation of 5-Amino-3-(3 '-(R)-Methoxy b-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (137)

The required sugar, acetic acid l,2,5-tri-O-acetyl-3)-methoxy-D-
xylofaranose cc and J3 mixture (91) was prepared as reported in Example 30.
Step 1: Preparation of5-Amino~3-(3'-(R)-Methoxy-0-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (136)

In a manner similar to Example 42, Step 1,230 mg of the title compound
136 was generated from acetic acid l,2,5-tri-O-acetyl-3)-methoxy-D-xylofuranose
a and.b mixture (91) in 31% yield as a white solid: 1H NMR (400 MHz, d6-DMSO)
511.21 (s, 1H), 6.94 (br s, 2H), 6.14 (m, 1H), 5.74 (d, = 6.2 Hz, 1H), 4.33 (m,
2H), 4.18 (m, 2H), 3.35 (s, 3H), 3.30 (s, 3H), 2.04 (s, 3H), 2.01 (s, 3H); [M+H]+
116

WO 2006/066080 PCT/US2005/045589
414.8; Elemental Analysis for CHNAS: calc'd: C, 43.48; H, 4.38; N, 13.52;
found: C, 43.12; H, 4.36; N, 13.17.
Step 2: Preparation of5-Amino-3-(3'-(R)-Methoxy -b-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (137)
In a manner similar to Example 42, Step 2,43 mg of the title compound 137
was generated in 29% yield as a white solid: 'H NMR (400 MHz, 4-DMSO)
5 11.50 (br s, 1H), 6.98 (br s, 2H), 5.68 (d, / = 5.5 Hz, 1H), 5.60 (d, /= 7.8 Hz, 1H),
5.10 (m, 1H), 4.48 (m, 1H), 4.08 (m, 1H), 3.84 (m, 1H), 3.57 (m, 2H), 3.35 (s, 3H);
[M+H]+ 330,9; Elemental Analysis for C1!Hi4N4O6S-0.7H2O-0.1iPrOH: calc'd: C,
38.89; H, 4.68; N, 16.06; found: C, 38.78; H, 4.30; N, 15.84.
Example 46
Preparation of5-Amino-3-(3'-(R)-(2-methoxy-ethoxy), 2 ',5'-di-hydroxy-j3-D-
xylqfuranosyl)-3H, 6H~thiazolo[4,5-d]pyrimidin-2-one (139)

The required sugar, acetic acid l,2,5-tri-O-acetyl-3-(2-methoxy-ethoxy)-D-
xylofuranose a and p mixture (98) was prepared as reported in Example 32.
Step 1: Preparation of5-Amino-3'(3'-(R)-(2-methoxy-ethoxy), 2 ',5'-di-O-acetyl ~fi-
D-xylofuranosyl)-3H, 6H-thiazolo[4,5-dJpyrimidin-2-one (138)

In a manner similar to Example 42, Step 1,118 mg of the title compound
138 was generated from Acetic acid l,2,5-tri-O-acetyl-3-(2-methoxy-ethoxy)-D-
xylofuranose a and @ mixture (98) in 21% yield as a white solid: 1H NMR (400
MHz, 4-DMSO) S 11.23 (s, 1H), 6.90 (br s, 2H), 6.13 (m, 1H), 5.74 (d, = 6.24Hz,
1H), 4.33 (m, 3H), 4.17 (m, 1H), 3.73 (m, 1H), 3.57 (m, 1H), 3.46 (m, 2H), 3.25 (s,
117

WO 2006/066080 PCT/US2005/045589
3H), 2.03 (s, 3H), 2.01 (s, 3H); [M+H]+ 459.3; Elemental Analysis for
CH22N4O9S-0.3H2O.0.5EtOAc: calc'd: C, 44.93; H, 5.28; N, 11.03; found: C,
44.93; H, 5.01; N, 11.14.
Step2: Preparation of5-Amino-3-(3'-(R)-(2-methoxy-eihoxy), 2',5'-di-hydroxy-fi-
D-xylofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2-one (139)
In a manner similar to Example 42, Step 2,43 mg of the title compound 139
was generated in 36% yield as a white solid: 1H NMR (400 MHz, d6-DMSO)
811.44 (br s, 1H), 6.97 (br s, 2H), 5.67 (d, 7= 5.46 Hz, 1H), 5.60 (d, J = 7.8 Hz,
1H), 5.10 (m, 1H), 4.39 (m, 1H), 4.08 (m, 1H), 3.98 (m, 1H), 3.71 (m, 1H), 3.59 (m,
3H), 3.45 (m, 2H), 3.27 (s, 3H); [M+H]+ 374.9; Elemental Analysis for
Ci3Hi8N4O7S-1.0H2O0.25Et0Ac: calc'd: C, 40.57; H, 5.35; N, 13.52; found: C,
40.81; H, 4.96; N, 13.40.
Example 47
Preparation of 5-Amino-3-(3 '-(S)-methyl, 2',3',5 '-tri-hydroxy-fi-D-
xylofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrirnidin-2-one (141)

The preparation of the required sugar, 1,2,3,5 tetra-O-acetyl-3(S)-methyl D-
xylofuranose a and p mixture (105), was prepared as reported in Example 34.
Step h Preparation of 5-Amino-3-(3f-(S)-methyl, T,3',5'-tri-O-acetyl-p-D-
xylofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2-one (140)

In a manner similar to Example 42, Step 1,110 mg of the title compound
140 was generated from 1,2,3,5 tetra-O-acetyI-3(S)-methyl D-xylofuranose
aand b mixture (105) in 15% yield as a white solid: 1H NMR (400 MHz, de-
118

WO 2006/066080 PCT/US2005/045589
DMSO) 5 11.27 (br s, 1H), 6.98 (br s, 2H), 6.25 (d, = 4.68 Hz, 1H), 5.77 (d, =
4.68 Hz, 1H), 4.40 (dd, J = 9.4, 3.1 Hz, 1H), 4.22 (m, 1H), 3.68 (dd, J= 4.7, 3.1 Hz,
1H), 2.08 (s, 3H), 2.04 (s, 3H), 2.02 (s, 3H), 1.56 (s, 3H); [M+H]+ 456.8; Elemental
Analysis for CHN-O.SHaO-OPrOH: calc'd: C, 44.27; H, 4.77; N, 11.73;
found: C, 44.45; H, 4.55; N, 11.62.
Step 2:' Preparation of 5-Amino-3-(3'-(S)-methyl, 2',3',5'-tri-hydroxy-b-D-
xylofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2-one (141)
In a manner similar to Example 42, Step 2,31 mg of the title compound 141
was generated in 54% yield as a white solid: 1H NMR (400 MHz,-DMSO)
511.36 (br s, 1H), 6.94 (br s, 2H), 5.70 (d = 5.46 Hz, 1H), 5.58 (d,=4.7 Hz,
1H), 5.09 (br s, 1H), 4.48 (m, 2H), 3.59 (m, 3H), 1.17 (s, 3H); [M+H]+ 330.9;
Elemental Analysis for CHOeS-LlHaO: calc'd: C, 37.73; H, 4.66; N, 16.00;
found: C, 37.66; H, 4.22; N, 15.60.
Example 48
Preparation of 5-Amino-3-(5'-( 1,2-diacetoxy-ethyl), 2',3'-di-hydroxy-j3-
D-glucofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2-one (143)

The required sugar, penta-O-acetylglucofuranose (108), was prepared as
described in Example 35.
Step 1: Preparation of'5-Amino-3-(5'-(1,2-diacetoxy-ethyl), 2',3'-di-O-acetyl-p-D-
glucofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2-one (142)

