CA2767253A1 - Novel nucleic acid prodrugs and methods of use thereof - Google Patents

Novel nucleic acid prodrugs and methods of use thereof Download PDF

Info

Publication number
CA2767253A1
CA2767253A1 CA2767253A CA2767253A CA2767253A1 CA 2767253 A1 CA2767253 A1 CA 2767253A1 CA 2767253 A CA2767253 A CA 2767253A CA 2767253 A CA2767253 A CA 2767253A CA 2767253 A1 CA2767253 A1 CA 2767253A1
Authority
CA
Canada
Prior art keywords
alkyl
aryl
nucleic acid
hydrogen
och2ch2s
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2767253A
Other languages
French (fr)
Inventor
Gregory L. Verdine
Meena
Naoki Iwamoto
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wave Life Sciences Pte Ltd
Original Assignee
ONTORII Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43429503&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2767253(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by ONTORII Inc filed Critical ONTORII Inc
Publication of CA2767253A1 publication Critical patent/CA2767253A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/712Nucleic acids or oligonucleotides having modified sugars, i.e. other than ribose or 2'-deoxyribose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/655Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms
    • C07F9/65515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having oxygen atoms, with or without sulfur, selenium, or tellurium atoms, as the only ring hetero atoms the oxygen atom being part of a five-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals

Abstract

Described herein are nucleic acid prodrugs and nucleic acid prodrugs comprising chiral phosphorous moieties. Also described herein are methods of making and using nucleic acid prodrugs and nucleic acid prodrugs comprising chiral phosphorous moieties.

Description

NOVEL NUCLEIC ACID PRODRUGS AND METHODS OF USE THEREOF
CROSS REFERENCE
[0001] This application claims the benefit of U.S. Provisional Application No.
61/223,369, filed July 6, 2009, and U.S. Provisional Application No. 61/242,722, filed September 15, 2009, each of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
[0002] Described herein are nucleic acid prodrugs and nucleic acid prodrugs comprising chiral phosphorous moieties and methods of making and using the same.
BACKGROUND OF THE INVENTION
[0003] Oligonucleotides are useful in therapeutic, diagnostic, research, and new and nanomaterials applications. The use of natural sequences of DNA or RNA is limited, for example, by their stability to nucleases. Additionally, in vitro studies have shown that the properties of antisense nucleotides such as binding affinity, sequence specific binding to the complementary RNA, stability to nucleases are affected by the configurations of the phosphorous atoms. Therefore, there is a need for prodrugs of stereodefined oligonucleotides to impart additional stability to oligonucleotide molecules in a number of in-vitro and in-vivo applications Prodrugs of stereodefined oligonucleotides which comprise phosphorus atom-modified nucleic acids and methods of use thereof are described herein.
SUMMARY OF THE INVENTION
[0004] One embodiment provides for a chiral nucleic acid prodrug.
[0005] One embodiment provides a nucleic acid prodrug having the following structure:
R1 O Ba X" 'O
[o"o_a]
PR2 n Formula 1 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+i, Li+', or K+i;

Y2 is 0, NR d' or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR , wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
at least one instance of X is -OCH2CH2S-S(O)2Rio, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, R12 O R12 O ,O,^,iS R11 ,S"-"S R11 --O-KO--,-Rl l --S O-J~R11 O O
0"~OR11 -S'-~O~rR11 S~R11 S)~ R11 I50R11 --O O)f R11 S O, R1o S O, ,O O, O R
~~ R1o ~~ R10 ~ ~S. ~ 11 A l O R11 O R11 ,0,-\@.Me ~~Me 0 0 Me , 9Me QMe ~.Me O NMe ,S N.Me Me Me , 0 , 0 Q Me Q Me i0 N. iS N" /Z R15R16 0 11 ~~~ Me \~~ Me- 'S-S-RW

O O q O or R1s Z I S-S{-/) q w.

R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
Rio is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl;
Z is S or O;
g is 0, 1, or 3;
w is 1, 2, 3, 4, 5, or 6;
R15 and R16 are independently hydrogen or methyl;
R17 is selected from alkyl, aryl or a CH2CH=CH2;

N O -N -N-N Me R18 is selected from N(CH3)2, -, , , , and I-N
McO2C ;and n is an integer of 1 to about 200.
[0006] Another embodiment provides for a nucleic acid prodrug having the following structure:
R1 Ba X, 'O
[o"o_a]
R2 n Formula 1 wherein R1 is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y1 is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;

each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

2 0 R12 0 ,0,^,/S R11 O O R11 \SD~R11 0 at least one instance of X is S O, R1o S"/~. O, \'O""_YO'RjO O R
R10 ~S. 11 / I~

~S O Ri l '5~/O R11 \,O,,.,~G),Me r -Me 0 0 Me , 9Me QMe \O(N .Me \S(NMe Me Me O O

Q Me 4 Me \,O,~,[N"Me ,S,'/ (N,Me 0 , or 0 R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
Rio is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl; and n is an integer of 1 to about 200.
[0007] A further embodiment provides the nucleic acid prodrug wherein each X-phosphonate moiety of the compound of Formula 1 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
[0008] A further embodiment provides the nucleic acid prodrug wherein each X-phosphonate moiety has a Rp configuration.
[0009] A further embodiment provides the nucleic acid prodrug wherein each X-phosphonate moiety has a SP configuration. A further embodiment provides the nucleic acid prodrug wherein each X-phosphonate independently has a RP configuration or a Sp configuration.
[0010] A further embodiment provides the nucleic acid prodrug wherein R10 is methyl. A further embodiment provides the nucleic acid prodrug wherein R11 is methyl. A further embodiment provides the nucleic acid prodrug wherein R12 is methyl.
[0011] A further embodiment provides the nucleic acid prodrug, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from /--O-KO--,-Rll , AS K

S R11 S O, \'O""-Yo' ~
~~ R10 R10 'S, \SR
1o O R11 \S O R11 R11 O O O
QMe Me Me Me , Me , 0 9Me Q Me 4 Me ,S, N.Me ,O, N.Me ,S\ ~N\Me 0 , 0 and 0 A
further embodiment provides the nucleic acid prodrug, wherein at least 50% of the X moieties of the nucleic ~2~0 11 R12OO~ R11 acid prodrug are independently selected from AOOR , ~ S K
S R11 S O, \'O""-Yo' ~ ~~ R1o R10 'S' VS O, R1o O R11 O R11 ,S,,-, ~O R11 O O O

QMe Me Me Me , Me , 0 QMe Q Me Q Me ,S,,,--yN.Me ,O,,,,-YN.Me \,S,,_,,--yN.Me 0 , 0 and 0 A
further embodiment provides the nucleic acid prodrug, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from \S R11 S O, ,R1 o l, 'S, S O, R1o O R11 ~S O R11 ,S,,,,,O R11 QMe NMe \.-S-."-& Me \,O,, Me I ~Me ~ ~Me Me , Me , 0 QMe Q Me Q Me vS, N.Me ,O, N.Me ,S,,/ N,Me 0 , 0 and 0 A
further embodiment provides the nucleic acid prodrug, wherein each X moiety of th`e` nucleic acid prodrug is R12 0 0 R12 O f ,O,-"S~ R11 O KO J" -"SKO~R11 101 independently selected from I'S O, Rio S O, \'O""^YO'Rjo O R
/ ~~ R10 is. 11 ~S O Rll S O R11 ` Ir -Me 0 0 , Me 9Me QMe Y.Me N. Me ,S N.Me Me , 0 , 0 Q Me Y--~ Me \,O,,,,-YN=Me ,S,,N,Me 0 ,and 0
[0012] A further embodiment provides the nucleic acid prodrug, wherein each X
moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2Rio, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, O O R11 "S O)~R11 0 , IS"--,,S YR11 ,10,-~,,OYR11 -"S,-"i0-R11 /'S SyR11 S)f R11 --S OyR11 s o, \'O"""-'YO' O O~R11 ~~ R1o R10 O O O S"S.

I'S O, Rio O R11 \S O R11 /O R11 O O O

QMe Me Me Me , Me , O
9Me 4 Me 4 Me ,S, N.Me ,e ,S,/ (N,Me 0 , 0 , 0 t S-S-R17 Z*S-S
) q ii q W
0 or R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
Rio is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl;
Z is S or 0;
g is 0, 1, or 3;
w is 1, 2, 3, 4, 5, or 6;
R15 and R16 are independently hydrogen or methyl;
R17 is selected from alkyl, aryl or a CH2CH=CH2; and -N O I-N ~-N ~-N NMe Rig is selected from N(CH3)2, , and I-N

McO2C
[0013] One embodiment provides a nucleic acid prodrug having the following structure:
R1 0 a 0 [>o_ia]

R2 n wherein R1 is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

Rio 2 S II O\ S.
at least one X is, S O R11 0 "S'R11 S O, Rio O R11 \S O R11 O R11 O O O
QMe Q Me v5~~(YD.Me VS N.Me VMe Me , 0 , or 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200.
[0014] A further embodiment provides a nucleic acid prodrug wherein R10 is methyl. A further embodiment provides a nucleic acid prodrug wherein R11 is methyl. A further embodiment provides a nucleic acid prodrug wherein R12 is methyl
[0015] A further embodiment provides a nucleic acid prodrug, wherein at least 25% of the X moieties of the X R1o 2 YO.
nucleic acid prodrug are independently selected from S O R11 0 S O, R1o O R11 \S O R11 /S\,,, O R11 QMe Q Me v5~~(YD.Me VS N.Me VMe Me , 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. A further embodiment provides a nucleic acid prodrug, wherein at least 50% of \
the X moieties of the nucleic acid prodrug are independently selected f r o m V S O, R1o S O=R O R11 ~S O R11 O 'S"S.R

QMe R11 ,S - Me \,S,,,,-YN,M
e ~ ~Me 0 Me , 0 , and YMe ,S, N.Me 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. A
further embodiment provides a nucleic acid prodrug, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently I'S O, 2 0 S\'/O'R1o S O'l R11 _ fl selected from S O R11 0 'S' R1 1 0 ~S O T R11 S O R11 ,S,,,,--,&,Me -Me 0 0 , Me QMe Q Me ,S,N.Me ,S,,N,Me 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; RI l is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
A further embodiment provides a nucleic acid prodrug, wherein at each X moiety of the nucleic acid prodrug 2 VS\ /O'S.
R1o is independently selected from S O R11 0 "S" R11 V S O, O R11 \S O R11 ,S,,,,,O R11 QMe Q Me Me S N,Me \,S,, N.Me e Me , 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0016] One embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the following structure:

R1 Ba X, .O
O Ba R2 n Formula 1 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na", Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
wherein at least one X moiety of the nucleic acid prodrug is independently selected from R12 O R12 O,S R11 \S ""'_Y0' __OO~R DSO-KR Obi R10 S O, R1o O 0.R O R11 R11 /S"'~'-'/O R11 R11 R11 O O O

QMe ~O 1 Me \SMe O NMe e Me Me , Me , 0 QMe Q Me Q Me _S(N.Me -_S~ (N,Me 0 0, and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of. (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
[0017] Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1, wherein each X-phosphonate moiety of the compound of Formula 1 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1 wherein each X-phosphonate moiety has a RP
configuration. Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1, wherein each X-phosphonate moiety has a SP configuration. Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1 wherein each X-phosphonate independently has a RP configuration or a Sp configuration.
[0018] Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1, wherein at least 25% of the X moieties of the nucleic acid prodrug are R12 0 R12 0 /,0,,-~S R11 O O R11 ~S 0 R11 independently selected from , 0 I'S O, Rio S O, \'0""^Y0'Rjo O R
/ ~~ R10 iS. 11 \S O R11 O R11 ~/SOR11 R11 \,O,,.,\G),Me \,S,,.,-\Y(@,Me r -Me -Me 0 , Me , Me 9Me QMe Q Me \,0,,,--,YN~Me vMe 0 , 0 and Y--~ Me ,e O ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from R12 O R12 O ~/S R11 iS O'--O-KO~R "S O~R O R10 S 01 Rio '0 R11 'S Ri i O /"S '&R1, 0 0 ~S O R1, ~S O R11 R11 , , YMe Me Me Me , Me , 0 YMe Q Me 4 Me ,S, N.Me \,0,,,,-YN~Me ,S (N,Me 0 , 0 , and 0 wherein R10 is an alkyl group having 1 to 4 carbon atoms; Rll is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from 0 0 R11 12 0 SvR11 ''~,"S O`R10 "O~YO'R1o S O R11 IOI O O\SR1o "S. O"T R11 ~S 0 R11 O R11 /SOR11 R11 ~.Me ~ ~Me O O Me YMe YMe \,S,.,~Y(D,Me ,\SyN.Me -Me Me O O

YMe YMe ,,S, N,Me 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1, wherein each instance of X is independently selected from S O, R1o R12 O \S"'-Y0'Rj0 O R11 /-'s "KO"R" R11 O 'S'S'R

O O O

R11 N.Me ~.Me I I ~Me -Me 0 , Me , Me 9Me QMe Q Me \,O,,,,-YN" Me 5/-y N11 Me ,Me 0 , 0 , 0 , and 4 Me ,S, N.Me O ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0019] Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1 wherein R10 is methyl. Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1 wherein R11 is methyl.
Another embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the structure of Formula 1 wherein R12 is methyl.
[0020] One embodiment provides a method of treating a disease associated with upregulated RNase L by administering a therapeutic amount of a chiral nucleic acid prodrug. Another embodiment provides a method of treating a disease associated with upregulated RNase L, wherein the disease is chronic fatigue syndrome. Another embodiment provides a method of treating a disease associated with downregulated RNase L by administering a therapeutic amount of a chiral nucleic acid prodrug. Another embodiment provides a method of treating a disease with downregulated RNase L, wherein the disease is cancer. IN

anothoer embodiment, the cancer is selected from prostate, colorectal, and pancreatic cancer. In one embodiment, the cancer with downregulated RNase L is pancreatic cancer. In another embodiment the cancer with downregulated RNase L is prostate cancer. In yet another embodiment, the cancer with downregulated RNase L is colorectal cancer.
[0021] One embodiment provides a method of treating cancer comprising administering a therapeutic amount of a nucleic acid prodrug having the following structure:
R1 O Ba X, 'O
O'~P "'O O Ba R2 n Formula 1 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na", Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

least one X moiety of the nucleic acid prodrug is independently selected from 2 0"~S1R11 S 01 R10 i ~ ( R10 S O, Rio R11 R11 O R11 O'l "S.
R11, S 0 0 0 ~S O R11 S O R11 \OMe -Me 0 0 , Me , 9Me QMe \O(N .Me \S(NMe Me Me O O

Q Me 4 Me \,O,,,,-YN=Me Me 0 , and 0 ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
R10 is an alkyl group having 1 to 4 carbon atoms;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of. (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
[0022] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein at least 25% of the X moieties of the nucleic acid prodrug are /independently selected from 'OOR
, S O, Rio S""-YO,R10 \'O""'^YO'Rjo "S. O R

O R11 ~S O R11 /SOR11 ,Me R11 N, .Me N, I Me I Me 0 , Me , Me 9Me QMe Q Me \,O,,,II N\ Me ,S\',,--YN,Me Me 0 , 0 and Q Me ,e O ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from R12 0 R12 0 S R11 \S ""-Y0' S O, '0 R11 ,S R11 iS O R11 /s0R11 O R11 QMe N.Me ,Me ,I " Me I ~Me Me , Me , 0 QMe Q Me YMe ,S,N.Me ,,S,N,Me 0 , 0 , and 0 wherein R10 is an alkyl group having 1 to 4 carbon atoms; Rll is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein at least 90% of the X

moieties of the nucleic acid prodrug are independently selected from 'OOR

12 SuR11 S O`R10 'O~YO'R1o S O R11 IOI O O\SR1o R11 /s0R11 ,S,,-, /O R11 "S' O'l S R11 0 0 0 ~S O R11 R11 ~.Me ~ QMe O Me QMe QMe Y.-Me Me ~O N.Me \S(NMe ~
Me O O
Q Me Q Me ,,S,,N,Me 0 , and 0 ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein each instance of X is independently selected from S O, R1o R1S\~/O.R1o O R11 /IS "K-O"R" ~ R11 O " S'S`R11 O

O O O
QMe R11 .\,e Y -Me O Me O and 4 Me ,S,, N.Me 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0023] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein R10 is methyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein R11 is methyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein R12 is methyl.
[0024] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein each X-phosphonate moiety of the compound of Formula 1 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein each X-phosphonate moiety has a RP
configuration. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein each X-phosphonate moiety has a SP
configuration. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein each X-phosphonate independently has a RP configuration or a Sp configuration.
[0025] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound of Formula 1, wherein the cancer is pancreatic cancer.
[0026] One embodiment provides a nucleic acid prodrug having the following structure:

R

x 0 FF OR3 n Fomnala 2 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+i, Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

2 0 R12 0 /,0,,,,/S R11 AOOR11_-S-O-_~_ R11 at least one instance of X is v S O, R1o S O, \'0""_Y0'Rj0 O R
/ ~~ R10 iS. 11 Al R11 R11 ~.Me ~~Me 0 0 Me , 9Me QMe VO N.Me \S(NMe Me Me O O

Q Me 4 Me \,O,,,,-YN.Me ,0 , or 0 R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
Rio is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl; and n is an integer of 1 to about 200.
[0027] One embodiment provides a nucleic acid prodrug of Formula 2, wherein each X-phosphonate moiety of the compound of Formula 2 is more than 98% diastereomerically pure as determined by 31P NMR
spectroscopy or reverse-phase HPLC. Another embodiment provides a nucleic acid prodrug of Formula 2, wherein each X-phosphonate moiety has a RP configuration. Another embodiment provides a nucleic acid prodrug of Formula 2, wherein each X-phosphonate moiety has a SP
configuration. Another embodiment provides a nucleic acid prodrug of Formula 2, wherein each X-phosphonate independently has a RP
configuration or a Sp configuration.
[0028] Another embodiment provides a nucleic acid prodrug of Formula 2 wherein Rio is methyl. Another embodiment provides a nucleic acid prodrug of Formula 2wherein R11 is methyl.
Another embodiment provides a nucleic acid prodrug of Formula 2 wherein R12 is methyl
[0029] Another embodiment provides a nucleic acid prodrug of Formula 2, wherein at least 25% of the X

moieties of the nucleic acid prodrug are independently selected from 'OOR

"2) 0 ""S S R11 S~YO`R10 /O~YO\R10 S O, R1o "S. O R11 ~S O R11 R11 QMe NMe ~ .Me \OyN Me I
"Me Me Me , Me , 0 QMe Q Me Q Me ,e \,O,,/~N,Me ,S,,,~N,Me 0 , 0 , and 0 Another embodiment provides a nucleic acid prodrug of Formula 2, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from R11 12 ,0,,~SyR11 -YO'R1 o /O"-"-YO\R10 \,-S O, R1o O R11 ~S O R11 ism/O R11 QMe \,O,,-~@ " Me Me Me Me QMe Me , Me , O
QMe Q Me Q Me ,Me ,e ,e 0 , 0 , and 0 Another embodiment provides a nucleic acid prodrug of Formula 2, wherein at least 90% of the X moieties ~2 0 of the nucleic acid prodrug are independently selected from O O R11 2 )~ \O~~SYR11 S O'R10 \'O""-YO'Rlo S O, O R11 \S O R11 R11 QMe Me Me Me , Me , O
QMe Q Me Q Me ,S, N.Me ,0,,,~N,Me ,e 0 , 0 , and 0 Another embodiment provides a nucleic acid prodrug of Formula 2, wherein each X moiety of the nucleic O O

acid prodrug is independently selected from R11 , AS

S R11 S O, \'O""-Yo' ~ ~~ R1o R 10 'S' VS O, O R11 /s0R11 R11 QMe Me Me Me , Me , 0 9Me Q Me 4 Me ,S,,,--yN.Me ,O,,,,-YN.Me \,S,,_,,--yN.Me 0 , 0 ,and 0
[0030] One embodiment provides a pharmaceutical composition comprising a nucleic acid prodrug having the following structure:
R

x 0 R'- OR3 n Fonrnila 2 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+i, Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

wherein at least one X moiety of the nucleic acid prodrug is independently selected from R12 O R12 O ,/S R11 iS O' ~OO~R ~SO~R O R10 S O, Rio 'O R11 ~S O R11 O AS 'N11 O O

S O R11 \S O R11 'S"_' --'iO R11 O O O
QMe \OMe '\SMe \OyNM
e QMe QMe Me , Me , 0 QMe Q Me Q Me _S N.Me -_N\Me 0 , 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of. (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
[0031] Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein each X-phosphonate moiety of the compound of Formula 2 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein each X-phosphonate moiety has a RP configuration. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein each X-phosphonate moiety has a SP configuration. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein each X-phosphonate independently has a RP configuration or a Sp configuration.
[0032] Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from R12 O R12 O O~/S R11 + S O' ~ ~ R 10 S O, R1o VON'-"'Y0'Rj0 O R11 'S R11 R11 ~S 0 R11 R11 O O O
QMe QMe QMe Me , Me , O
QMe Q Me Q Me , Me ,Me , S, N.Me 0 0, and 0 wherein R10 is an alkyl group having 1 to 4 carbon atoms; Rll is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2, wherein at least 50% of the X moieties of the nucleic acid prodrug are I ,0~~/S~ R11 I
independently selected from 'OOR11~S K , V S O, R1o S'/~. O, ,O R
R10 ~S. "T 11 / I~

0 R11 ~S O R11 /SOR11 ,Me R11 N, .Me N, I Me I Me 0 , Me , Me YMe YMe Q Me \,O,,,II N\ Me ,S\',,--YN,Me Me 0 , 0 , 0 , and Q Me ,S,, N, Me 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2, wherein at least 90% of the X moieties of ~ R 11 the nucleic acid prodrug are independently selected from Ao--K0)~Rjj S R11 S~
O, \'O""-Yo' ~

~ R10 R10 'S' V S O, O R11 /s Rii R11 O R11 ,/O R11 \,0,,,-,,N@,Me 'T I ,Me 0 0 Me YMe YMe N, .Me ~O N. Me Me I Me Me , 0 , 0 Q Me 4 Me ,Me 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2, wherein each instance of X is independently selected from S O
, Rio jS O\R1o O R11 R11 O O O
QMe NMe \SMe ,O N~Me "Me ~Me Me , Me , 0 9Me 9 Me 9Me ,S,N.Me ,0,'/-YN,Me ,S,,N,Me 0 , 0 , and 0 wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0033] Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein R10 is methyl. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein R11 is methyl. Another embodiment provides a pharmaceutical composition comprising a compound of Formula 2 wherein R12 is methyl.
[0034] One embodiment provides a method of treating cancer comprising administering a therapeutic amount of a nucleic acid prodrug having the following structure:

R

x 0 FF bW n Fomnala 2 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+i, Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

Ao__K)~
at least one X moiety of the nucleic acid prodrug is independently selected from O R11 2 0 ""S R11 SO\R10 \'0"-"_Y0'Rj0 S O, R1o 0 R11 \S ~ ~ R11 R11 S O R11 O R11 0.-"~@.Me ~r ~r I,Me 0 0 , Me , 9Me QMe '\O(N .Me ,S N.Me I ~Me Me O O

Q Me 4 Me , Me Me 0 , and 0 ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
R10 is an alkyl group having 1 to 4 carbon atoms;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of. (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
[0035] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein at least 25%
of the X moieties of the ~2O/JII0 , R11 R12OO~ R11 -"S'K
nucleic acid prodrug are independently selected from \ O , S R11 S O, \'O""-Yo' ~
~~ R10 R10 'SI

S O, R1o O R11 ~O 'T R11 /S~/O R11 O O O

O R11 '5,'~-"/O R11 \,O,,,-~Y(D,Me -Me O O Me YMe YMe \,S,,~Y0,Me ,e \SyN.Me -Me Me , 0 , 0 Q Me Q Me \,0,,,,-YN~Me ,S,/ (N,Me 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein at least 50%
of the X moieties of the ~2OII 0 R11 ~SR1K2OO~ R11 nucleic acid prodrug are independently selected from \ O

AO S R11 VS O, ,0i R1o 'S' V S O, Rio O R11 ~S O R11 ,S,,,,,O R11 /SOR11 R11 ,0,,/\@.Me ,Me 0 0 Me YMe YMe \,S,,~Y0,Me ,e \SyN.Me -Me Me , 0 , 0 YMe YMe ,,S, N,Me 0 , and 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein at least 90%
of the X moieties of the O O R11 \SO~
nucleic acid prodrug are independently selected from , R11 AO---~ S R11 S O, V0""-Y0'Rj0 ~ ~~ R
1o 'S' \SR1o O R11 \S O R11 R11 O O O
O R11 S O R11 \,0,,,-~Y(D,Me -Me O O Me 9Me QMe Y.Me N. Me ,S N.Me Me O O

Q Me 4 Me 'Me ,0 , and 0 ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein each instance of X is independently \I- S O, R1o selected from 11 0 "S"S'R11 0 /SOR11 ism/O R11 ~S O R11 O O O
QMe ,S,,,,, ~N-, O R11 N.Me ,S N.Me I ,Me O Me O and Y__~ Me ,S, N.Me 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0036] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein R10 is methyl.
Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein R11 is methyl. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein R12 is methyl
[0037] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein each X-phosphonate moiety of the compound of Formula 2 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
[0038] Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein each X-phosphonate moiety has a RP
configuration. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein each X-phosphonate moiety has a SP configuration. Another embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein each X-phosphonate independently has a RP configuration or a Sp configuration.
[0039] One embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein the cancer is pancreatic cancer.
[0040] One embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein the compound has the following formula:

R,1 O 0 A
RilyS
O O HO
1111 Stiff O OA
I HO
R, 1S-',--p'~ O A
1 f -HO OH
Formula A3-2 wherein each A is adenine and each R11 is independently selected from alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl. A further embodiment provides a method of treating pancreatic cancer comprising administering a therapeutic amount of a compound of Formula A3-2.
[0041] One embodiment provides a method of treating cancer comprising administering a therapeutic amount of a compound having the structure of Formula 2, wherein the compound has the following formula:
,i's-'\__0, //O

~y s O A

S^/0.11!00 O
HO

HO OH Formula A3-3.
[0042] A further embodiment provides a method of treating pancreatic cancer comprising administering a therapeutic amount of a compound of Formula A3-3.
[0043] One embodiment provides a compound or its pharmaceutically acceptable salt having the following formula:

X, P,,O

X \ 0-'~ O A
HO
X"O 'O A

HO
X, .O

HO OH
Formula A3-1 ~ R11 wherein each A is adenine; and at least one X moiety is /--O-KO--,- S R11 S O, \'O""-YO'Rjo ~ ~~ R10 "S.