In a manner similar to Example 42, Step 1,100 mg of the title compound
142 was generated from penta-O-acetylglucofuranose (108) in 10% yield as a white
119

WO 2006/066080 PCT/US2005/045589
solid: 1H NMR (400 MHz, CDC13) 5 5.91 (m, 1H), 5.81 (br s, 2H), 5.73 (d, J = 6.2
Hz, 1H), 5.51 (m, 1H), 5.41 (m, 1H), 4.55 (dd, / = 12.5,2.3 Hz, 1H), 4.29 (t, 7 =
7.02 Hz, 1H), 3.90 (m, 1H), 1.96 (s, 3H), 1.93 (s, 3H), 1.90 (s, 3H), 1.55 (br s, 1H);
[M+H]+ 515.3; Elemental Analysis for C19N11S-O.l5MeOH: calc'd: C, 44.29;
H, 4.39; N, 10.79; found: C, 44.69; H, 4.44; N, 10.41.
Step 2: Preparation of 5~Amino-3-(5'-(l,2-diacetoxy-ethyl), 2',3'-di-hydroxy-b-D-
glucofuranosyl)-3H, 6H-thiazolo[4,5-dJpyrimidin-2-one (143)
In a manner similar to Example 42, Step 2,45 mg of the title compound 143
was generated in 83% yield as a white solid: lH NMR (400 MHz, aVDMSO)
5 11.34 (br s, 111), 6.96 (br s, 2H), 5.72 (d, =4.7 Hz, 1H), 5.63 (d, = 3.12 Hz,
1H), 5.11 (d, /= 9.4 Hz, 1H), 4.56 (m, 2H), 4.39 (t,= 5.46 Hz, 1H), 3,96 (m, 1H),
3.74 (m, 2H), 3.51 (m, 1H), 3.36 (m, 1H); [M+H]+ 346.9; Elemental Analysis for
C11H14N4O7S4.OH2O: calc'd: C, 36.26; H, 4.43; N, 15.38; found: C, 36.20; H, 4.37;
N, 15.01.
Example 49
Preparation of 5-Amino-3-(3'-(S)-acetoxymethyl-, 2'.,3',5'-tri-O-acetyl-p-D-
xylofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2~one (144)

AXVO
144
The required sugar, tetra-O-acetyl-3-acetoxymethyl-D-xylofuranos
a and (3 mixture (113) was prepared as reported in Example 36.
Step 1: Preparation of5-Amino-3-(3'-(S)-acetoxymetltyl-, 2',3',5'-tri-O-acetyl-p-D-
xylofuranosyl)-3H,6H-thiazolo[4,5-d]pyrimidin-2-one (144)
In a manner similar to Example 42, Step 1,80 mg of the title compound 144
was generated from tetra~0-acetyl-3-acetoxymethyl-D-xylofuranos a and /?mixture
(113) in 13% yield as a white solid: JH NMR (400 MHz, d6-DMSO) 811.23 (s,
1H), 6.93 (br s, 2H), 6.37 (d, / = 5.46 Hz, 1H), 5.69 (d, J = 5.46 Hz, 1H), 4.77 (d, J
= 11.7 Hz, 1H), 4.52 (m, 3H), 4.39 (dd, 7 = 7.8,1.6 Hz, 1H), 4.17 (dd /=7.8,3.9
Hz, 1H), 2.08 (s, 3H), 2.06 (s, 3H), 2.05 (s, 3H), 2.03 (s, 3H); [M+H]+ 514.8;
120

ft.
WO 2006/066080 PCT/US2005/045589
Elemental Analysis for C19H22N4O11S: calc'd: C, 44.36; H, 4.31; N, 10.89; found: C,
44.16; H, 4.37; N, 10.69.
Example 50
Preparation of 5-Amino-3-(3'-(R)-Butoxy-b-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5-d]pyrimidin-2-one (146)

The required sugar acetic acid l,2,5-tri-0-acetyl-3-butoxy-D-xylofuranose
a and {3 mixture (101) was prepared as reported in Example 33.
Step 1: Preparation of5-Amino-3-(3'-(R)-Butox-2',5'-di-O-acetyl-P~D-
xytofuranosyl)-3H, 6H-ihiazolo[4,5-d]pyrimidin-2-one (145)

In a manner similar to Example 42, Step 1, the title compound 145 was
generated from acetic acid l,2,5-tri-0-acetyl-3-butoxy-D-xylofuranose
a and b mixture (101) and carried on crude to Step 2.
Step 2: Preparation of 5-Amino-3-(3'-(R)-Butoxy-b-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5'd]pyrimidin-2-one (146)
In a manner similar to Example 42, Step 2, 5.3 mg of the title compound 146
was generated in 16% yield as a white solid: :H NMR (400 MHz, ds-DMSO)
5 11.22 (br s, 1H), 6.92 (br s, 2H), 5.64 (d, 7= 5.46 Hz, 1H), 5.60 (d, =7.8 Hz,
1H), 5.08 (m, 1H), 4.39 (t, /= 6.24 Hz, 1H), 4.07 (m, 1H), 3.92 (t, =7.02 Hz, 1H),
3.57 (m, 3H), 3.42 (m, 1H), 1.48 (m, 2H), 1.33 (m, 2H), 0.88 (t, = 7.02 Hz, 3H);
[M+H]+ 372.9; Elemental Analysis for CHaoNS-LOHaO-OMeOH: calc'd: C,
42.89; H, 5.90; N, 13.89; found: C, 43.18; H, 5.68; N, 13.65.
121

WO 2006/066080 PCT/US2005/045589

Step 1: Preparation of 5-Amino-3-(5'-0-acetoxymethyl-2',3'-di-0-acetyl-5'-deoxy-
b-D-ribofuranosyl)-3H, 6H~thiazolo[4,5-d]pyrimidin-2,7-dione (149)

In a manner similar to Example 42, Step 1,96 mg of the title compound
149 was generated from 5-O-acetoxymethyl-l, 2,3-tri-O-acetyl-5-deoxy-a,p-D-
ribofuranose [prepared according to the method of Pakulski et al. Polish J.
Chem. 1995, 69,912-917] in 34% yield as a white solid: 'H NMR (400 MHz,
CDCk) 5 11.92 (s, 1H), 6.15 (d, =6.4,1H), 5.94 (s, 2H), 5.71 (m, 1H), 4.91
(m, 1H), 4.40 (m 1H), 4.16 (m, 2H), 2.09 (s, 9H), 2,00 (m, 2H); [M+H]+ 457.0;
Elemental Analysis for (CENS-OSHbO): calc'd: C, 44.30; H, 4.48; N,
12.16; S, 6.96; found: C, 44.79; H, 4.62; N, 11.55; S, 6.59.
Step 2: Preparation of 5-Amino-3-(5'-deoxy-5'-hydroxymethyl-fi-D-
ribofiiranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2,7-dione (150)
In a manner similar to Example 42, Step 2,28 mg of the title compound
150 was generated in 56% yield as a white solid: lU NMR (400 MHz, dg-
DMSO) 511.23 (s, 1H), 6.94 (s, 2H), 5.74 (m, 1H), 5.22 (m, 1H), 4.87 (m, 1H),
4.40 (m, 1H), 4.00 (m, 2H), 3.44 (s, 3H), 1.72 (m, 2H); [M+H]+330.9.
Example 53
Preparation of 5-Amino-3-[3'-deoxy-3'-O-p-toluenesulfonyl-fi-D-
xylofuranosyl]-3H,6H-tliiazolo-[4,5-d]pyrimidine-2,7-dione (151)
123