S O, Rio O R11 /s rR11 R11 O Rll S O R11 \,O,,,-~Y(D,Me -Me 0 0 Me 9Me QMe Y-.Me N. Me ,e Me Me , 0 , 0 Q Me Q Me , Me ,e 0 , or 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0044] Another embodiment provides a compound or its pharmaceutically acceptable salt having the structure of Formula A3-1, wherein at least two of the X moieties of the nucleic acid prodrug are ,O,.,~S~ R11 independently selected f r o m 'OOR11~S O ~ R11 0 , S O, R1o /S~O,R10 ,"S. O"T R11 O R11 R11 ~S 0 R11 O O O
R11 ~Me ,S~\(@.Me r -Me -Me 0 , Me , Me 9Me QMe 4 Me \,O,,,II N\ Me ,S\' (N\Me Me 0 , 0 , 0 , and 4 Me \,S,,,,,-yN,Me O ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0045] Another embodiment provides a compound or its pharmaceutically acceptable salt having the structure of Formula A3-1, wherein at least three of the X moieties of the nucleic acid prodrug are Y
S ~ R11 independently selected from 'OOR11~SK , S O, R1o S O, \'O""'^YO'RjO O R
/ ~~ R10 iS. ~ 11 \S O R11 ,S,,,,,O R11 \S O R11 R11 \,O,,.,\G),Me \,S,,.,-\(@,Me r -Me Y -Me 0 , Me , Me 9Me QMe Q Me ,ON I~ Me vS, Nl~ Me Me 0 , 0 and Y--~ Me ,e 0 ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0046] Another embodiment provides a compound or its pharmaceutically acceptable salt having the structure of Formula A3- 1, wherein each X moiety of the nucleic acid prodrug is independently selected from R12 O R12 O ~/S R11 iS O'--O-KO~R "S O~R O R10 'O R11 R11 ~S O R11 ~S O R11 R11 QMe Me Me Me , Me , 0 9Me Q Me 4 Me ,S, N.Me \,0,,,,-yN~Me ,S (N,Me 0 , 0 , and 0 wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
[0047] Another embodiment provides a compound or its pharmaceutically acceptable salt having the structure of Formula A3- 1, wherein the compound has the following formula:

1 'S-N-Q
-ySJ ,O A
Cy -~
O O HO
~StipO, O
; A
O HO
)Stio O
A
0-1;4 HO OH Formula A3-3.
INCORPORATION BY REFERENCE
[0048] All publications and patent applications disclosed herein in this specification are herein incorporated by reference in their entirety to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWINGS
[0049] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings.
[0050] Figure 1 provides a representative analytical HPLC profile for compound 64 and GSH.
[0051] Figure 2 provides provided a representative HPLC profile of compound 64a, a glutathione adduct, and the final product after release from the pro-moeity.
[0052] Figure 3 provides a plot of conversion over time for compound 64a and 64b.
[0053] Figure 4 provides a reaction timecourse as determined by LC-MS for the glutathione assisted prodrug release of compound 64a.
DETAILED DESCRIPTION OF THE INVENTION
[0054] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in the application including, without limitation, patents, patent applications, articles, books, manuals, and treatises are hereby expressly incorporated by reference in their entirety for any purpose.
[0055] Unless otherwise stated, the following terms used in this application, including the specification and claims, have the definitions given below. It must be noted that, as used in the specification and the appended claims, the singular forms "a" "an" and "the" include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. In this application, the use of "or" or "and" means "and/or" unless stated otherwise. Furthermore, use of the term "including" as well as other forms, such as "include", "includes" and "included" is not limiting.
Certain Chemical Terminology
[0056] Unless otherwise noted, the use of general chemical terms, such as though not limited to "alkyl,"
"amine," "aryl," are unsubstituted.
[0057] As used herein, Ci-C,, includes Ci-C2, Ci-C3 ... C,-CX. By way of example only, a group designated as "Ci-C4" indicates that there are one to four carbon atoms in the moiety, i.e. groups containing 1 carbon atom, 2 carbon atoms, 3 carbon atoms or 4 carbon atoms, as well as the ranges Ci-C2 and Ci-C3. Thus, by way of example only, "Ci-C4 alkyl" indicates that there are one to four carbon atoms in the alkyl group, i.e., the alkyl group is selected from among methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Whenever it appears herein, a numerical range such as "1 to 10" refers to each integer in the given range; e.g., "1 to 10 carbon atoms" means that the group may have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, 6 carbon atoms, 7 carbon atoms, 8 carbon atoms, 9 carbon atoms, or carbon atoms.
[0058] The terms "heteroatom" or "hetero" as used herein, alone or in combination, refer to an atom other than carbon or hydrogen. Heteroatoms are may be independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but are not limited to these atoms. In embodiments in which two or more heteroatoms are present, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others.
[0059] The term "alkyl" as used herein, alone or in combination, refers to a straight-chain or branched-chain saturated hydrocarbon monoradical having from one to about ten carbon atoms, or one to six carbon atoms.
Examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, 2-methyl-l-propyl, 2-methyl-2-propyl, 2-methyl-l-butyl, 3-methyl-l-butyl, 2-methyl-3-butyl, 2,2-dimethyl-l-propyl, 2-methyl-l-pentyl, 3-methyl-l-pentyl, 4-methyl-l-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-l-butyl, 2-ethyl-l-butyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neopentyl, tert-amyl and hexyl, and longer alkyl groups, such as heptyl, octyl and the like. Whenever it appears herein, a numerical range such as "C1-C6 alkyl" or "Cl_6 alkyl", means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. In one embodiment, the "alkyl" is substituted. Unless otherwise indicated, the "alkyl" is unsubstititued.
[0060] The term "alkenyl" as used herein, alone or in combination, refers to a straight-chain or branched-chain hydrocarbon monoradical having one or more carbon-carbon double-bonds and having from two to about ten carbon atoms, or two to about six carbon atoms. The group may be in either the cis or trans conformation about the double bond(s), and should be understood to include both isomers. Examples include, but are not limited to ethenyl (-CH=CH2), 1-propenyl (-CH2CH=CH2), isopropenyl [-C(CH3)=CH2], butenyl, 1,3-butadienyl and the like. Whenever it appears herein, a numerical range such as "C2-C6 alkenyl"
or "C2-6 alkenyl", means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. In one embodiment, the "alkenyl" is substituted. Unless otherwise indicated, the "alkenyl" is unsubstititued.
[0061] The term "alkynyl" as used herein, alone or in combination, refers to a straight-chain or branched-chain hydrocarbon monoradical having one or more carbon-carbon triple-bonds and having from two to about ten carbon atoms, or from two to about six carbon atoms. Examples include, but are not limited to ethynyl, 2-propynyl, 2-butynyl, 1,3-butadiynyl and the like. Whenever it appears herein, a numerical range such as "C2-C6 alkynyl" or "C2-6 alkynyl", means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms. In one embodiment, the "alkynyl" is substituted. Unless otherwise indicated, the "alkynyl" is unsubstititued.
[0062] The terms "heteroalkyl", "heteroalkenyl" and "heteroalkynyl" as used herein, alone or in combination, refer to alkyl, alkenyl and alkynyl structures respectively, as described above, in which one or more of the skeletal chain carbon atoms (and any associated hydrogen atoms, as appropriate) are each independently replaced with a heteroatom (i.e. an atom other than carbon, such as though not limited to oxygen, nitrogen, sulfur, silicon, phosphorous, tin or combinations thereof), or heteroatomic group such as though not limited to -0-0-, -S-S-, -O-S-, -S-O-, =N-N=, -N=N-, -N=N-NH-, -P(O)2-, -O-P(O)2-, -P(O)2-0-1 -S(O)-, -S(O)2-, -SnH2- and the like.
[0063] The terms "haloalkyl", "haloalkenyl" and "haloalkynyl" as used herein, alone or in combination, refer to alkyl, alkenyl and alkynyl groups respectively, as defined above, in which one or more hydrogen atoms is replaced by fluorine, chlorine, bromine or iodine atoms, or combinations thereof. In some embodiments two or more hydrogen atoms may be replaced with halogen atoms that are the same as each another (e.g. difluoromethyl); in other embodiments two or more hydrogen atoms may be replaced with halogen atoms that are not all the same as each other (e.g. 1-chloro-l-fluoro-l-iodoethyl). Non-limiting examples of haloalkyl groups are fluoromethyl, chloromethyl and bromoethyl. A
non-limiting example of a haloalkenyl group is bromoethenyl. A non-limiting example of a haloalkynyl group is chloroethynyl.
[0064] The term "carbon chain" as used herein, alone or in combination, refers to any alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl or heteroalkynyl group, which is linear, cyclic, or any combination thereof. If the chain is part of a linker and that linker comprises one or more rings as part of the core backbone, for purposes of calculating chain length, the "chain" only includes those carbon atoms that compose the bottom or top of a given ring and not both, and where the top and bottom of the ring(s) are not equivalent in length, the shorter distance shall be used in determining the chain length. If the chain contains heteroatoms as part of the backbone, those atoms are not calculated as part of the carbon chain length.
[0065] The term "cycloalkyl" as used herein, alone or in combination, refers to a saturated, hydrocarbon monoradical ring, containing from three to about fifteen ring carbon atoms or from three to about ten ring carbon atoms, though may include additional, non-ring carbon atoms as substituents (e.g.
methylcyclopropyl). Whenever it appears herein, a numerical range such as "C3-C6 cycloalkyl " or "C3.6 cycloalkyl ", means that the cycloalkyl group may consist of 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, i.e., is cyclopropyl, cyclobutyl, cyclopentyl or cyclohepty, although the present definition also covers the occurrence of the term " cycloalkyl " where no numerical range is designated. The term includes fused, non-fused, bridged and spiro radicals. A fused cycloalkyl may contain from two to four fused rings where the ring of attachment is a cycloalkyl ring, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Examples include, but are not limited to cyclopropyl, cyclopentyl, cyclohexyl, decalinyl, and bicyclo [2.2.1] heptyl and adamantyl ring systems.
Illustrative examples include, but are not limited to the following moieties:
^,0, 0,0,0,00, E>, <:>, CI:), CEI, CC, C-0, and the like.
In one embodiment, the "cycloalkyl" is substituted. Unless otherwise indicated, the "cycloalkyl" is unsubstititued.
[0066] The terms "non-aromatic heterocyclyl" and "heteroalicyclyl" as used herein, alone or in combination, refer to a saturated, partially unsaturated, or fully unsaturated nonaromatic ring monoradicals containing from three to about twenty ring atoms, where one or more of the ring atoms are an atom other than carbon, independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but are not limited to these atoms. In embodiments in which two or more heteroatoms are present in the ring, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others. The terms include fused, non-fused, bridged and spiro radicals. A fused non-aromatic heterocyclic radical may contain from two to four fused rings where the attaching ring is a non-aromatic heterocycle, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Fused ring systems may be fused across a single bond or a double bond, as well as across bonds that are carbon-carbon, carbon-hetero atom or hetero atom-hetero atom. The terms also include radicals having from three to about twelve skeletal ring atoms, as well as those having from three to about ten skeletal ring atoms. Attachment of a non-aromatic heterocyclic subunit to its parent molecule can be via a heteroatom or a carbon atom. Likewise, additional substitution can be via a heteroatom or a carbon atom. As a non-limiting example, an imidazolidine non-aromatic heterocycle may be attached to a parent molecule via either of its N atoms (imidazolidin-1-yl or imidazolidin-3-yl) or any of its carbon atoms (imidazolidin-2-yl, imidazolidin-4-yl or imidazolidin-5-yl). In certain embodiments, non-aromatic heterocycles contain one or more carbonyl or thiocarbonyl groups such as, for example, oxo- and thio-containing groups. Examples include, but are not limited to pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl. Illustrative examples of heterocycloalkyl groups, also referred to as non-aromatic heterocycles, include:

o 010 OH I/ N \I II/O11I O
1 1 I I 1 I 7 1 V V ' ' \V/ V S (N) N
UNH HUH Off/ (0) NC ,0 J ~
H S H N

N C~ UN , ~ N (N) SO
O O O O o ON 0 O- o ~O O~O ('-NH HNvNH S HN~O N)H
~~~~6////// ~//6// `--' and the like.
[0067] The terms also include all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides. In one embodiment, the "non-aromatic heterocyclyl" or "heteroalicyclyl" is substituted. Unless otherwise indicated, the "non-aromatic heterocyclyl" or "heteroalicyclyl" is unsubstititued.
[0068] The term "aryl" as used herein, alone or in combination, refers to an aromatic hydrocarbon radical of six to about twenty ring carbon atoms, and includes fused and non-fused aryl rings. A fused aryl ring radical contains from two to four fused rings where the ring of attachment is an aryl ring, and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. Further, the term aryl includes fused and non-fused rings containing from six to about twelve ring carbon atoms, as well as those containing from six to about ten ring carbon atoms. A non-limiting example of a single ring aryl group includes phenyl; a fused ring aryl group includes naphthyl, phenanthrenyl, anthracenyl, azulenyl; and a non-fused bi-aryl group includes biphenyl. In one embodiment, the "aryl" is substituted. Unless otherwise indicated, the "aryl" is unsubstititued.
[0069] The term "heteroaryl" as used herein, alone or in combination, refers to an aromatic monoradicals containing from about five to about twenty skeletal ring atoms, where one or more of the ring atoms is a heteroatom independently selected from among oxygen, nitrogen, sulfur, phosphorous, silicon, selenium and tin but not limited to these atoms and with the proviso that the ring of said group does not contain two adjacent 0 or S atoms. In embodiments in which two or more heteroatoms are present in the ring, the two or more heteroatoms can be the same as each another, or some or all of the two or more heteroatoms can each be different from the others. The term heteroaryl includes fused and non-fused heteroaryl radicals having at least one heteroatom. The term heteroaryl also includes fused and non-fused heteroaryls having from five to about twelve skeletal ring atoms, as well as those having from five to about ten skeletal ring atoms. Bonding to a heteroaryl group can be via a carbon atom or a heteroatom. Thus, as a non-limiting example, an imidazole group may be attached to a parent molecule via any of its carbon atoms (imidazol-2-yl, imidazol-4-yl or imidazol-5-yl), or its nitrogen atoms (imidazol-1-yl or imidazol-3-yl). Likewise, a heteroaryl group may be further substituted via any or all of its carbon atoms, and/or any or all of its heteroatoms. A fused heteroaryl radical may contain from two to four fused rings where the ring of attachment is a heteroaromatic ring and the other individual rings may be alicyclic, heterocyclic, aromatic, heteroaromatic or any combination thereof. A non-limiting example of a single ring heteroaryl group includes pyridyl; fused ring heteroaryl groups include benzimidazolyl, quinolinyl, acridinyl; and a non-fused bi-heteroaryl group includes bipyridinyl. Further examples of heteroaryls include, without limitation, furanyl, thienyl, oxazolyl, acridinyl, phenazinyl, benzimidazolyl, benzofuranyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, benzothiophenyl, benzoxadiazolyl, benzotriazolyl, imidazolyl, indolyl, isoxazolyl, isoquinolinyl, indolizinyl, isothiazolyl, isoindolyloxadiazolyl, indazolyl, pyridyl, pyridazyl, pyrimidyl, pyrazinyl, pyrrolyl, pyrazinyl, pyrazolyl, purinyl, phthalazinyl, pteridinyl, quinolinyl, quinazolinyl, quinoxalinyl, triazolyl, tetrazolyl, thiazolyl, triazinyl, thiadiazolyl and the like, and their oxides, such as for example pyridyl-N-oxide.
Illustrative examples of heteroaryl groups include the following moieties:
H H H H
U U U U> C \N U U
N N N N
INN CNJ N NN CNN
N N S
H
H H N N \ \ \
N O CC N~/~
N N N N N and the like.

In one embodiment, the "heteroaryl" is substituted. Unless otherwise indicated, the "heteroaryl" is unsubstititued.
[0070] The term "heterocyclyl" as used herein, alone or in combination, refers collectively to heteroalicyclyl and heteroaryl groups. Herein, whenever the number of carbon atoms in a heterocycle is indicated (e.g., Ci-C6 heterocycle), at least one non-carbon atom (the heteroatom) must be present in the ring. Designations such as "Ci-C6 heterocycle" refer only to the number of carbon atoms in the ring and do not refer to the total number of atoms in the ring. Designations such as "4-6 membered heterocycle"
refer to the total number of atoms that are contained in the ring (i.e., a four, five, or six membered ring, in which at least one atom is a carbon atom, at least one atom is a heteroatom and the remaining two to four atoms are either carbon atoms or heteroatoms). For heterocycles having two or more heteroatoms, those two or more heteroatoms can be the same or different from one another. Non-aromatic heterocyclic groups include groups having only three atoms in the ring, while aromatic heterocyclic groups must have at least five atoms in the ring. Bonding (i.e.
attachment to a parent molecule or further substitution) to a heterocycle can be via a heteroatom or a carbon atom. In one embodiment, the "heterocyclyl" is substituted. Unless otherwise indicated, the "heterocycyl" is unsubstititued.
[0071] The terms "halogen", "halo" or "halide" as used herein, alone or in combination refer to fluoro, chloro, bromo and/or iodo.
Certain Pharmaceutical Terminology
[0072] The term "subject", "patient" or "individual" as used herein in reference to individuals suffering from a disorder, and the like, encompasses mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fish and the like. In one embodiment of the methods and compositions provided herein, the mammal is a human.
[0073] The terms "effective amount", "therapeutically effective amount" or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound being administered that is sufficient to treat or prevent the particular disease or condition. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in a disease. An appropriate "effective" amount in any individual case may be determined using techniques, such as a dose escalation study.
[0074] As used herein, "treatment" or "treating," or "palliating" or "ameliorating" are used interchangeably herein. These terms refers to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
[0075] A "therapeutic effect," as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
[0076] The term "pharmaceutically acceptable" as used herein, refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compounds described herein, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
[0077] The term "pharmaceutical composition," as used herein, refers to a biologically active compound, optionally mixed with at least one pharmaceutically acceptable chemical component, such as, though not limited to carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
[0078] The term "carrier" as used herein, refers to relatively nontoxic chemical compounds or agents that facilitate the incorporation of a compound into cells or tissues.
[0079] The term "prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein. Thus, the term "prodrug"
refers to a precursor of a biologically active compound that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam). A discussion of prodrugs is provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed.
Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein. The term "prodrug" is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrugs of an active compound, as described herein, may be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to acyloxy, thioacyloxy, 2-carboalkoxyethyl, disulfide, thiaminal, and enol ester derivatives of a phosphorus atom-modified nucleic acid.
[0080] The term "pro-oligonucleotide" or "pronucleotide" or "nucleic acid prodrug" refers to an oligonucleotide which has been modified to be a prodrug of the oligonucleotide.
Certain Nucleic Acid Terminology
[0081] Natural nucleic acids have a phosphate backbone; artificial nucleic acids may contain other types of backbones, but contain the same bases.
[0082] The term "nucleotide" as used herein refers to a monomeric unit of a polynucleotide that consists of a heterocyclic base, a sugar, and one or more phosphate groups. The naturally occurring bases, (guanine, (G), adenine, (A), cytosine, (C ), thymine, (T), and uracil (U)) are derivatives of purine or pyrimidine, though it should be understood that naturally and non-naturally occurring base analogs are also included. The naturally occurring sugar is the pentose (five-carbon sugar) deoxyribose (which forms DNA) or ribose (which forms RNA), though it should be understood that naturally and non-naturally occurring sugar analogs are also included. Nucleic acids are linked via phosphate bonds to form nucleic acids, or polynucleotides, though many other linkages are known in the art (such as, though not limited to phosphorothioates, boranophosphates and the like). Artificial nucleic acids include PNAs (peptide nucleic acids), phosphothionates, and other variants of the phosphate backbone of native nucleic acids.
[0083] The term "nucleoside" refers to a moiety wherein a nucleobase or a modified nucleobase is covalently bound to a sugar or modified sugar.
[0084] The term "sugar" refers to a monosaccharide in closed and/or open form.
Sugars include, but are not limited to, ribose, deoxyribose, pentofuranose, pentopyranose, and hexopyranose moieties.
[0085] The term "modified sugar" refers to a moiety that can replace a sugar.
The modified sugar mimics the spatial arrangement, electronic properties, or some other physicochemical property of a sugar.
[0086] The terms "nucleic acid" and "polynucleotide" as used herein refer to a polymeric form of nucleotides of any length, either ribonucleotides (RNA) or deoxyribonucleotides (DNA). These terms refer to the primary structure of the molecules and, thus, include double- and single-stranded DNA, and double-and single-stranded RNA. These terms include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs and modified polynucleotides such as, though not limited to, methylated and/or capped polynucleotides. The terms encompass poly- or oligo-ribonucleotides (RNA) and poly- or oligo-deoxyribonucleotides (DNA); RNA or DNA derived from N-glycosides or C-glycosides of nucleobases and/or modified nucleobases; nucleic acids derived from sugars and/or modified sugars; and nucleic acids derived from phosphate bridges and/or modified phosphorous-atom bridges. The term encompasses nucleic acids containing any combinations of nucleobases, modified nucleobases, sugars, modified sugars, phosphate bridges or modified phosphorous atom bridges. Examples include, and are not limited to, nucleic acids containing ribose moieties, the nucleic acids containing deoxy-ribose moieties, nucleic acids containing both ribose and deoxyribose moieties, nucleic acids containing ribose and modified ribose moieties. The prefix poly- refers to a nucleic acid containing about 1 to about 10,000 nucleotide monomer units and wherein the prefix oligo- refers to a nucleic acid containing about 1 to about 200 nucleotide monomer units.
[0087] The term "nucleobase" refers to the parts of nucleic acids that are involved in the hydrogen-bonding that binds one nucleic acid strand to another complementary strand in a sequence specific manner. The most common naturally-occurring nucleobases are adenine (A), guanine (G), uracil (U), cytosine (C), and thymine (T).
[0088] The term "modified nucleobase" refers to a moiety that can replace a nucleobase. The modified nucleobase mimics the spatial arrangement, electronic properties, or some other physicochemical property of the nucleobase and retains the property of hydrogen-bonding that binds one nucleic acid strand to another in a sequence specific manner. A modified nucleobase can pair with all of the five naturally occurring bases (uracil, thymine, adenine, cytosine, or guanine) without substantially affecting the melting behavior, recognition by intracellular enzymes or activity of the oligonucleotide duplex.
[0089] The term "chiral reagent" refers to a compound that is chiral or enantiopure and can be used for asymmetric induction in nucleic acid synthesis.
[0090] The term "chiral ligand" or "chiral auxiliary" refers to a moiety that is chiral or enantiopure and controls the stereochemical outcome of a reaction.
[0091] In a condensation reaction, the term "condensing reagent" refers to a reagent that activates a less reactive site and renders it more susceptible to attack by a nucleophile.
[0092] The term "blocking group" refers to a group that transiently masks the reactivity of a functional group. The functional group can be subsequently unmasked by removal of the blocking group.
[0093] The terms "boronating agents", "sulfur electrophiles", "selenium electrophiles" refer to compounds that are useful in the modifying step used to introduce BH3, S, and Se groups, respectively, for modification at the phosphorus atom.
[0094] The term "moiety" refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.
[0095] The term "solid support" refers to any support which enables synthetic mass production of nucleic acids and can be reutilized at need. As used herein, the term refers to a polymer, that is insoluble in the media employed in the reaction steps performed to synthesize nucleic acids, and is derivatized to comprise reactive groups.
[0096] The term "linking moiety" refers to any moiety optionally positioned between the terminal nucleoside and the solid support or between the terminal nucleoside and another nucleoside, nucleotide, or nucleic acid.
[0097] A "DNA molecule" refers to the polymeric form of deoxyribonucleotides (adenine, guanine, thymine, or cytosine) in its either single stranded form or a double-stranded helix. This term refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms.
Thus, this term includes double-stranded DNA found, inter alia, in linear DNA
molecules (e.g., restriction fragments), viruses, plasmids, and chromosomes. In discussing the structure of particular double-stranded DNA molecules, sequences can be described herein according to the normal convention of giving only the sequence in the 5' to 3' direction along the non-transcribed strand of DNA
(i.e., the strand having a sequence homologous to the mRNA).
[0098] A DNA "coding sequence" or "coding region" is a double-stranded DNA
sequence which is transcribed and translated into a polypeptide in vivo when placed under the control of appropriate expression control sequences. The boundaries of the coding sequence (the "open reading frame" or "ORF") are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxyl) terminus. A coding sequence can include, but is not limited to, prokaryotic sequences, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic (e.g., mammalian) DNA, and synthetic DNA
sequences. A polyadenylation signal and transcription termination sequence is, usually, be located 3' to the coding sequence. The term "non-coding sequence" or "non-coding region" refers to regions of a polynucleotide sequence that are not translated into amino acids (e.g. 5' and 3' un-translated regions).