WO 2006/066080 PCT/US2005/045589

Step 1: Preparation of5-Amino-3-[2'5'-di-0-acetyl-3'-deoxy-3'-0-p-
toluenesulfonyl-p-D-xylofuranosyl]-3H,6H'thiazolo-l4,5-d]pynmidine-2,7-dione
(151)
In a manner similar to Example 42, Step 1,24.6 mg of the title compound
151 was generated in 12% yield as an off-white solid: *H NMR (400 MHz, CDCl3)
5 11.89 (s, 1H), 7.84 (d, = 8.4,2H), 7.40 (d, 7= 8.4,2H), 6.22 (d, J= 4.4,1H),
5.92 (br s, 2H), 5.75 (d, = 4.8,1H), 4.95 (d, =4.8,1H), 4.30 (m, 1H), 4.25 (d, =
6,2H), 2.48 (s, 3H), 2.05 (s, 6H); [M+H]+ 555.3.
Example 54
Preparation of (3 'R)-5-Amino-3-(3 '-deoxy-3 '-fluoro-J3-D-xylofuranosyl)-3H, 6H-
thiazolo[4,5'd]pyrimidin~2,7-dione (153)
Step 1: Preparation of(3'R)-5-Amino-3-(2'-0-acetyl-5'-0-benzoyl-3'~deoxy-3'-
fluoro-fi-D-xylofuranosyl)-3H, 6H-thiazolo[4,5-d]pyrimidin-2,7-dione (152)

In a manner similar to Example 42, Step 1,149 mg of the title compound
152 was generated from 1,2-Di-0-acetyl-5-0-benzoyl-3-deoxy-3-(R)-fluoro-a,p-D-
xylofurariose [prepared according to the method of Gosselin et al. Carbohydrate
Research 1993,249,1-17] in 24% yield as a yellow solid: !H NMR (400 MHz,
124

WO 2006/066080 PCT/US2005/045589
CDCl3) 5 11.57 (s, 1H), 8.04 (d, / = 6.8, 2H), 7.56 (t, J = 7.6,1H), 7.43 (t, J = 7.6,
2H), 6.35 (dd, J = 22.4,4.8,1H), 5.92 (s, 2H), 5.32 (dd, /= 51.6, 4.8,1H), 5.20 (s,
1H), 4.79 (dd, J = 11.2,4,1H), 4.59 (m, 2H), 4.5 (m, 1H), 2.06 (s, 3H); [M+H]+
465.3.
Step 2: (3'R)-5-amino-3-(3'-deoxy-3 '-fluoro-P-D-xylofumnosyl)-3H,6H-
thiazolo[4,5-d]pyrimidin-2,7-dione (153)
In a manner similar to Example 42, Step 2,14.3 mg of the title compound
153 was generated in 45 % yield as white solid: *H NMR (400 MHz, DMSO-dg)
8 11.41 (s, 1H), 6.97 (s, 2H), 5.77 (m, 1H), 5.19 (rn, 1H), 4.98 (m, 1H), 4.01 (m,
1H), 3.60 (m, 2H), 2.09 (s, 2H); [M+H]+ 318.9; Elemental analysis for
(CHFN4OsS-0.4EA-2H20): calc'd: C, 35.76; H, 4.71; N, 14.3; found: C, 35.71;
H, 3.68; N, 14.15.
Example 55
Preparation of 3-Allyl-5-amino-3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-dione
(154)

Step 1: Preparation of 3-Allyl-5-amino-3H, 6H-thiazoh[4,5-d]pyrimidine-2,7-dione
(154)
In a manner similar to Example 15, Step 1, schemel, 178 mg of the title
compound 154 was generated in 35 % yield as a pale yellow solid: 'H NMR (400
MHz, DMSO-rf(j) 810.26 (s, 1H), 6.09 (s, 2H), 5.06-5.01 (m, 1H), 4.32 (dd, / =
10.3,1.5,1H), 4.196 (dd, /=16.9,1.5,1H), 3.55 (d, J~4.4,2H); [M+H]+ 225.1.
Example 56
Preparation of 5-Amino-3-pyridin-3-ylmethyl-3H,6E-thiazolo[4,5-d]pynmidine-
2,7-dione (155)
125

WO 2006/066080 PCT/US2005/045589
Step 1; Preparation of5-Amino-3-pyridin-3-ylmethyl-3H,6H-thiazolo[4,5-
d]pyrimidine-2,7-dione (155)
In a manner similar to Example 15, Step 1,143 mg of the title compound
155 was generated in 24% yield as a white solid: 'H NMR (400 MHz, TMSO-d6)
510.33 (s, 1H), 7.76 (d, .= 2.2,1H), 7.68 (dd, = 2.2,1.5,1H), 6.88 (m, 1H), 6.55
(s, 2H), 6.15 (s, 2H), 4.17 (s, 2H); [M+Hf 276.1.
Example 57
Preparation of5-Amino-3-(4-chloro-but-2-enyl)-3H, 6H-thiazblo[4,5-
d]pyrimidine-2,7-dione (156)

Step 1: Preparation of5-Amino-3-(4-chloro-but-2-enyl)-3H,6H-ihiazolo[4,5-
d]pynmidine-2,7-dione (156)
In a manner similar to Example 15, Step 1,440 mg of the title compound
156 was generated in a 63 % yield as a light yellow solid: 'H NMR (400 MHz,
DMSO-4J) 5 10.33 (s, 1H), 6.90 (s, 2H), 5.82-5.74 (m, 1H), 5.65-5.59 (m, 1H), 4.45 .
(d, /=7.0,2H), 4.39 (d, / =7.8,2H); [M+Hf 273.1.
Example 58
Preparation of 5-Amino-3-hexyl-3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-dione (157)

Step 1: Preparation of5-Amino-3-hexyl-3H,6H~thiazolo[4,5-d]pyrimidine-2,7-
dione (157)
In a manner similar to Example 15, Step 1,154 mg of the title compound
157 was generated in a 35 % yield as an off-white solid: !H NMR (400 MHz,
126

WO 2006/066080 PCT/US2005/045589
DMSO-4) 5 11.03 (s, 1H), 6.88 (s, 2H), 3.74 (t, = 6.8,2H), 1.61 (m, 4H), 1.26 (m,
4H), 0.85 (t, = 6.8,3H); [M+H]+ 269.31.

Example 59
Preparation of(l %2 'S,3%4'R)-5~Amino-3-(2 ',3'-dio\y-4'-hydroxymethyl-
cyclopentan-1 '-yl)-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione (158)
(rR,2'S,3'R,4'R)N7-[7-chloro-2-oxo-3-(2',3'-0-isopropylidene-4'-vinyl-
cyclopentan4'-yl)-23-dihydro-thiozolo[4,5-iflpyriniidin-5-yl]-N,N-dimethyl-
formamidine (85) (85 mg, 0.20 mmol) was dissolved in CH3OH (3.0 mL) and H2O
(1.5 mL) at ambient temperature. The solution was cooled to 0 °C. Sodium periodate
(90 mg, 0.42 mmol) and Osmium tetroxide (2 mg, catalytic) were then added to the
solution. The reaction was stirred at 0 °C for 1 h and room temperature for 2 h
before it was filtered and concentrated. The resulting mixture was dissolved in
CH2CI2 (10 mL), then washed with H2O (2 x 10 mL). The organic layer was dried
over MgSO4, filtered, then concentrated.
The above product was dissolved in CH3OH (3 mL) at ambient temperature.
Sodium borohydride (12 mg, 0.32 mmol) was then added to the solution. The
reaction was stirred for 1 h, then concentrated. The resulting mixture was dissolved
in CH2CI2, and washed with H2O (2 x 10 mL). The organic layer was dried over
MgSO4, filtered, then concentrated.
The above product was dissolved CH3OH (1 mL) and 2 M HCl (5 rnL) at
ambient temperature. The reaction was stirred at reflux for 5 h, then concentrated.
The resulting mixture was purified by reverse phase HPLC affording 9.1 mg (13%)
of 158 as a white solid: 1H (400 MHz, CD3OD) 8 4.95(m, IE), 4.69 (dd, = 7.2, 5.6,
127

WO 2006/066080 PCT/US2005/045589
1H), 4.03 (t, = 5.2,1H), 3.71 (dd, J = 11.2,6.4,1H), 3.60 (dd, J = 11.2,6.4,1H),
3.35 (s,lH), 1.93-2.14 (m,3H).
Example 60
5-Amino-3-(2 '-O-acetyl-3 '-deoxy-j3-D-ribofuranosyl)-3H, 6H-thiazolo[4,5-
d]pyrimidine-2,7-dione (160)

a. Candida Arctica, acetone, pH 7 phosphate buffer, 97%.
Preparation of 5-Amino-3-(2'-0-acetyl-3'-deoxy-b-D-ribofuranosyl)-3H,6H-
thiazolo[4,5-dJpyrimidine-2,7-dwne (160)
To a suspension of nucleoside diacetate 121 (200.0 mg, 0.52 mmol) in
acetone (5.0 mL), pH 7 phosphate buffer (2.5 mL), and H2O (22.5 mL) was added
Candida Arctica immobilized acrylic resin (0.10 g). The mixture decolorized within
5 mia after enzyme addition. The reaction was stirred at room temperature for 16 h.
Celite (1.0 g) was added and, after stirring for 10 min, the mixture was filtered
through a pad of celite. The filter cake was rinsed with acetone (3 x 10 mL), and the
acetone was reduced in vacuo. The remaining aqeous layer was then heavily salted
with solid NaCl. Ethyl acetate was added the biphasic mixture was vigorously stirred
for 30 min before separating. The organic layer was dried over NaaSO4,, decanted,
concentrated, and purified via flash chromatography (SiC2,50-100% EtOAc-
hexanes + 2% MeOH). This afforded 155.0 mg of monoacetate 160 (97%): lH
(400MHz, DMSO-rftf) 8 11.20 (s, 1H), 6.94 (br s, 2H), 5.79 (d, = 2.4,1H), 5.63 (d,
=8.0,1H), 4.76 (t, 7=6.0,1H),4.11 (dt, 7 = 5.2,10.4,1H), 3.43-3.52 (m, 2H),
2.44-2.5 (m, 1H), 2.05 (s, 3H), 1.95 (dd, = 6.0,13.6,1H); [M+H]+ m/z 342.
Analysis cal'd for: C12Hi4N4O6S "0.5 H2O: C, 42.10; H,4.12; N, 16.37; S, 9.37.
Found: C, 42.30; H, 4.26; N, 16.37; S, 9.23.
Example 61
5-Amino-3-(2,'3'-dideoxy-p-D-ribofuranosyl)-3H,6H-thiazolo[4,5-d]pyrimidine-
2,7-dione (165)
128

WO 2006/066080 PCT/US2005/045589



Step 1) Preparation of 5-N-Acetylamino-3-(5'-tert-butyldimethylsily-2'-deoxy-2'-O-
thiocarbonylimidazole-b-D-ribofuranosyl)-3H,6H-ihiazolo[4,5-d]pyrimidine-2,7- .
dione (161)

To a mixture of alcohols 127 and 128 (247 mg, 0.541 mmol) in MeCN (8
niL) at rt was added TCDI (193 mg, 1.08 mmol). The reaction mixture was stirred
18 h whereupon it was concentrated and submitted to flash chromatography (SiO2,
EtOAc), affording 150 mg (49%) of a mixture of thiocarbamates 161 and 162 as a
solid material: lH NMR (400 MHz, rf6-DMSO) 8 12.18 (br s, 1H), 11.72 (br s, 1H),
8.54 (s, 1H), 7.86 (s, 1H), 7.10 (s, 1H), 6.36 (m, 1H), 6.04 (s, 1H), 4.38-4.43 (m,
1H), 3.68-3.80 (m, 2H), 2.63-2.69 (br m, 1H), 2.36-2.41 (m, 1H), 2.17 (s, 3H), 0.84
(s, 9H), 0.00 (s, 6H); [M+H]+ m/z 567.
Step 2) Preparation of 5-N-Acetylamino-3-(5'-tert-butyldimethylsily-2',3'-dideoxy-
3-D-ribofuranosyl)-3H,6H-thiazolo[4,5-d]pyrimidine-2,7-dione (163)

129

WO 2006/066080 PCT/US2005/045589
To a mixture of thiocarbamates 161 and 162 (57 mg, 0.10 mmol) and
Bu3SnH (187 uL, 0.705 mmol) in PhMe (10 mL) at rt was added AIBN (1.6 mg,
0.010 mmol). The reaction mixture was immersed into a 130° C oil bath, stirred 20
min, concentrated and then submitted to flash chromatography (S1O2,60-80%
EtO Ac-CHCLO, affording 28 mg (64%) of compound 163 as a white solid: lH NMR
(400 MHz, 4$-DMSO) 812.13 (br s, 1H), 11.80 (br s, 1H), 6.11 (dd, / = 8.4,3.7,
1H), 3.95-4.02 (m, 1H), 3.64 (d, = 5.1,2H), 2.54-2.59 (m, 1H), 2.23-2.33 (m, 1H),
2.19 (s, 3H), 2.04-2.13 (m, 1H), 1.94-1.97 (m, 1H), 0.82 (s, 9H), -0.01 (6H); [M+H]+
m/z 441.
Step 3)Preparation of 5~N-Acetylamino-3-(2',3' -dideoxy-j3-D-ribofuranosyl)-
3H, 6H-thiazolo[4,5-d]pyrimidine-2,7-dione (164)

A solution of siloxane 163 (84 mg, 0.19 mmol) in 2 M HF-MeCN (20 mL)
was stirred for 10 min, then concentrated and submitted to flash chromatography
(SiO2, 5-10% MeOH-CHCl3) to afford 10 mg (16%) of alcohol 164 as a white solid:
*H NMR (400 MHz, -DMSO) 812.15 (br s, 1H), 11.78 (br s, 1H), 6,10 (dd, =
8.4,4.0,1H), 4.66 (t, = 5.9,1H), 3.91-3.97 (m, 1H), 3.46 (t, /= 5.9,2H), 2.51-2.58
(m, 1H), 2.21-2.32 (m, 1H), 2.19 (s, 3H), 2.01-2.18 (m, 1H), 1.89-1.97 (m, 1H);
[M+HT m/z 327.
Step 4) Preparation of 5-Amino-3-(2',3'-dideoxy-j3-D-ribofuranosyl)-3H,6H~
thiazolo[4,5-d]pyrimidine-2,7-dione (165)

To a solution of acetamide 164 (19 mg, 0.058 mmol) in MeOH (3 mL) at rt
was added K2CO3 (64 mg, 0.46 mmol). The reaction mixture was stirred 18 h
whereupon it was quenched with HO Ac (53 uL), concentrated and triturated with
130