[0099] The term "reading frame" refers to one of the six possible reading frames, three in each direction, of the double stranded DNA molecule. The reading frame that is used determines which codons are used to encode amino acids within the coding sequence of a DNA molecule.
[00100] As used herein, an "antisense" nucleic acid molecule comprises a nucleotide sequence which is complementary to a "sense" nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule, complementary to an mRNA sequence or complementary to the coding strand of a gene. Accordingly, an antisense nucleic acid molecule can hydrogen bond to a sense nucleic acid molecule.
[00101] The term "base pair" or ("bp"): a partnership of adenine (A) with thymine (T), or of cytosine (C) with guanine (G) in a double stranded DNA molecule. In RNA, uracil (U) is substituted for thymine.
[00102] As used herein a "codon" refers to the three nucleotides which, when transcribed and translated, encode a single amino acid residue; or in the case of UUA, UGA or UAG encode a termination signal.
Codons encoding amino acids are well known in the art and are provided for convenience herein in Table 1.
Table 1 Codon Usage Table Codon Amino acid AA Abbr. Codon Amino acid AA Abbr.
Lr U Phenylalanine Phe F UCU Serine Ser S
UUC Phenylalanine Phe F UCC Serine Ser S
UUA Leucine Leu L UCA Serine Ser S
UUG Leucine Leu L UCG Serine Ser S

Codon Amino acid AA Abbr. Codon Amino acid AA Abbr.
CUU Leucine Leu L CCU Proline Pro P
CUC Leucine Leu L CCC Proline Pro P
CUA Leucine Leu L CCA Proline Pro P
CUG Leucine Leu L CCG Proline Pro P
AUU Isoleucine Ile I ACU Threonine Thr T
AUC Isoleucine Ile I ACC Threonine Thr T
AUA Isoleucine Ile I ACA Threonine Thr T
AUG Methionine Met M ACH Threonine Thr T
GUU Valine Val V GCU Alanine Ala A
GUC Valine Val V GCC Alanine Ala A
GUA Valine Val V GCA Alanine Ala A
GUG Valine Val V GCG Alanine Ala A
UAU Tyrosine Tyr Y UGU Cysteine Cys C
UAC Tyrosine Tyr Y UGC Cysteine Cys C
UUA Stop UGA Stop UAG Stop UGG Tryptophan Trp W
CAU Histidine His H CGU Arginine Arg R
CAC Histidine His H CGC Arginine Arg R
CAA Glutamine Gln Q CGA Arginine Arg R
CAG Glutamine Gln Q CGG Arginine Arg R
AAU As ara ine Asn N AGU Serine Ser S
AAC As ara ine Asn N AGC Serine Ser S
AAA Lysine Lys K AGA Arginine Arg R
AAG Lysine Lys K AGG Arginine Arg R
GAU Aspartate Asp D GGU Glycine Gly G
GAC As artate Asp D GGC Glycine Gly G
GAA Glutamate Glu E GGA Glycine Gly G
GAG Glutamate Glu E GGG Glycine Gly G
[00103] As used herein, a "wobble position" refers to the third position of a codon. Mutations in a DNA
molecule within the wobble position of a codon, in some embodiments, result in silent or conservative mutations at the amino acid level. For example, there are four codons that encode Glycine, i.e., GGU, GGC, GGA and GGG, thus mutation of any wobble position nucleotide, to any other nucleotide, does not result in a change at the amino acid level of the encoded protein and, therefore, is a silent substitution.
[00104] Accordingly a "silent substitution" or "silent mutation" is one in which a nucleotide within a codon is modified, but does not result in a change in the amino acid residue encoded by the codon. Examples include mutations in the third position of a codon, as well in the first position of certain codons such as in the codon "CGG" which, when mutated to AGG, still encodes Arg.
[00105] The terms "gene," "recombinant gene" and "gene construct" as used herein, refer to a DNA
molecule, or portion of a DNA molecule, that encodes a protein or a portion thereof. The DNA molecule can contain an open reading frame encoding the protein (as exon sequences) and can further include intron sequences. The term "intron" as used herein, refers to a DNA sequence present in a given gene which is not translated into protein and is found in some, but not all cases, between exons. It can be desirable for the gene to be operably linked to, (or it can comprise), one or more promoters, enhancers, repressors and/or other regulatory sequences to modulate the activity or expression of the gene, as is well known in the art.
[00106] As used herein, a "complementary DNA" or "cDNA" includes recombinant polynucleotides synthesized by reverse transcription of mRNA and from which intervening sequences (introns) have been removed.
[00107] "Homology" or "identity" or "similarity" refers to sequence similarity between between two nucleic acid molecules. Homology and identity can each be determined by comparing a position in each sequence which can be aligned for purposes of comparison. When an equivalent position in the compared sequences is occupied by the same base, then the molecules are identical at that position;
when the equivalent site occupied by the same or a similar nucleic acid residue (e.g., similar in steric and/or electronic nature), then the molecules can be referred to as homologous (similar) at that position.
Expression as a percentage of homology/similarity or identity refers to a function of the number of identical or similar nucleic acids at positions shared by the compared sequences. A sequence which is "unrelated" or "non-homologous" shares less than 40% identity, less than 35% identity, less than 30% identity, or less than 25% identity with a sequence described herein. In comparing two sequences, the absence of residues (amino acids or nucleic acids) or presence of extra residues also decreases the identity and homology/similarity.
[00108] The term "homology" describes a mathematically based comparison of sequence similarities which is used to identify genes with similar functions or motifs. The nucleic acid sequences described herein can be used as a "query sequence" to perform a search against public databases, for example, to identify other family members, related sequences or homologs. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to nucleic acid molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and BLAST) can be used (See www.ncbi.nlm.nih.gov).
[00109] As used herein, "identity" means the percentage of identical nucleotide residues at corresponding positions in two or more sequences when the sequences are aligned to maximize sequence matching, i.e., taking into account gaps and insertions. Identity can be readily calculated by known methods, including but not limited to those described in (Computational Molecular Biology, Lesk, A.
M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A.
M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988). Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in publicly available computer programs.
Computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FASTA (Altschul, S. F. et al., J. Molec. Biol. 215: 403-410 (1990) and Altschul et al. Nuc.
Acids Res. 25: 3389-3402 (1997)).
The BLAST X program is publicly available from NCBI and other sources (BLAST
Manual, Altschul, S., et al., NCBI NLM NIH Bethesda, Md. 20894; Altschul, S., et al., J. Mol. Biol.
215: 403-410 (1990). The well-known Smith Waterman algorithm can also be used to determine identity.
[00110] A "heterologous" region of a DNA sequence is an identifiable segment of DNA within a larger DNA
sequence that is not found in association with the larger sequence in nature.
Thus, when the heterologous region encodes a mammalian gene, the gene can usually be flanked by DNA that does not flank the mammalian genomic DNA in the genome of the source organism. Another example of a heterologous coding sequence is a sequence where the coding sequence itself is not found in nature (e.g., a cDNA where the genomic coding sequence contains introns or synthetic sequences having codons or motifs different than the unmodified gene). Allelic variations or naturally-occurring mutational events do not give rise to a heterologous region of DNA as defined herein.
[00111] The term "transition mutations" refers to base changes in a DNA
sequence in which a pyrimidine (cytidine (C) or thymidine (T) is replaced by another pyrimidine, or a purine (adenosine (A) or guanosine (G) is replaced by another purine.
[00112] The term "transversion mutations" refers to base changes in a DNA
sequence in which a pyrimidine (cytidine (C) or thymidine (T) is replaced by a purine (adenosine (A) or guanosine (G), or a purine is replaced by a pyrimidine.
Nucleic acid prodrugs comprising a chiral X-phosphonate moiety
[00113] The general principles of prodrug design are outlined by Bundgard (Design and Application of Prodrugs. In a Textbook of Drug Design and Development; Krogsgaard-Larsen, P., Bundgard, H., Eds.;
Harwood: Reading, UK, 1991).
[00114] One strategy to improve the pharmaceutical properties of molecules with desirable biological activity but poor pharmaceutical properties is to administer the molecule of interest as a prodrug derivative. These prodrugs can exhibit one or more of the properties of increased oral bioavailability, increased cell permeability, increased water solubility, reduced first-pass effect, increased stability, active transport by intestinal transporters, or avoidance of efflux transporters, when compared to the parent molecule.
[00115] Oligonucleotides have several pharmaceutical properties which can be improved through the application of prodrug strategies. In particular, oligonucleotides are rapidly degraded by nucleases and exhibit poor cellular uptake through the cytoplasmic cell membrane (Poijarvi-Virta et al., Curr. Med. Chem.
(2006), 13(28);3441-65; Wagner et al., Med. Res. Rev. (2000), 20(6):417-51;
Peyrottes et al., Mini Rev.
Med. Chem. (2004), 4(4):395-408; Gosselin et al., (1996), 43(1):196-208;
Bologna et al., (2002), Antisense & Nucleic Acid Drug Development 12:33-41). In one example, Vives et al., (Nucleic Acids Research (1999), 27(20):4071-76) found that tert-butyl SATE pro-oligonucleotides displayed markedly increased cellular penetration compared to the parent oligonucleotide.
[00116] In some embodiments, the prodrug moiety is removed selectively by esterases, nucleases or a cytochrome P450 enzyme, including but not limited to those listed below.

Family Gene Family Cent CYP1 CYPIAI, CYPIA2, CYPIBI CYP17: CYP17A1 CYP2A6, CYP2A7, CYP2A13, CYP2B6, CYP19 !CYP19A1 CYP2C8, CYP2C9, CYP2C 18, CYP2C 19, CYP2 CYP2D6, CYP2E1, CYP2F1, CYP2J2, CYP20 : CYP20A1 CYP21 : CYP21A2 CYP3 CYP3A4, CYP3A5, CYP3A7, CYP3A43 CYP4A11, CYP4A22, CYP4B 1, CYP4F2, ........ ..... ......... .........
......... ......... ......... .........
CYP4 CYP4F3, CYP4F8, CYP4F11, CYP4F 12 CYP26 CYP26A1 CYP26B 1 CYP26C1 ................... CYP4F22, CYP4V2, CYP4X1, CYP4Z1 ... CYP27A 1 (bile acid biosynthesis), CYP27B 1 CYP5 CYPSAI CYP27 (vitamin D3 1-alpha hydroxylase, activates vitamin D3), CYP27C1 (unknown function) CYP7 CYP7A1, CYP7B 1 CYP8 CYP8A1 (prostacyclin synthase), CYP8B 1 (bile acid biosynthesis) CYP46 CYP46A1 CYP11 CYP11A1 CYP11B1 CYP11B2 CYP51: CYP51A1 (lanosterol 14-alpha demethylase) ...............................................................................
................................................
:..............................................................................
................................................
[00117] In some embodiments, the prodrug is removed when the prooligonucleotide has not yet been transported through the cell membrane. In other embodiments, the prodrug is removed from the pro-oligonucleotide only after it is transported through the cell membrane.
Alternatively, the prodrug is removed only after it is transported into an organelle within the cell. In some embodiments, the prodrug moiety is removed through a non-enzymatic removal including but not limited to the spontaneous reduction inside the cell.
[00118] Described herein are prodrugs of a nucleic acid comprisng a modification of a chiral X-phosphonate, wherein the modification improves one or more physicochemical, pharmacokinetic or pharmacodynamic property of the nucleic acid. A prodrug moiety is connected to an oxygen or sulfur atom which is connected to the phosphorus atom of a phosphonate or phosphothiorate group of the nucleotide. The prodrug moiety includes but is not limited to S-acyl-2-thioethyl, acyloxy, thioacyloxy, 2-carboalkoxyethyl, disulfide, thiaminal, and enol ester derivatives.
[00119] In one embodiment, the prodrug moiety is an S-acyl-2-thioethyl moiety having the following structure:

/'~0"-"/S Y R11 O
wherein R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl. In some embodiments, R11 is methyl, ethyl or cyclopropyl.
[00120] In other embodiments, the prodrug moiety is an acyloxy moiety having following structure:

\

wherein R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl, and R12 is hydrogen or alkyl. In some embodiments, R11 is methyl and R12 is hydrogen.
[00121] Alternatively, the prodrug moiety is a thioacyloxy moiety having the following structure:

A'S "KO~R

wherein R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl, and R12 is hydrogen or alkyl. In some embodiments, R11 is methyl and R12 is hydrogen.
[00122] The invention also provides a prodrug 2-carboalkoxyethyl moeity having one of the following structures:

/,0 or 0 wherein R10 is an alkyl group having 1 to 4 carbon atoms. In some embodiments, R10 is methyl or ethyl.
[00123] In yet other embodiments, the prodrug moiety is a disulfide moiety having the following structure:

/SSRll wherein R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl. In some embodiments, R11 is methyl, ethyl or benzyl.
[00124] In further embodiments, the prodrug moiety is a thioacetal moiety having the following structure:

S 0, O ~'T R11 O
wherein R10 is an alkyl group having 1 to 4 carbon atoms and R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl. In some embodimenst, R10 is methyl and R11 is methyl or phenyl.
[00125] The invention also provides enol ester prodrug moieties having one of the following structures: 0 ~r R11 O R11 0 or 0 C4 enol ester C3 enol ester wherein R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl. In some embodiments, the C3- enol ester prodrug moiety or the C4 enol ester prodrug moiety is in the cis form. In some embodiments of the C3- enol ester prodrug moiety or the C4 enol ester prodrug moiety, R11 is methyl, ethyl or phenyl.
[00126] In one embodiment, the prodrug moiety is a trialkylammoniumethyl moiety having one of the following structures:

,0,,,-,,@.Me Me N, N"
I Me I Me Me , or Me
[00127] In one embodiment, the prodrug moiety is a alkylhydroxamate moiety having one of the following structures:
QMe QMe ,e ,e 0 , or 0
[00128] In one embodiment, the prodrug moiety is a acylhydroxamate moiety having one of the following structures:

4 Me 4 Me V,Me 0or 0[00129] One embodiment provides a nucleic acid prodrug having the following structure:

R1 O Ba X" 'O
[o"o_a]
~R2 n Formula 1 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -P(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na", Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
at least one instance of X is -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, R12 O R12 O /,0,,,,/S R11 R11 \O_KO_I__R 11 \S-O-_~-R 11 O O
0 OYR11 /~S,-,,,OYR11 S~R11 O O O
S)~ R11 O~R11 )~ R11 \11 S O, Rio S"/~. O, \'0""_Y0'Rjo O R
R10 ~ ~S. ~ 11 / I~

S O R11 0 R11 0._"~@.Me ~r ~r I,Me 0 0 , Me , QMe QMe '\O(N .Me ,S N.Me I ~Me Me O O
Q Me Q Me /O /~ N~ iS N Z R15R16 O
\ I~ Me \/~ Me ~S-S-RW

0 0 q O or R15R16 R1s Z ~ S-S{-/) q W.

R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