WO 2006/066080 PCT/US2005/045589
MeOH-H2O to afford 6 mg (36%) of the title compound 165 as a white solid: !H
NMR (400 MHz, 4-DMSO) 811.15 (s, 1H), 6.85 (br s, 2H), 6.07 (dd, /= 8.4,4.4,
1H), 4.63 (t, /= 5.9,1H), 3.89-3.95 (m, 1H), 3.46 (t, J = 5.5,2H), 2.44-2.48 (m,
1H), 2.17-2.27 (m, 1H), 2.01-2.11 (m, 1H), 1.86-1.93 (m, 1H); [M+Hf m/z 285.
Anti-Viral Activity of Compounds
A number of assays may be employed in accordance with the present
invention in order to determine the degree of anti-viral activity of a compound of the
invention such as cell culture, animal models, and administration to human subjects.
The assays described herein may be used to assay viral growth over time to
determine the growth characteristics of a virus in the presence of a compound of the
invention.
In another embodiment, a virus and a compound of the invention are
administered to animal subjects susceptible to infection with the virus. The
incidence, severity, length, virus load, mortality rate of infection, etc. can be
compared to the incidence, severity, length, virus load, mortality rate of infection,
etc. observed when subjects are administered the virus alone (in the absence of a
compound of the invention). Anti-virus activity of the compound of the invention is
demonstrated by a decrease in incidence, severity, length, virus load, mortality rate
of infection, etc. in the presence of the compound of the invention. In a specific
embodiment, the virus and the compound of the invention are administered to the
animal subject at the same time, hi another specific embodiment, the virus is
administered to the animal subject before the compound of the invention. In another
specific embodiment, the compound of the invention is administered to the animal
subject before the virus.
In another embodiment, the growth rate of the virus can be tested by
sampling biological fluids/clinical samples (e.g.t nasal aspirate, throat swab, sputum,
broncho-alveolar lavage, urine, saliva, blood, or serum) from human or animal
subjects at multiple time points post-infection either in the presence or absence of a
compound of the invention and measuring levels of virus. In specific embodiments,
the growth rate of a virus is assayed by assessing the presence of virus in a sample
after growth in cell culture, growth on a permissible growth medium, or growth in
subject using any method well-known in the art, for example, but not limited to,
immunoassay (e.g., ELISA; for discussion regarding ELISAs see, e.g., Ausubel et
131

WO 2006/066080 PCT/US2005/045589
al, eds, 1994, Current Protocols in Molecular Biology, Vol. I, John Wiley & Sons,
Inc., New York at 11.2.1), immunofiuorescent staining, or immunoblot analysis
using an antibody which immunospecifically recognizes the virus to be assayed or
detection of a virus-specific nucleic acid (e.g., by Southern blot or RT-PCR analysis,
etc.).
In a specific embodiment, viral titers can be determined by obtaining
biological fluids/clinical samples from infected cells or an infected subject,
preparing a serial dilution of the sample and infecting a monolayer of cells that are
susceptible to infection with the virus (e.g. primary cells, transformed cell lines,
patient tissue samples, etc) at a dilution of the virus that allows for the emergence of
single plaques. The plaques can then be counted and the viral liter expressed as
plaque forming units per milliliter of sample.
In one specific embodiment, the growth rate of a virus in a subject can be
estimated by the titer of antibodies against the virus in the subject. Antibody serum
liter can be determined by any method well-known in the art, for example, but not
limited to, the amount of antibody or antibody fragment in serum samples can be
quantitated by, e.g., ELISA. Additionally, in vivo activity of a Formula I compound
can be determined by directly administering the compound to a test animal,
collecting biological fluids (e.g., nasal aspirate, throat swab, sputum, broncho-
alveolar lavage, urine, saliva, blood, or serum) and testing the fluid for anti-virus
activity.
In embodiments where samples to be assayed for virus levels are biological
fluids/clinical samples (e.g.t nasal aspirate, throat swab, sputum, broncho-alveolar
lavage, urine, saliva, blood, or serum), the samples may or may not contain in tact
cells. Samples from subjects containing intact cells can be directly processed,
whereas isolates without intact cells may or may not be first cultured on a
permissive cell line (e.g. primary cells, transformed cell lines, patient tissue samples,
etc) or growth medium (e.g., LB broth/agar, YT broth/agar, blood agar, etc.). Cell
suspensions can be cleared by centrifugation at, e.g., 300xg for 5 minutes at room
temperature, followed by a PBS, pH 7.4 (Ca++ and Mg++ free) wash under the same
conditions. Cell pellets can be resuspended in a small volume of PBS for analysis.
Primary clinical isolates containing intact cells can be mixed with PBS and
centrifuged at 300xg for 5 minutes at room temperature. Mucus is removed from the
interface with a sterile pipette tip and cell pellets can be washed once more with PBS
132

WO 2006/066080 PCT/US2005/045589
under the same conditions. Pellets can then be resuspended in a small volume of
PBS for analysis.
In another embodiment, a compound of the invention is administered to a
human subject infected with a virus. The incidence, severity, length, viral load,
mortality rate of infection, etc. can be compared to the incidence, severity, length,
viral, load, mortality rate of infection, etc. observed in human subjects infected with
a virus in me absence of a compound of the invention or in the presence of a
placebo. Anti-viral activity of the compound of the invention is demonstrated by a
decrease in incidence, severity, length, viral load, mortality rate of infection, etc, in
the presence of the compound of the invention. Any method known in the art can be
used to determine anti-viral activity in a subject such as those described previously.
Additionally, in vivo activity of a Formula I prodrug can be determined by
directly administering the compound to an animal or human subject, collecting
biological fluids/clinical samples (e.g., nasal aspirate, throat swab, sputum, broncho-
alveolar lavage, urine, saliva, blood, or serum) and testing the biological
fluids/clinical samples for anti-viral activity (e.g., by addition to cells in culture in
the presence of the virus).
Metabolism of Formula I Prodrugs
The Formula I prodrugs of the present invention, must be metabolized to
Formula II compounds and other compounds of the invention in the body if they are
to serve as effective prodrugs. Hepatocyes often are used to assess the degree to
which a compound may be transformed in the body of an animal, and it is known
that such transformations may vary with hepatocytes from different species in a way
that reflects metabolism in the whole animal. See Seddon T. et ah, Biochem
Pharmacol, 38(10), 1657-65 (1989).
A study was undertaken to evaluate the metabolic stability of Formula I
compounds 14,15,13,114,30,103,67,65, and 76 in the presence of fresh
cynomolgus monkey hepatocytes and monitor the formation of 6-oxy metabolites,
i.e., Formula II compounds and other compounds of the invention. For comparison,
the metabolic stability of famciclovir was also assessed.
Preparation of Fresh Hepatocyte Suspension
Fresh cynomolgus monkey hepatocyte suspension (Lot #: Cyl41) was
purchased from CellzDirect (Tucson, AZ). Hepatocyte Incubation Medium (serum-
free, sterile) was purchased from In Vitro Technologies (Baltimore, MD).
133

WO 2006/066080 PCT/US2005/045589
The cynomolgus monkey hepatocyte suspension was prepared from fresh
cynomolgus monkey hepatocytes in hepatocyte incubation medium at the
concentration of 1.25 million cells/mL. The final incubation concentration (after test
article addition) was 1.0 million cells/mL.
Preparation of Stock Solutions
Existing 100 mM stock solutions in DMSO were used. The concentrations
of the test articles were checked using UV-vis microplate reader. Correction
coefficients were determined using the absorbance of a freshly prepared DMSO
stock of 122.
Incubations
Reaction suspensions were prepared in removable 96-well tubes, each
containing 320 uL of fresh cynomolgus monkey hepatocyte suspension at the
density of 1.25 million cells per mL and 40 uL of hepatocyte incubation medium.
The above mixtures were pre-incubated open at 37 °C, 95% humidity and 5% CO2
for 30 minutes. Reactions were initiated by the addition of 40 uL of test article at
lOx concentration to each tube to achieve the final concentrations of 50 |xM for the
test article(s) and 1 million/mL cell density. The reaction suspension in each tube
was mixed by inverting the tube several times. Aliquots of 50 uL from each
reaction suspension were distributed into six additional removable 96-well tubes
(one tube per time point taken at 15,30,45,60,90, and 120 minutes). The open
tubes were incubated at 37 °C under 95% humidity and 5% CO2.
Preparation of Samples for Analysis
At predetermined time points, reactions were terminated by the addition of
150 L of the stop solution to each tube containing 50 uL of the reaction
suspension. The composition of the stop solution was the following: 15 mL of
acetonitrile (containing 1 g/mLnebularine as an internal standard and 0.1% formic
acid) combined with 1 mL water.
The calibration curves were prepared in the following way. To 80 uL of cell
suspension (at the cell density of 1.25 million/mL) 10 uL of hepatocyte incubation
medium and 10 uL of the appropriate concentration of the compound in hepatocyte
incubation medium were added. Immediately following the compound addition, 300
uL of the stop solution (see above) was added.
134

WO 2006/066080 PCT/US2005/045589
All quenched samples were kept on wet ice until they were processed for
analysis. Then they were mixed using a bench top Multi-Tube Vortexer (VWR
Scientific Products) for approximately 30 seconds, and centrifuged at 4,000 rpm
(3,220 rcf) for 10 minutes at 4°C. Clear supernatant (100 JJL) was transferred into a
clean deep well 96-well plate, evaporated to dryness under nitrogen, reconstituted in
100 pL of 90:10 water: acetonitrile, and analyzed for the parent form and metabolites
of the test article using an appropriate LC/MS/MS method.
Bioanalvsis
The compounds were quantified on an AP13000 LC/MS/MS instrument in
the ESI-Positive MRM (multiple reaction monitoring) mode. The summary of the
results on Formula I prodrug degradation and product generation is given in Table 1.
Table 1
Concentration of the Metabolized Product Formed in Cvnomolgus Monkey
Hepatocvtes after 2 hrs Incubation of 50 HIM of a Formula I Prodrug:

Formula ICompound MetabolizedProduct Product Concentration (jaM) Response
14 122 9.7 +
15 122 31.8 ++++
13 122 24.2 +++
114 134 21.7 +++
30 117 21.5 +++
103 141 13.1 ++
57 157 1.7 +
65 148 14.7 ++
76 132 21.4 +++
Famciclovir Penciclovir '5.9 +
In fresh cynomolgus monkey hepatocytes compounds 14,15,13,114,30,
103,67,65, and 76 as well as famciclovir are metabolized to yield the corresponding
6-oxy metabolites: 122 from the first three Formula Iprodrugs and 134,117,141,
157,148, and 132, respectively. Famciclovir produces penciclovir.
135

WO 2006/066080 PCT/US2005/045589
IFN-a Induction from Peripheral Blood Mononuclear Cells (PBMC)
Peripheral blood mononuclear cells (PBMCs) are prepared by standard
methods from human blood and are primarily comprised of monocytes, NK cells,
circulating dendritic cells and both T and B cells. Briefly, they are purified by
density gradient centrifugation from a buffy coat, which is the component of whole
blood that contains leukocytes and platelets. In turn, buffy coats are prepared by
centrifuging whole blood and isolating the thin cream colored layer between the
upper plasma layer and the lower red blood cell portion of the separated mixture.
PBMC Purification
Freshly collected donor buffy coats were obtained from the San Diego
Blood Bank. PBMCs were isolated from the buffy coats using histopaque-1077
gradient (Sigma), essentially as described in the manufacturer's protocol. Buffy
coats were transferred into 50 ml centrifuge tubes and PBS added to a total
volume of 35 ml. Next 10 ml histopaque-1077 was underlayed at the bottom of
each tube, which were then centrifuged at 259 x g for 30 minutes at room
temperature without brake in a 5804 R centrifuge (Eppendorf). The top PBS
level from each tube was removed and discarded and the buffy coat layer
transferred to a fresh tube. The total volume was made up to 50 ml with PBS
and the tubes were then centrifuged for 10 minutes at 259 x g at room
temperature. The cells were washed an additional 3 times with PBS in this
manner.
The cell (PBMC) pellet was then resuspended in 30-40 ml complete
(RPMI1640) media. PBMCs were seeded at either 2.5 or 7.5 x 106 cells/ml
complete media (lx and 3x seedings, respectively) and allowed to rest overnight
before compound exposure for 24 hours. The cells and media were then
collected, centrifuged for 5 minutes at 735 x g in a 5415 C microfuge
(Eppendorf) at room temperature and the supernatant analyzed by IFN-a ELISA.
The ability of Formula I prodrugs, Formula II compounds, and other compounds
of the invention to demonstrate favorable oral delivery characteristics and to
induce immune responses when administered by a selected route can be
compared with the results of similar experiments with compounds described in
the literature. Hereby incorporated by reference in their entireties are U.S.
Patent Nos. 5,041,426 and 4,880,784, and U.S. Patent Application No.
136

WO 2006/066080 PCT/US2005/045589
10/861,430 (U.S. Patent Application Publication No. US 2005/0070556), which
disclose, inter alia, IFN-ot induction of isatoribine.

Accordingly, relative activities of the compounds of the invention are
expressed as a percentage of the level of IFN-cc induction by either 32 or 100
uM isatoribine.
ELISA Protocol
Human IFN-a ELISA (#KHC4012) was performed as described in
Biosource protocol. However, to ensure that readings were within the linear
range of detection, PBMC supernatant samples were typically diluted 1:3 (2.5 x
106 cells/ml seeding) or 1:15 (7.5 x 106 cells/ml seeding) and then analyzed
together with undiluted supernatant samples. The concentration in each sample
was calculated from the O.D. by reference to a standard curve.
The compounds of the invention exhibited TFN-a induction from PBMCs
relative to isatoribine in the following ranges:
0-10%: Compounds 142,157,141,148, and 33
11-50%: Compounds 152 and 165
51-100%: Compounds 32,160,130, and 137
>100%: Compounds 133,34,147,121,122,134,117,132, and 153.
Comparison to Isatoribine
The results demonstrate the remarkable superiority of 134 and 122 to
isatoribine with respect to enhancement of IFN-a production from human PBMCs in
vitro. Figure 1 and Figure 2 show plots of pg/ml IFN-a induced in human PBMCs
from compounds 134 and 122 vs pg/ml IFN-a induced by an identical concentration
of isatoribine. The results may be summarized as follows.
At IX PBMC seeding, both 134 and 122 induce significantly and
substantially more EFN-a production at 100 uM test compound than does isatoribine,
especially for weak responders to isatoribine (left panel on Figures 1 and 2). When
seeding is increased to 3X, the difference between the amount of IFN-a produced at
100 uM test agent is less clear-cut although visually apparent and statistically
137


WO 2006/066080 PCT/US2005/045589
significant (right panel, Figures 1 and 2). When the concentration of both 134 and
122 is 32 uM at 3X seeding, however, both 134 and 122 dramatically and
unexpectedly outperform isatoribine (right panel and inserts of Figures 1 and 2).
It is to be understood that the foregoing description is exemplary and
explanatory in nature, and is intended to illustrate the invention and its preferred
embodiments. Through routine experimentation, the artisan will recognize
apparent modifications and variations that may be made without departing from
the spirit of the invention. Thus, the invention is intended to be defined not by
the above description, but by the following claims and their equivalents.
138