Rio is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl;
Z is S or O;
g is 0, 1, or 3;
w is 1, 2, 3, 4, 5, or 6;
R15 and R16 are independently hydrogen or methyl;
R17 is selected from alkyl, aryl or a CH2CH=CH2;

-N O I-N I-N I-N'NMe Rig is selected from N(CH3)2, , and I-N

McO2C ;and n is an integer of 1 to about 200.
[00130] In one aspect the invention provides a nucleic acid prodrug having the following structure:

Rl Ba O Ba R2 n Formula 1 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -OP(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+i, Li+', or K+i;
Y2 is 0, NRd, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR, wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

R12 O R12 O ,0,,,,/S Ri l -O-O~R "'S O~R
each instance of X is 11 11 O
S O, Rio ,,O\ O, R11 (Rio ""T /SSRll O
~r O R11 ,O,,,--,,!.Me ~/s0Rll ~r I,Me O O Me 9Me We ,_N.Me ,,S N.Me Me e, 0 , 0 Q Me Q Me ,S
e 0 or 0 R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
R10 is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl; and n is an integer of 1 to about 200.
[00131] In one aspect the invention provides a nucleic acid prodrug having the following structure:
R

x 0 FF OW n Fomnala 2 wherein R' is -OH, -SH, -NR dRd, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -OP(O)(Re)2, -HP(O)(Re), -OR' or -SR';
Y' is 0, NRd, S, or Se;
Ra is a blocking group;
Re is a blocking group;
each instance of Rd is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(Re)2, or -HP(O)(Re);
each instance of Re is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+i, Li+', or K+i;
Y2 is 0, NRd, or S;

each instance of R2 is independently hydrogen, -OH, -SH, -NR dRd, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y'-, alkenyl-Y'-, alkynyl-Y'-, aryl-Y'-, heteroaryl-Y'-, -ORb or -SR , wherein Rb is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

R12 O R12 0 0S Ri l each instance of X is 'OOR 11 R S 0 11 0 \.-S 0, ,S IIO,R1o ,O\ O,R10 / 0 R11 ~
/SSRll 0 AIS O
"T ~r R11 O R11 I-01"-.~!.Me Y--Me O O Me 9Me QMe ~.Me O II N.Me lS N.Me I ~~
Mee O O
Q Me Q Me ,e S
e 0 or 0 R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
R10 is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl; and n is an integer of 1 to about 200.
[00132] A further embodiment provides the nucleic acid prodrug of Formula 1 or Formula 2, wherein each X
moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-~2II 0 11~S ROO

SCH2CH2OH, -OCH2CH2CO2H 'OOR
, S R11 R11 uR11 /-"S,-",OYR11 AIS SyR11 /--O S)~ R11 /-IS O)~ R11 0)~ R11 \S"-"--YO'R1o S O, R1o 'O R11 'S R11 R11 ~O ' Me ~.Me T-Me-Me 0 , Me , Me 9Me QMe 4 Me ,O,N,Me , Me \,O,,,,~N, Me O O O
Me /Z R15R16 O- Z `R1~5R16 R18 Me 1' S-S R17 `' '~~" `S-Si-/q 11 ~
O O or q w ;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
Rio is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl;
Z is S or O;
g is 0, 1, or 3;
w is 1, 2, 3, 4, 5, or 6;
R15 and R16 are independently hydrogen or methyl;

R17 is selected from alkyl, aryl or a CH2CH=CH2; and -N O I-N I-N I-N NMe Rig is selected from N(CH3)2, , and I-N

McO2C
[00133] In some embodiments, n is an integer of 1 to about 50; 1 to about 40;
1 to about 30; 1 to about 25; 1 to about 20; 1 to about 15; or 1 to about 10.
[00134] One embodiment provides a non-racemic pro-oligonucleotide wherein the pro-oligonucleotide is an analog of 2-5A, having a structure of the following formula:

X

HO
x 10 O

HO

x .O
_Y_2~
HO OH
Formula A3-1 wherein X is any of the prodrug moieties descibed herein.
[00135] In some embodiments, the non-racemic pro-oligonucleotide is a 2-5A
analog having the following structure:

~" A
R11yS
O Q H
R111 S'~-- 0 A
OL.

RiS'~' P~~yA
1-f HO OH
Formula A3-2 wherein R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl.
[00136] In one embodiment, non-racemic pro-oligonucleotide is a 2-5A analog having the following structure:

JAS--_o O
o -YSJ' A
O O HO
a '0 A
HO
V ~10A
' O
H H
Formula A3-3 EXEMPLARY METHODS OF SYNTHESIS
General discussion of the methods of synthesis of nucleic acid prodrugs comprising a chiral X-phosphonate moiety [00137] The methods described herein provide for an efficient synthesis of phosphorus atom-modified nucleic acid prodrugs wherein the stereochemical configuration at a phosphorus atom is controlled, thus producing a stereodefined oligonucleotide. While the exemplary methods of synthesis described herein provide for a 3'-5' nucleotide likage, the 2'-5' nucleotide linkage is also contemplated.
[00138] The pro-oligonucleotides of the invention may be synthesized by modifying either a chiral phosphorothioate or chiral H-phosphonate of a nucleotide or nucleic acid.
[00139] A S-acyl- 2-thioethyl pronucleotide may be synthesized from a nucleic acid or nucleotide comprising a chiral H-phosphonate as shown in the following scheme:
Scheme 1 R 0 Ba HO-S1Rõ R, Ba IZ NCS O
R O ?2 H` iO Rõ'1S'\_0. 'O
C/P"õ=C Ba per o Ba RZ RZ

_0-"S R11 [00140] In some embodiments, R1 is -OP(O)(Re)2, wherein Re is 0 . The chiral H-phosphonate is treated with N-chlorosuccinimde and then reacted with S-acyl- 2-thioethyl alcohol to produce a S-acyl- 2-thioethyl prodrug. Protecting groups present at R', R2, and/or R3 may be subsequently removed.
[00141] An acyloxy nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral H-phosphonate as shown in the following scheme:

Scheme 2 R1 O Ba R1 Ba RZ RZ
H~ 000 NCS R11 OOH R11~n O, ~O
[o_'1-11,,0 Ba O ~0~,,,,,0 Ba O O
RZ n RZ

[00142] The chiral H-phosphonate is treated with N-chlorosuccinimide and then reacted with a hydroxymethyl acetate compound to produce an acyloxy prodrug. Protecting groups present at R1, R2, and/or R3 may be subsequently removed.
[00143] A thioacyloxy nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral phosphorothioate as shown in the following scheme:
Scheme 3 R1 O Ba R1 Ba RZ
RZ Q R11\/]/_D\S\
-S, 0 O~PO Ba R11/p\O^CI 0~ O Ba O

RZ n RZ n [00144] The chiral phosphorothioate is treated with a chloromethyl acyloxy compound to produce an acyloxy prodrug. Protecting groups present at R1, R2, and/or R3 may be subsequently removed.
[00145] A 2-carboalkoxyethyl nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral phosphorothioate as shown in the following scheme:
Scheme 4 R1 O Ba R1 Ba MeO Rz Me0 S\ i0 S\ i0 O ",O O Ba O O~PBa O
R2 n R2 n [00146] The deprotonated chiral phosphorothioate is reacted with an alkyl acrylate to produce a 2-carboalkoxyethyl pronucleotide. Protecting groups present at R1, R2, and/or R3 may be subsequently removed.
[00147] A disulfide nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral phosphorothioate as shown in the following scheme:

Scheme 5 R1 O Ba R1 Ba -sue ~o R11S-SR11 R11. S O
S> O S/ o Ba O\, O O Ba R2 n R2 n The deprotonated chiral phosphorothioate is reacted with a dialkyl sulfide to produce an alkyl disulfide pronucleotide. Protecting groups present at R1, R2, and/or R3 may be subsequently removed.
[00148] A thioacetal nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral phosphorothioate as shown in the following scheme:
Scheme 6 R1 O Ba R1 Ba R10pply -S-, #00 R100 0 R11r R2 O S '.O
[OOBa] O R
O R1o0 O O O Ba R2 n R2 n [00149] A 1,1,-dialkyoxy 3-acyloxy propane is reacted with trimethylsilyltriflate and the deprotonated chiral phosphorothioate is then added to the reaction mixture to produce an thioacetal pronucleotide. Protecting groups present at R1, R2, and/or R3 may be subsequently removed.
[00150] A C3 enol ester nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral phosphorothioate as shown in the following scheme:
Scheme 7 R1 Ba R1 Ba R2 CI~~O~R11 o R11 o 2 Ba 0 O Ba R2 n R2 n [00151] The deprotonated chiral phosphorothioate is reacted with an (E) -3-chloro- 1-acyloxy-prop-l-ene compound to produce the C3 enol ester nucleic acid prodrug. Protecting groups present at R1, R2, and/or R3 may be subsequently removed.
[00152] A C4 enol ester nucleic acid prodrug may be synthesized from a nucleic acid or nucleotide comprising a chiral phosphorothioate as shown in the following scheme:

Scheme 8 ,Ba Rl O R1 O Ba CIC-yR11 O R11~ Rz -S~ O C~S' 'O
[o'o_a] O'=õO Ba O
Rz n Rz n [00153] The deprotonated chiral phosphorothioate is reacted with an (E) -3-chloro- 1-acyloxy-but-l-ene compound to produce the C3 enol ester nucleic acid prodrug. Protecting groups present at R', R2, and/or R3 may be subsequently removed.
[00154] In some embodiments the nucleic acid comprising a chiral phosphorothioate or chiral H-phosphonate is synthesized as described herein. In other embodiments, other methods of synthesis may be used to provide the nucleic acid comprising a chiral phosphorothioate or chiral H-phosphonate.
Scheme 9. Synthesis of chiral phosphorothioate precursors of the prodrug oligonucleotides of the invention. (Route A) R õ Ba R1 0 Ba RBa R 0Ba activation chiral reagent Rz z z O Rz 0H' O Z' CR P\ R R A WZPO Ba L/ L WZ W1 W nGl Formula 2 II G1,C4 condensatio G4"-G3GZ Rz G2 G3 J~ O capping III
[Aa]
W1r GG z G4'G3 R V
HO O Ba HO Ba V OA R' ~_ Ba z RZ "~~R2 =P~O R
Ba HOB 0 remove chiral auxilliary Jp.O A Wp O rR
[o_a] A WZ 'O Ba chain W X11õG
elongation }~ z 1G cycle 4 sG3G Gz 2 n 4 'G3 Rz m J~ O
VIII OA O A W~0 Ba z IV WõG, remove other protecting groups, R G2 rR cleave from solid support G3 v O Ba G4 R1 Ba VI O4 Rz Rz " ' 1 [>Ba] deblock 5-eWO Ba modiing WnGl Ga G3Gz V

Rz n VII OA

Formula 1' [00155] The reaction of a molecule comprising an achiral H-phosphonate moiety of Formula 2 with a nucleoside comprising nucleophilic moiety of Formula IV results in the formation of a condensed intermediate (V); which is converted to a nucleic acid comprising a chiral X-phosphonate moiety which can be further modified to produce the prodrug oligonucleotide of Formula I
comprising a chiral X-phosphonate moiety. The synthesis of the condensed intermediate comprises the steps of (a) activation of the compound of Formula 2 with a condensing agent to form intermediate II, (b) reaction with a chiral reagent to form intermediate III, followed by (c) reaction with the compound of Formula IV.
[00156] The condensed intermediate may be converted to a nucleic acid comprising a chiral X phosphonate moiety of Formula 1' by capping the chiral auxiliary with a moiety A, which is an acyl, aryl, alkyl, aralkyl, or silyl moiety, and modifying the phosphorus to introduce J, which is S, Se, or BH3, thus producing a compound of Formula VII.
[00157] The compound of Formula VII may be converted to the compound of Formula 1', where X is S, Se, or BH3, and n is 1, by cleaving the chiral reagent, and deblocking blocking groups and cleaving from solid support if desired. Alternatively the compound of Formula VII is subjected to chain elongation by deblocking the 5' terminus, and repeating coupling steps to produce a condensed intermediate as before. The steps of capping, modifying, deblocking, and chain elongation are repeated until the desired n is achieved. At that point, the chiral reagents at each phosphonate are cleaved, the remaining blocking groups are cleaved, including cleaving from a solid support, if desired, to produce the compound of Formula 1', where X is S, Se, or BH3, and n is greater than or equal to 2 and less than about 200. The compound of Formula 1', where X is S is then converted by the methods described herein to form the pro-oligonucleotide compound of Formula 1.
Modifying agents used to introduce S, Se, or BH2 at chiral phosphorus of the condensed intermediate V in Route A.
[00158] In some embodiments, the modifying agent is a sulfur electrophile, selenium electrophile, or boronating agent. In some embodiments, the sulfur electrophile is a compound having one of the following formulas:
Ss (Formula B), Z10-S-S-Z", or Z10-S-X-Z"
wherein Z10 and Z" are independently alkyl, aminoalkyl, cycloalkyl, heterocyclic, cycloalkylalkyl, heterocycloalkyl, aryl, heteroaryl, alkyloxy, aryloxy, heteroaryloxy, acyl, amide, imide, or thiocarbonyl, or Z10 and Z" are taken together to form a 3 to 8 membered alicyclic or heterocyclic ring, which may be substituted or unsubstituted; X is 502, 0, or NRf; and Rf is hydrogen, alkyl, alkenyl, alkynyl, or aryl. In other embodiments, the sulfur electrophile is a compound of Formula B, C, D, E, or F:
N H2 OEt O O
A S HN-<
S N S~ I S S-S
Ss -~ O O 1NH
Formula B Formula C Formula D Formula E Formula F
[00159] In other embodiments, the sulfur electrophile is Formula F, Formula E
or Formula B.
[00160] In some embodiments, the selenium electrophile is a compound having one of the following formulas:
Se (Formula G), Z10-Se-Se-Z", or Z10-Se-X-Z"

wherein Z10 and Z" are independently alkyl, aminoalkyl, cycloalkyl, heterocyclic, cycloalkylalkyl, heterocycloalkyl, aryl, heteroaryl, alkyloxy, aryloxy, heteroaryloxy, acyl, amide, imide, or thiocarbonyl, or Z10 and Z" are taken together to form a 3 to 8 membered alicyclic or heterocyclic ring, which may be substituted or unsubstituted; X is SO2, S, 0, or NRf; and Rf is hydrogen, alkyl, alkenyl, alkynyl, or aryl.
[00161] In other embodiments, the selenium electrophile is a compound of Formula G, H, I, J, K, or L.
S
Se I ~Se Ph Ph-P-Ph Se-~,e NCB Se-Se CN
Se KSeCN Ph Ph Formula G Formula H Formula I Formula J Formula K Formula L
[00162] In some embodiments, the selenium electrophile is Formula G or Formula L.
[00163] In some embodiments, the boronating agent is borane-N,N-diisopropylethylamine (BH3=DIPEA), borane-pyridine (BH3=Py), borane-2-chloropyridine (BH3=CPy), borane-aniline (BH3=An), borane-tetrahydrofuran (BH3=THF), or borane-dimethylsulfide (BH3=Me2S), aniline-cyanoborane, triphenylphosphine-carboalkoxyboranes.
[00164] In other embodiments, the boronating agent is borane-N,N-diisopropylethylamine (BH3=DIPEA), borane-2-chloropyridine (BH3=CPy), borane-tetrahydrofuran (BH3=THF), or borane-dimethylsulfide (BH3=Me2S).
Scheme 10.
Synthesis of a precursor of a prodrug oligonucleotide via chiral H-phosphonates (Route B).

Ra a z activation R1~a chiral reagent R~a O Rz H a '.0- R CR Rz Rz 'PCO
i W2 o H- z' A
W WinG9 Formula 2 II G G4 condensation G4'G3G2 Rz Gz G3 H, O
III ~~ [o'oa]
X acidify (deblock T-end) Rz V

HO, n_ a ~4a rl~' chain elongation O, ~O modifying O1.O cycle X O O a [H'oa]
HOa Rz n Rz m +, Rz XII O~ IX O A HER O a remove other protecting groups, H W1 ""G1 cleave from solid support G4 'G3 Gz R2 R a chiral auxilliary H, .O
is removed O"'O
during acidification Rz 0.!O A R2 X~ O a 11 1 Og XI
G4_ ~''Gt Rz n G3 Gz OH
Formula 1 [00165] In another embodiment, described in Scheme 10, an achiral H-phosphonate of Formula 2 is treated with a condensing reagent to form an intermediate of structure II. The intermediate of structure II is not isolated and is treated in the same pot with a chiral reagent to form a chiral intermediate of structure III. The intermediate of structure III is not isolated and undergoes a reaction in the same pot with a nucleoside or modified nucleoside of structure IX to provide a chiral phosphite compound of structure X. In some embodiments, structure X is extracted into a solvent to separate it from side products, impurities, and/or reagents. In other embodiments, when the method is performed via solid phase synthesis, the solid support comprising the compound of structure X is filtered away from side products, impurities, and/or reagents.