WO 2006/066080 PCT/US2005/045589
What is claimed is:
1. A compound of Formula I
whereia
X is O or S,
Y is O or S,
R1 is H, alkyl, aryl, cycloalkyl, or heterocyclyl,
R2 is NH2, -NHC(O)R4, -NHR5, -N=CHNR6R7,
R3 is H,Cl,Br,or OR8,
R4 is -C1-C7-alkyl or -O(C1-C7-alkyl),
R5 is -C1-C7-allcyl,
R6 and R7 are independently -C1-C7-alkyl or together with nitrogen form a 5- or 6-
membered heterocyclic ring,
Rs-CHRR10,
R9 is H, -C1-C7-alkyl, cycloalkyl, aryl, heterocyclyl, ~NRnR12, or OR5,
R10 is -C1-C7-alkyl, cycloalkyl, aryl, heterocyclyl, -NRnR12, or OR5,
Rn and R12 are independently H, -C1-C7-alkyl, or -C(O)R4,
wherein when X is O, Y is S, and R3 is H, Cl, Br, or OR8, R1 is not H or p-D-ribose
or esters thereof,
wherein the above alkyl, aryl, cycloalkyl, or heterocyclyl moieties are optionally
substituted by 1-4 substituents selected from
hydrogen,
alkanoyl,
alkylamine,
amino,
aryl, cycloalkyl, heterocyclyl,
azido,
C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 alkoxy, C1-C6
alkylamine, C1-C6 dialkylamine, C2-C6 alkenyl, or C2-C6 alkynyl, wherein
each of which may be interrupted by one or more hetero atoms,
139

WO 2006/066080 PCT/US2005/045589
carboxyl,
cyano,
halo,
hydroxy,
mercapto,
nitro,
thioalkyl,
-N=N-NH2,
-C(O)2-(C1-C6 alkyl), -C(O)2-(aryl), -C(OMcyeloalkyl), -C(O)2-
(lieterocyclyl), -O-(C1-C6 haloalkyl), -O-(C1-C6 alkyl)aryl, -C1-C6-Ce
a]kyl)cycloalkyi, -O-(C1-C6 alkyl)heterocyclyl, -O-(C1-C6 alkyl)amino, -O-
(C1-C6 alkyl)alkylamino, -O-(C1-C6 alkyl)dialkylamino, -O-(C1-C6 alkyl)-
C(O)-amino, -O-(C1-C6 alkyl)-C(O)-alkylamino, -O-(C1-C6 alkyl)-S(O)2-
amino, -O-(C1-C6 alkyl)- S(O)2-alkylamino, -O-(C1-C6 alkyl)- S(O)2-
dialkylamino, -O-(C1-C6 alkyl)-C(O)-dialkylamino, -O-aryl, -O-heterocyclyl,
-NHC(O)-(C1-C6 alkyl), -NHC(O)-(C1-C6 alkenyl), -NHC(OMaryl),
-NHC(0)-(cycloalkyl), -NHC(OHheterocyclyl), -NHC(O)-(C1-C6
alkyl)aryl, -NHC(OMC1-C6 alkyl)cycloalkyl, -NHC(O)-(C1-C6
alkyl)heterocyclyl, -NHC(O)-(C1-C6 alkyl)amino, -NHC(O)-(C1-C6
alky)alkylamine, -NHC(O)-(C1-C6 alkyl)dialkylamine, -NHC(O)-(C1-C6
alkyl)C(O)amino, -NHC(O)-(C1-C6 alkyl)C(O)alkylamine, -NHC(O)-(C1-C6
alkyl)C(O)dialkylamine, -NHC(O)-(C1-C6 alkyl)N(H)-(C1-C6 alkyl)C(O)2-.
(C1-C6 alkyl), -NH-(C1-C6 alkyl)-C(O)-amino, -NH-(C1-C6 alkyl)-C(O)-
alkylamino, -NH-(C1-C6 aIkyl)-C(O)-dialkylamino, -NHC(O)(C1-C6
alkyl)S(O)2(C1-C6 alkyl), -NHC(O)-(C1-C6 alkyl)-S-(heterocyclyl),
-NHS(O)2-(C1-C6 alkyl), -NHS(O)2-(aryl), -NH-(C1-C6 alkyl)- S(O)2-
amino, -NH-(C1-C6 alkyl)- S(O)2-alkylamino, -NH-(C1-C6 alkyl)- S(O)2-
dialkylamino, -NHS(O)2-(cycloalkyl), -NHS(O)2-(heterocyclyl),
-NHS(O)(C1-C6 alkyl), -NHS(O)(aryl), -NHS(O)(cycloalkyl),
-NHS(O)(heterocyclyl), -NHS(C1-C6 alkyl), -NHS(aryl),
-NHS(cycloalkyl), and-NH-S-(heterocyclyl),
wherein each of the above substituents can be further optionally substituted
by 1-5 substituents selected from
amino,
140

WO 2006/066080 PCT/US2005/045589
C1-C6 alkylaraine, C1-C6 dialkylamine,
C1-C6 alkyl, C1-C6 alkoxy, C1-C6 alkenyl, C1-C6 hydroxyl, and C1-C6
hydroxyalkyl, each optionally substituted by
cyano,
halo, and
nitro,
or a pharmaceutically acceptable salt or hydrate thereof.
2. The compound or pharmaceutically acceptable salt or hydrate according to
claim 1 wherein R2 is NH2.
3. The compound or pharmaceutically acceptable salt or hydrate according to
claim 2 wherein R3 is H.
4. The compound or pharmaceutically acceptable salt or hydrate according to
claim 3 wherein X is O and Y is S.
5. The compound or pharmaceutically acceptable salt or hydrate according to
claim 4 wherein R1 is selected from

141

142
WO 2006/066080 PCT/US2005/045589


143
WO 2006/066080 PCT/US2005/045589


144
WO 2006/066080 PCT/US2005/045589


145
WO 2006/066080 PCT/US2005/045589


146
WO 2006/066080 PCT/US2005/045589


147
WO 2006/066080 PCT/US2005/045589


WO 2006/066080 PCT/US2005/045589
8. A compound of Formula II
wherein
X is O or S,
Y is O or S,
Z is O or CH2,
R2 is -NH2, -NHC(O)R4, -NHR5, -N=CHNR6R7,
R4 is -C1-C7-alkyl or -O(C1-C7-alkyl),
R5 is -C1-C7-alkyl,
R6 and R7 are independently -C1-C7-alkyl or together with nitrogen form a 5- or 6-
membered heterocyclic ring,
R13 is OH or SH,
R14 is H, -CH2OH, or -CH2-O-C(O)C1-C18 alkyl,
R1S is OH, alkenyl, -OC(O)C1-C18 alkyl, -OC(O)aryl, or -OC(O)heterocyclyl,
R16, R17, R18, and R19 are independently H, halo, N3, alkyl, -(CH2)mOR20,
-(CH2)mOC(O)C1-C18 alkyl, -OC(O)aryl, -OS(O)2aryl, or R16 and R17 are an alkenyl, or
R17 and R19 combine together to form a dioxole ring,
R20 is H or alkyl, .
m is 0 or 1,
n is 1 or 2,
wherein if R2 is NH2, then one of the following must be present:
Z is CH2;
either n is 2 or m is 1;
at least one of R16, R17, R18, and R19 is halo, N3, alkyl, or -(OH2)mOR20 wherein m is
1, and wherein if R17 is N3, then R18 and R19 are not H, and wherein if R17 is OH and
R16 and R19 are H, then R18 is not F; or
R16 and R17 are an alkenyl,
148