The compound of structure X is treated with an acid to remove the blocking group at the 5'-end of the growing nucleic acid chain (structure XI). The acidification step also removes the chiral auxiliary ligand to provide a chiral H-phosphonate IX. The 5'-deblocked intermediate is optionally allowed to re-enter the chain elongation cycle to form a condensed intermediate containing a blocked 5'-end, which is then acidified to remove the 5'-end blocking group and chiral auxiliary ligand.
[00166] When the desired chain length has been reached, the 5'-deprotected intermediate undergoes a modifying step to introduce a moiety X bonded to each of the phosphorus atoms to provide a compound of structure XII. The modified intermediate is deblocked by removal of remaining protecting groups, e.g., nucleobase, modified nucleobase, sugar or modified sugar protecting groups are removed, to provide a nucleic acid of Formula 1. In embodiments where a solid support is used, the phosphorus-atom modified nucleic acid is then cleaved from the solid support. In certain embodiments, the nucleic acids is left attached on the solid support for purification purposes and then cleaved from the solid support following purification.
In one embodiment, the synthesis described in Scheme 10 is useful when the GI
and G2 positions of the chiral auxiliary ligand of Formula A are not hydrogen.
Modification of the compound of Formula IX obtained via Route B to introduce an X- or X'-phosphonate moiety.
[00167] Other methods used to modify the compound of Formula IX obtained via Route B are illustrated in Reaction Schemes 10a and 10b. Phosphonate and phosphite are known to tautomerize and exist in equilibrium. The phosphite tautomer is less stable than the phosphonate tautomer. Equilibrium lies toward the phosphonate tautomer under neutral conditions due to the very strong P=O
bond. Under acidic conditions, the phosphoryl group of the phosphonate becomes reversibly protonated. Cleavage of the P-H
bond in the intermediate occurs slowly to produce the phosphite intermediate.
Structure IX is then modified to form structure XII, using reagents shown in Reaction Schemes I Oa and IOb.

Reaction Scheme 10a.
Modification of the phosphorus center of intermediates synthesized via Route B, using an initial halogenation at phosphorus.
HO-J_ Ba -Wo. 3a R2N' O

R' m amore O

HO 0\Ba HO Ba HO-, 0. Ba HO-, O 6a 2 positive R R' _ halogen [80 R
O; H0. reagent alcohol H F..O O
- -----------------X'R.,O Ba 5 ,.O Ba ~/ 0 O
R m 1~"--R( ! m IX OA D, thiol HO ~, .0 Ba O'P,,O
RS- .O Ba R' m Reaction Scheme 10b.
Modification of phosphorus in intermediates synthesized via Route B, using an initial silylation.
HO-- 0 Ba HO- O Ba O. P. HO O.,PO
[OBa Y O Ba R Rz m R 7R2 HO Ba OA OA R' acylating 0. O
agent R O Ba HO 0Ba HO- O Ba HO Ba aldehyde alkylating Rz m R2 agent '~R2 silylng OA
SiO. O
O..P.O HO O reagegent a H' O O Ba .P"O Ba P-,..0 Ba R2 M R2 R` m boronating HO 0 Ba m -~ agent 0,.0 sulfur Z H,E, O Ba selenium lectrophile electrophild 2 t ~~R M
HO 0 Ba OA
HO Ba O.1P,O
O
Z Se 0 0 Ba O 0 Z --S 0 , 0 Ba 7R2 im Rz M
OA
OA

[00168] In some embodiments, the modifying step is performed by reacting structure IX with a halogenating reagent followed by reacting with a nucleophile. In specific embodiments, the halogenating reagent is CC14, CBr4, C14,, C12, Br2, 12, sulfuryl chloride (S02C12), phosgene, triphosgene, sulfur monochloride, sulfur dichloride, chloramine, CuC12, N-chlorosuccinimide (NCS), N-bromosuccinimide (NBS), or N-iodosuccinimide (NIS). In other specific embodiments, the halogenating reagent is CC14, CBr4, C12, sulfuryl chloride (S02C12), or N-chlorosuccinimide (NCS). In some embodiments, the nucleophile is primary or secondary amines, alcohols, or thiols. In other embodiments, the nucleophile is NRfRfH, RfOH, or RfSH, wherein Rf is hydrogen, alkyl, alkenyl, alkynyl, or aryl, and at least one of Rf of NRfRfH is not hydrogen.
[00169] The modifying step can also be performed by reacting structure IX with a silylating reagent followed by reaction with a sulfur electrophile, a selenium electrophile, a boronating agent, an alkylating agent, an aldehyde, or an acylating agent.
[00170] In specific embodiments, the silylating reagent is chlorotrimethylsilane (TMS-Cl), triisopropylsilylchloride (TIPS-CI), t-butyldimethylsilylchloride (TBDMS-Cl), t-butyldiphenylsilylchloride (TBDPS-Cl), 1,1,1,3,3,3-hexamethyldisilazane (HMDS), N-trimethylsilyldimethylamine (TMSDMA), N-trimethylsilyldiethylamine (TMSDEA), N-trimethylsilylacetamide (TMSA), N,O-bis(trimethylsilyl)acetamide (BSA), or NO-bis(trimethylsilyl)trifluoroacetamide (BSTFA).
[00171] In other specific embodiments, the sulfur electrophile is a compound having one of the following formulas:
Ss (Formula B), Z10-S-S-Z", or Z10-S-X-Z"
wherein Z10 and Z" are independently alkyl, aminoalkyl, cycloalkyl, heterocyclic, cycloalkylalkyl, heterocycloalkyl, aryl, heteroaryl, alkyloxy, aryloxy, heteroaryloxy, acyl, amide, imide, or thiocarbonyl, or Z10 and Z" are taken together to form a 3 to 8 membered alicyclic or heterocyclic ring, which may be substituted or unsubstituted; X is SO2, 0, or NR f; and Rf is hydrogen, alkyl, alkenyl, alkynyl, or aryl. In other embodiments, the sulfur electrophile is a compound of Formula B, C, D, E, or F:
NH2 OEt O O
A S HN-<
N I S S-S
S--( S--( 'O O)-NH
Sa S O
Formula B Formula C Formula D Formula E Formula F
[00172] In other embodiments, the sulfur electrophile is Formula F, Formula E
or Formula B.
[00173] In some embodiments, selenium electrophile is a compound having one of the following formulas:
Se (Formula G) , Z10-Se-Se-Z", or Z10-Se-X-Z"
wherein Z10 and Z" are independently alkyl, aminoalkyl, cycloalkyl, heterocyclic, cycloalkylalkyl, heterocycloalkyl, aryl, heteroaryl, alkyloxy, aryloxy, heteroaryloxy, acyl, amide, imide, or thiocarbonyl, or Z10 and Z" are taken together to form a 3 to 8 membered alicyclic or heterocyclic ring, which may be substituted or unsubstituted; X is SO2, S, 0, or NRf; and Rf is hydrogen, alkyl, alkenyl, alkynyl, or aryl.
[00174] In other embodiments, the selenium electrophile is a compound of Formula G, H, I, J, K, or L.
Se I 'Se Ph Ph-P-Ph Se-~,e NCB Se-Se CN
Se KSeCN Ph Ph Formula G Formula H Formula I Formula J Formula K Formula L
[00175] In some embodiments, the selenium electrophile is Formula G or Formula L.
[00176] In some embodiments, the boronating agent is borane-N,N-diisopropylethylamine (BH3=DIPEA), borane-pyridine (BH3=Py), borane-2-chloropyridine (BH3=CPy), borane-aniline (BH3=An), borane-tetrahydrofuran (BH3=THF), or borane-dimethylsulfide (BH3=Me2S), aniline-cyanoborane, triphenylphosphine-carboalkoxyboranes. In other embodiments, the boronating agent is borane-N,N-diisopropylethylamine (BH3=DIPEA), borane-2-chloropyridine (BH3=CPy), borane-tetrahydrofuran (BH3=THF), or borane-dimethylsulfide (BH3=Me2S).
[00177] In other embodiments, the alkylating agent is an alkyl halide, alkenyl halide, alkynyl halide, alkyl sulfonate, alkenyl sulfonate, or alkynyl sulfonate.
[00178] In other embodiments, the aldehyde is (para)-formaldehyde, alkyl aldehyde, alkenyl aldehyde, alkynyl aldehyde, or aryl aldehyde.
[00179] In yet other embodiments, the acylating agent is a compound of Formula M or N:
O O O

G7A, M G7jO'A'G7 Formula M Formula N
[00180] wherein G7 is alkyl, cycloalkyl, heterocyclic, cycloalkylalkyl, heterocycloalkyl, aryl, heteroaryl, alkyloxy, aryloxy, or heteroaryloxy; and M is F, Cl, Br, I, 3-nitro-1,2,4-triazole, imidazole, alkyltriazole, tetrazole, pentafluorobenzene, or 1-hydroxybenzotriazole.

Scheme 11. Synthesis of a chiral dinucleoside phosphorothioate by stereoselective synthesis.
TBDPS a TBDPS a HO ~ HO-1a +
Ph ~
Ph TBDPSO
1.Ac2O
TBDPS O a 2. / TBDPS O a \ I /S
O. OHaO S HDBU
NH 06. < 0 0 O a o -],O a 3. DBU
Ph TBDPSO TBDPSO

[00181] One method of stereoselective dinucleoside phosphorothioate synthesis involves the use of stereochemically pure 3'-phosphoramidites as described by Oka et at, (J. Am.
Chem. Soc. (2003), 125:8307-17). As shown in the Scheme 6a (above), 2-chlorooxazaphospholidine derivatives are allowed to react with a 5'-O-(TBDPS)nucleoside to afford the 3'-O-oxazaphospholidine derivative.
Reaction of a 3'-O-(TBDPS)nucleoside with the 3'-O-oxazaphospholidine derivative in the presence of an activator such as N-(cyanomethyl)pyrrolidine gives the dinucleoside phosphite as a single diastereomer. The dinucleoside phosphite can be converted to the phosphorothioate by a three-step process involving acetylation with acetic anhydride, sulfurization with the Beaucage reagent (3H-1,2-benzodithiol-3-one-1,1-dioxide; Iyer et at, J. Am.
Chem. Soc. (1990), 112:1253-4), and cleavage of the chiral auxiliary with excess DBU. The protected dinucleoside phosphorthioate is then converted to the prodrug by the methods disclosed herein.
[00182] Other methods useful for the synthesis of dinucleoside phosphorthioates include enzymatic methods (Hacia et al. Biochemistry (1994), 33:5367-9; Tang et al. Nucleosides Nucleotides (1995), 14:985-990), methods involving separation of diasteromeric phosphorthioate mixtures prepared by non-stereoselective methods (Zon et al Oligonucleotides and Analogues: A Practical Approach; IRL
Press: London, 1991, pp 87-108) and methods involving stereoselective synthesis of phosphorthioates (Wilk et al. J. Am. Chem. Soc.
2000, 122, 2149-2156; Lu et al, Angew. Chem., Int. Ed. 2000, 39, 4521-4524;
Iyer et al Tetrahedron:Asymmetry 1995, 6, 1051-1054. Iyer et al Tetrahedron Lett. 1998, 39, 2491-2494; Lu et al Tetrahedron 2001, 57, 1677-1687. Stec et al Nucleic Acids Res. 1991, 19, 5883-5888; Stec et al J. Am.
Chem. Soc. 1995, 117, 12019-12029; Uznan'ski et al J. Am. Chem. Soc. 1992, 114, 10197-10202.

Reverse 5' to 3' Nucleic Acid Synthesis [00183] A nucleic acid of Formula 1 comprising a chiral X-phosphonate moiety alternatively is synthesized from the 5' to 3' direction. In embodiments where a solid support is used, the nucleic acid is attached to the solid support through its 5' end of the growing nucleic acid, thereby presenting its 3' group for reaction, including enzymatic reaction (e.g. ligation and polymerization). In some embodiments, this orientation is engineered by preparing nucleoside monomers comprising an achiral H-phosphonate moiety at the 5' position and protected hydroxyl group at the 3' position. In an embodiment, the nucleic acid is synthesized according to Scheme 12. In Scheme 12, -R4 is -ORb as defined above or, in the last cycle of synthesis, is R4, which is equivalent to R' as defined herein.

Scheme 12.
5' to 3' synthesis of a pro-oligonucleotide of Formula 1 comprising a chiral X-phosphonate moiety.
8 O [ra] OA a reagent H-p- O a acts chiral Gz, yg CR ORq RzP-O O a Rz OR4 Rz G3 4 W~ O\
Ir G OR4 Rz H "="O O a I I, fRZ v acidify A W (deblock 3'-end) III, condensation (d + 'O O Ba W, i1 Gi G4 zG3G2 Rz XIV
OA O a OA O a OR4 Rz Rz O> n modifying O~~.-O
X a a chain O H O elongation OA a cycle Rz Rz M Rz O
XVI OH OH 0. H""=O
XIII
remove other protecting groups, O a v cleave from solid support Rz HOa chiral auxilliary O is removed H *~O

ringacidification H P""0y/gga OO R A H GI~Gz' Rz X O a OH
XV
~~R2 G4 G3 Gz G' n OH
Formula 1r [00184] In the embodiment described in Scheme 12, an achiral H-phosphonate of structure Ir is treated with a condensing reagent to form an intermediate of structure IIr.
The intermediate of structure IIr is not isolated and is treated in the same pot with a chiral reagent to form an intermediate of structure IIIr. The intermediate of structure IIIr is not isolated and undergoes a reaction in the same pot with a nucleoside or modified nucleoside of structure XIII to provide a chiral phosphite compound of structure XIV. In some embodiments, structure XIV is extracted into a solvent to separate it from side products, impurities, and/or reagents. In other embodiments, when the method is performed via solid phase synthesis, the solid support comprising the compound of structure XIV is filtered away from side products, impurities, and/or reagents.
The compound of structure XIV is treated with an acid to remove the blocking group at the 3'-end of the growing nucleic acid chain (structure XV). The acidification step also removes the chiral auxiliary ligand to provide a compound of structure XIII. The 3'-deblocked intermediate is optionally allowed to re-enter the chain elongation cycle to form a condensed intermediate containing a blocked 3'-end, which is then acidified to remove the 3'-end blocking group and chiral auxillary ligand. Following at least one round of chain elongation cycle, the 3'-deprotected intermediate undergoes a modifying step to introduce a moiety X bonded to each of the phosphorus atoms to provide a compound of structure XVI. The modified intermediate is deblocked by removal of remaining protecting groups, e.g., nucleobase, modified nucleobase, sugar or modified sugar protecting groups are removed, to provide a nucleic acid of Formula 1. In other embodiments, the nucleoside comprising a 3'-OH moiety is an intermediate from a previous chain elongation cycle as described herein. In yet other embodiments, the nucleoside comprising a 3'-OH moiety is an intermediate obtained from another known nucleic acid synthetic method. After a cycle of synthesis with the first nucleoside, nucleosides, nucleotides, or nucleic acids that contain an unprotected -OH moiety can be used for subsequent elongation cycles. In embodiments where a solid support is used, the phosphorus-atom modified nucleic acid can then be cleaved from the solid support, located at the 5' end. In certain embodiments, the nucleic acids can optionally be left attached on the solid support for purification purposes and then cleaved from the solid support following purification. In one aspect, the synthesis described in Scheme 12 is useful when both of the G1 and G2 position of the chiral auxiliary ligand of Formula A are not hydrogen. The reverse 5' to 3' synthesis can be accomplished using the same starting materials in Scheme 12 in a mechanism analogous to steps in Route A.
Generation of phosphothiotriesters with reversible protecting groups from H-phosphonate [001851 Phosphorothioates can be synthesized in a stereospecific manner from H-phosphonates with retention of configuration at phosphorus atom (J. Org. Chem. 1991, 3861-3869). Also contemplated is the use of this reaction to synthesize phosphorothiotriesters using thiol-containing moiety that also carries bioreversible protecting group, see Scheme 13. Additionally, stereocontrolled solid-phase synthesis of oligonucleoside H-phosphonates has also been reported (Angew. Chem. Int. Ed.
2009, 48, 496-499) and it is contemplated that this method, combined with alkylation during solid support synthesis, to prepare phosphothiotriesters on support.
Scheme 13 NH NH NH
DMTrO N'k, O DMTrO Nl-~ O HO N~O
jNH NH NH
0P''O N~O O ;P 'O NO O P N~O
HO
_1 _1 O R- SO O R SO O

DMTrO DMTrO HO
i)12' ACN: Py (3:2), TBDPSCI and the respective thiol ii) 3% DCA/DCM

Reaction Conditions and Reagents used in the methods of the invention.