WO 2006/066080 PCT/US2005/045589
wherein the above alkyl, aryl, cycloalkyl, or heterocyclyl moieties are optionally
substituted by 1-4 substituents selected from
hydrogen,
alkanoyl,
alkylamine,
amino,
aryl, cycloalkyl, heterocyclyl,
azido,
C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 alkoxy, C1-C6
allcylamine, C1-C6 dialkylamine, C2-C6 alkenyl, or C2-C6 alkynyl, wherein
each of which may be interrupted by one or more hetero atoms,
carboxyl,
cyano,
halo,
hydroxy,
mercapto,
nitro,
ihioalkyl,
-N=N-NH2,
-C(O)2(C1-C6 alkyl), -C(O)2-(aryl), -C(O)2-(cycldalkyl),-C(O)2-
(heterocyciyl), -O-(C1-C6 haloalkyl), -O-(C1-C6 alkyl)aryl, -O-(C1-C66
alkyl)cycloalkyl, -O-(C1-C6 alkyl)heterocyclyl, -O-(C1-C6 alky)amino, -O-
(C1-C6 alkyl)alkylamino, -O-(C1-C6 alkyl)dialkylamino, -O-(C1-C6 alkyl)-
C(O)-amino, -O-(C1-C6 alkyl)-C(O)-alkylamino, -O-(C1-C6 alkyl)-S(O)2-
amino, -O-(C1-C6 alkyl)- S(O)2-alkylamino, -O-(C1-C6 alkyl)- S(O)2-
dialkylamino, -O-(C1-C6 alkyl)-C(O)-dialkylamino, -O-aryl, -O-heterocyclyl,
-NHC(O)-(C1-C6 alkyl), -NHC(O)-(C1-C6alkenyl), -NHC(OHaryl),
-NHC(OHcycloalkyl),-NHC(O)-(heterocyclyi),-NHC(O)-(C1-C6
alkyl)aryl,-NHC(O)~(C1-C6 alkyl)cycloalkyl,-NHC(O)-(C1-C6
alkyl)heterocyclyl, -NHC(O)-(C1-C6 alkyl)amino, -NHC(O)-(C1-C6
alkyl)alkylamine, -NHC(O)-(C1-C6 alkyl)dialkylamine, -NHC(O)-(C1-C6
alkyl)C(O)amino, -NHC(O)-(C1-C6 alkyl)C(O)alkylamine, -NHC(O)-(C1-C6
alkyl)C(O)dialkylamine, -NHC(O)-(C1-C6 alkyl)N(H)-(C1-C6 alkyl)C(O)2-
(C1-C6 alkyl), -NH-(C1-C6 alkyI)-C(O)-amino, -NHK(C1-C6 alkyl)-C(O)-
149

WO 2006/066080 PCT/US2005/045589
alkylamino, -NH-(C1-C6 alkyl)-C(O)-dial]cylamino, -NHC(O)-(C1-C6
alkyl)S(O)2(C1-C6 alkyl), -NHC(O)-(C1-C6 alkyl)-S-(heterocyclyl),
-NHS(O)2-(C1-C6 alkyl), -NHS(OMaryl), -NH-(C1-C6 alkyl)- S(O)2-
amino, -NH-(C1-C6 alkyl)- SCO-alkylamino, -NH-(C1-C6 alkyl)- SiPV
dialkylamino, -NHS (O)2(cycloalkyl), -NHS(O)2-(heterocyclyl),
-NHS(O)(C1-C6 alkyl), -NHS(O)(aryl), -NHS(O)(cycloaIkyl),
-NHS(O)(heterocyclyl), -NHS(C1-C6 alkyl), -NHS(aryl),
-NHS(cycloalkyl), and -NH-S-(heterocyclyl),
wherein each of the above substituents can be further optionally substituted
by 1-5 substituents selected from
amino,
C1-C6 alkylamine, C1-C6 dialkylamine,
C1-C6 alkyl, C1-C6 alkoxy, C1-C6 alkenyl, C1-C6hydroxyl, andC1-C6
hydroxyalkyl, each optionally substituted by
cyano,
halo, and
nitro,
or a pharmaceutically acceptable salt, hydrate, or tautomer thereof.
9. The compound or pharmaceutically acceptable salt, hydrate, or tautomer
according to claim 8 wherein R2 is NH2.
10. . The compound or pharmaceutically acceptable salt, hydrate, or tautomer
according to claim 9 wherein R13 is OH.
11. The compound or pharmaceutically acceptable salt, hydrate, or tautomer
according to claim 10 wherein X is O and Y is S.
150

WO 2006/066080 PCT/US2005/045589
151
12. The compound or pharmaceutically acceptable salt, hydrate, or tautomer
according to claim 8 selected from


152
WO 2006/066080 PCT/US2005/045589


153
WO 2006/066080 PCT/US2005/045S89


WO 2006/066080 PCT/US2005/045589


WO 2006/066080 PCT/US2005/045589

or a pharmaceutically acceptable salt or solvate thereof
17. A method of modulating immune cytokine activities in a patient comprising:
administrating to the patient a therapeutically or prophylactically effective
amount of a compound of claim 1, or pharmaceutically acceptable salt or solvate
thereof.
18. The method according to claim 17 wherein the compound is selected from

19. A method of treating a hepatitis C virus infection in a patient comprising:
administrating to the patient a therapeutically or prophyiactically effective
amount of a compound of claim 1, or pharmaceutically acceptable salt or solvate
thereof.
155
155
20. The method according to claim 19 wherein the compound is selected from

The invention is directed to 3,5-disubstituted and 3,5,7-trisubstituted-3-H-oxazolo and 3H-thiazolo[4,5-d]pyrimidin-2-one compounds and prodrugs thereof that have immunomodulatory activity. The invention is also directed
to the therapeutic or prophylactic use of such compounds and pharmaceutical compositions containing them, and to methods of
treating diseases and disorders described herein, by administering effective amounts of such compounds and prodrugs.

Documents:


Patent Number 253821
Indian Patent Application Number 2552/KOLNP/2007
PG Journal Number 35/2012
Publication Date 31-Aug-2012
Grant Date 28-Aug-2012
Date of Filing 09-Jul-2007
Name of Patentee ANADYS PHARMACEUTICALS, INC.
Applicant Address 3115 MERRYFIELD ROW, SAN DIEGO CALIFORNIA
Inventors:
# Inventor's Name Inventor's Address
1 WEBBER, STEPHEN E. 3531, MILLIKIN AVE. SAN DIEGO, CALIFORNIA 92122
2 LENNOX, JOSEPH R. 7174, SCHILLING AVE., APT. 54, SAN DIEGO, CALIFORNIA 92126
3 XIANG, ALAN X. 10414-95, SCRIPPS POWAY PKWY, SAN DIEGO, CALIFORNIA 92131
4 RUEDEN, ERIK J. 9249, CARLTON OAKS, APT. 40, SANTEE, CALIFORNIA 92071
5 HALEY, GREGORY J. 14711, CAMINITO MAR DE PLATA, DEL MAR, CALIFORNIA 92014
PCT International Classification Number A61K 31/435
PCT International Application Number PCT/US05/045589
PCT International Filing date 2005-12-16
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 NA 2007-07-09 Not Applicable