Conditions [001861 The steps of reacting a molecule comprising an achiral H-phosphonate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate can occur without isolating any intermediates. In some embodiments, the steps of reacting a molecule comprising an achiral H-phosphonate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate occurs is a one-pot reaction. In an embodiment, a molecule comprising an achiral H-phosphonate moiety, condensing reagent, chiral reagent, and compound comprising a free nucleophilic moiety are added to the reaction mixture at different times. In another embodiment, a molecule comprising an achiral H-phosphonate moiety, condensing reagent, and chiral reagent are present in the same reaction vessel or same pot. In another embodiment, a molecule comprising an achiral H-phosphonate moiety, condensing reagent, chiral reagent, and compound comprising a free nucleophilic moiety are present in the same reaction or same pot. This allows the reaction to be performed without isolation of intermediates and eliminates time-consuming steps, resulting in an economical and efficient synthesis. In specific embodiments, the achiral H-phosphonate, condensing reagent, chiral amino alcohol, 5'-OH nucleoside are present at the same time in a reaction. In a further embodiment, the formation of the chiral intermediate for condensation is formed in situ and is not isolated prior to the condensation reaction. In another embodiment, a molecule comprising an achiral H-phosphonate moiety has been activated by reaction with a condensing reagent, chiral reagent in a different reaction vessel from that used when reacting the chiral intermediate with the compound comprising a free 5'-OH moiety.
Synthesis on solid support [00187] In some embodiments, the synthesis of the nucleic acid is performed in solution. In other embodiments, the synthesis of the nucleic acid is performed on solid phase.
The reactive groups of a solid support may be unprotected or protected. During oligonucleotide synthesis a solid support is treated with various reagents in several synthesis cycles to achieve the stepwise elongation of a growing oligonucleotide chain with individual nucleotide units. The nucleoside unit at the end of the chain which is directly linked to the solid support is termed "the first nucleoside" as used herein. The first nucleoside is bound to the solid support via a linker moiety, i.e. a diradical with covalent bonds to both the polymer of the solid support and the nucleoside. The linker stays intact during the synthesis cycles performed to assemble the oligonucleotide chain and is cleaved after the chain assembly to liberate the oligonucleotide from the support.
[00188] Solid supports for solid-phase nucleic acid synthesis include the supports described in, e.g., US
patents 4,659,774 , 5,141,813, 4,458,066; Caruthers U.S. Pat. Nos. 4,415,732, 4,458,066, 4,500,707, 4,668,777, 4,973,679, and 5,132,418; Andrus et at. U.S. Pat. Nos. 5,047,524, 5,262,530; and Koster U.S. Pat.
Nos. 4,725,677 (reissued as Re34,069). In some embodiments, the solid phase is an organic polymer support. In other embodiments, the solid phase is an inorganic polymer support. In some embodiments, the organic polymer support is polystyrene, aminomethyl polystyrene, a polyethylene glycol-polystyrene graft copolymer, polyacrylamide, polymethacrylate, polyvinylalcohol, highly cross-linked polymer (HCP), or other synthetic polymers, carbohydrates such as cellulose and starch or other polymeric carbohydrates, or other organic polymers and any copolymers, composite materials or combination of the above inorganic or organic materials. In other embodiments, the inorganic polymer support is silica, alumina, controlled polyglass (CPG), which is a silica-gel support, or aminopropyl CPG. Other useful solid supports include fluorous solid supports (see e.g., WO/2005/070859), long chain alkylamine (LCAA) controlled pore glass (CPG) solid supports (see e.g., S. P. Adams, K. S. Kavka, E. J. Wykes, S. B.
Holder and G. R. Galluppi, J.
Am. Chem. Soc., 1983, 105, 661-663; G. R. Gough, M. J. Bruden and P. T.
Gilham, Tetrahedron Lett., 1981, 22, 4177-4180). Membrane supports and polymeric membranes (see e.g. Innovation and Perspectives in Solid Phase Synthesis, Peptides, Proteins and Nucleic Acids, ch 21 pp 157-162, 1994, Ed. Roger Epton and U.S. Pat. No. 4,923,901) are also useful for the synthesis of nucleic acids.
Once formed, a membrane can be chemically functionalized for use in nucleic acid synthesis. In addition to the attachment of a functional group to the membrane, the use of a linker or spacer group attached to the membrane may be used to minimize steric hindrance between the membrane and the synthesized chain.
[00189] Other suitable solid supports include those generally known in the art to be suitable for use in solid phase methodologies, including, for example, glass sold as PrimerTM 200 support, controlled pore glass (CPG), oxalyl-controlled pore glass (see, e.g., Alul, et al., Nucleic Acids Research, 1991, 19, 1527), TentaGel Support-an aminopolyethyleneglycol derivatized support (see, e.g., Wright, et al., Tetrahedron Lett., 1993, 34, 3373), and Poros-a copolymer of polystyrene/divinylbenzene.
[00190] Surface activated polymers have been demonstrated for use in synthesis of natural and modified nucleic acids and proteins on several solid supports mediums. The solid support material can be any polymer suitably uniform in porosity, has sufficient amine content, and sufficiently flexible to undergo any attendant manipulations without losing integrity. Examples of suitable selected materials include nylon, polypropylene, polyester, polytetrafluoroethylene, polystyrene, polycarbonate, and nitrocellulose. Other materials can serve as the solid support, depending on the design of the investigator. In consideration of some designs, for example, a coated metal, in particular gold or platinum can be selected (see e.g., US
publication No. 20010055761). In one embodiment of oligonucleotide synthesis, for example, a nucleoside is anchored to a solid support which is functionalized with hydroxyl or amino residues. Alternatively, the solid support is derivatized to provide an acid labile trialkoxytrityl group, such as a trimethoxytrityl group (TMT). Without being bound by theory, it is expected that the presence of the trialkoxytrityl protecting group will permit initial detritylation under conditions commonly used on DNA
synthesizers. For a faster release of oligonucleotide material in solution with aqueous ammonia, a diglycoate linker is optionally introduced onto the support.
Linking moiety [00191] A linking moiety or linker is optionally used to connect the solid support to the compound comprising a free nucleophilic moiety. Suitable linkers are known such as short molecules which serve to connect a solid support to functional groups (e.g., hydroxyl groups) of initial nucleosides molecules in solid phase synthetic techniques. In some embodiments, the linking moiety is a succinamic acid linker, or a succinate linker (-CO-CH2-CH2-CO-), or an oxalyl linker (-CO-CO-). In other embodiments, the linking moiety and the nucleoside are bonded together through an ester bond. In other embodiments, the linking moiety and the nucleoside are bonded together through an amide bond. In further embodiments, the linking moiety connects the nucleoside to another nucleotide or nucleic acid. Suitable linkers are disclosed in, for example, Oligonucleotides And Analogues A Practical Approach, Ekstein, F. Ed., IRL Press, N.Y., 1991, Chapter 1.

[00192] A linker moiety is used to connect the compound comprising a free nucleophilic moiety to another nucleoside, nucleotide, or nucleic acid. In some embodiments, the linking moiety is a phosphodiester linkage. In other embodiments, the linking moiety is an H-phosphonate moiety.
In yet other embodiments, the linking moiety is an X-phosphonate moiety.
Solvents for synthesis [00193] Synthesis of the nucleic acids is performed in an aprotic organic solvent. In some embodiments, the solvent is acetonitrile, pyridine, tetrahydrofuran, or dichloromethane. In some embodiments, when the aprotic organic solvent is not basic, a base is present in the reacting step. In some embodiments where a base is present, the base is pyridine, quinoline, or N,N-dimethylaniline. Other examples of bases include pyrrolidine, piperidine, N-methyl pyrrolidine, pyridine, quinoline, N,N-dimethylaminopyridine (DMAP), or N,N-dimethylaniline. In some embodiments, the aprotic organic solvent is anhydrous. In other embodiments, the anhydrous aprotic organic solvent is freshly distilled. In some embodiments, the freshly distilled anhydrous aprotic organic solvent is pyridine. In other embodiments, the freshly distilled anhydrous aprotic organic solvent is tetrahydrofuran. In other embodiments, the freshly distilled anhydrous aprotic organic solvent is acetonitrile.
Acidification conditions to remove blocking groups.
[00194] Acidification to remove blocking groups is accomplished by a Bronsted acid or Lewis acid. In some embodiments, acidification is used to remove R' blocking groups. Useful Bronsted acids are carboxylic acids, alkylsulfonic acids, arylsulfonic acids, phosphoric acid and its derivatives, phosphonic acid and its derivatives, alkylphosphonic acids and their derivatives, arylphosphonic acids and their derivatives, phosphinic acid, dialkylphosphinic acids, and diarylphosphinic acids which have a pKa (25 C in water) value of -0.6 (trifluoroacetic acid) to 4.76 (acetic acid) in an organic solvent or water (in the case of 80%
acetic acid). The concentration of the acid (1 to 80%) used in the acidification step depends on the acidity of the acid. Consideration to the acid strength must be taken into account as strong acid conditions will result in depurination/depyrimidination, wherein purinyl or pyrimidinyl bases are cleaved from ribose ring.

R = H, alkyl, aryl R = alkyl, aryl R1, R2 = H, alkyl, aryl R1, R2 = H, alkyl, aryl R1, R2 = H, alkyl, aryl [00195] In some embodiments, acidification is accomplished by a Lewis acid in an organic solvent. Useful Lewis acids are ZnX2 wherein X is Cl, Br, I, or CF3SO3.
[00196] In some embodiments, the acidifying comprises adding an amount of a Bronsted or Lewis acid effective to convert the condensed intermediate into the compound of Formula 4 without removing purine moieties from the condensed intermediate.
[00197] Acids that are useful in the acidifying step also include, but are not limited to 10% phosphoric acid in an organic solvent, 10% hydrochloric acid in an organic solvent, 1%
trifluoroacetic acid in an organic solvent, 3% dichloroacetic acid in an organic solvent or 80% acetic acid in water. The concentration of any Bronsted or Lewis acid used in the process is selected such that the concentration of the acid does not exceed a concentration that causes cleavage of the nucleobase from the sugar moiety.
[00198] In some embodiments, acidification comprises adding 1% trifluoroacetic acid in an organic solvent.
In some embodiments, acidification comprises adding about 0.1% to about 8%
trifluoroacetic acid in an organic solvent. In other embodiments, acidification comprises adding 3%
dichloroacetic acid in an organic solvent. In other embodiments, acidification comprises adding about 0.1% to about 10% dichloroacetic acid in an organic solvent. In yet other embodiments, acidification comprises adding 3% trichloroacetic acid in an organic solvent. In yet other embodiments, acidification comprises adding about 0.1% to about 10%
trichloroacetic acid in an organic solvent. In some embodiments, acidification comprises adding 80% acetic acid in water. In some embodiments, acidification comprises adding about 50%
to about 90%, or about 50%
to about 80%, about 50% to about 70%, about 50% to about 60%, about 70% to about 90% acetic acid in water. In some embodiments, the acidification comprises the further addition of cation scavengers to the acidic solvent. In specific embodiments, the cation scavengers can be triethylsilane or triisopropylsilane. In some embodiments, R' is deblocked prior to the step of acidifying the condensed intermediate. In some embodiments, R' is deblocked by acidification, which comprises adding 1%
trifluoroacetic acid in an organic solvent. In some embodiments, R' is deblocked by acidification, which comprises adding 3% dichloroacetic acid in an organic solvent. In some embodiments, R' is deblocked by acidification, which comprises adding 3% trichloroacetic acid in an organic solvent.
Removal of blocking moieities or groups [00199] Functional groups such as hydroxyl or amino moieties which are located on nucleobases or sugar moieties are routinely blocked with blocking (protecting) groups (moieties) during synthesis and subsequently deblocked. In general, a blocking group renders a chemical functionality of a molecule inert to specific reaction conditions and can later be removed from such functionality in a molecule without substantially damaging the remainder of the molecule (see e.g., Green and Wuts, Protective Groups in Organic Synthesis, 2nd Ed., John Wiley & Sons, New York, 1991). For example, amino groups can be blocked with nitrogen blocking groups such as phthalimido, 9-fludrenylmethoxycarbonyl (FMOC), triphenylmethylsulfenyl, t-BOC, 4,4'-dimethoxytrityl (DMTr), 4-methoxytrityl (MMTr), 9-phenylxanthin-9-yl (Pixyl), trityl (Tr), or 9-(p-methoxyphenyl)xanthin-9-yl (MOX). Carboxyl groups can be protected as acetyl groups. Hydroxy groups can be protected such as tetrahydropyranyl (THP), t-butyldimethylsilyl (TBDMS), 1-[(2-chloro-4-methyl)phenyl]-4-methoxypiperidin-4-yl (Ctmp), 1-(2-fluorophenyl)-4-methoxypiperidin-4-yl (Fpmp), 1-(2-chloroethoxy) ethyl, 3-methoxy-1,5-dicarbomethoxypentan-3-yl (MDP), bis(2-acetoxyethoxy)methyl (ACE), triisopropylsilyloxymethyl (TOM), 1-(2-cyanoethoxy)ethyl (CEE), 2-cyanoethoxymethyl (CEM), [4-(N-dichloroacetyl-N-methylamino)benzyloxy]methyl, 2-cyanoethyl (CN), pivaloyloxymethyl (PivOM), levunyloxymethyl (ALE). Other representative hydroxyl blocking groups have been described (see e.g., Beaucage et at., Tetrahedron, 1992, 46, 2223). In some embodiments, hydroxyl blocking groups are acid-labile groups, such as the trityl, monomethoxytrityl, dimethoxytrityl, trimethoxytrityl, 9-phenylxanthin-9-yl (Pixyl) and 9-(p-methoxyphenyl)xanthin-9-yl (MOX). Chemical functional groups can also be blocked by including them in a precursor form.
Thus an azido group can be considered a blocked form of an amine as the azido group is easily converted to the amine. Further representative protecting groups utilized in nucleic acid synthesis are known (see e.g. Agrawal et al., Protocols for Oligonucleotide Conjugates, Eds., Humana Press, New Jersey, 1994, Vol. 26, pp. 1-72).
[00200] Various methods are known and used for removal of blocking groups from the nucleic acids. In some embodiments, all blocking groups are removed. In other embodiments, the blocking groups are partially removed. In yet other embodiments, reaction conditions can be adjusted to remove blocking groups on certain moieties. In certain embodiments where R2 is a blocking group, removal of the blocking group at R2 is orthogonal to the removal of the blocking group at R'. The blocking groups at R' and R2 remain intact during the synthesis steps and are collectively removed after the chain assembly. In some embodiments, the R2 blocking group are removed simultaneously with the cleavage of the nucleic acids from the solid support and with the removal of the nucleobase blocking groups. In specific embodiments, the blocking group at R1 is removed while the blocking groups at R2 and nucleobases remain intact.
Blocking groups at R1 are cleavable on solid supports with an organic base such as a primary amine, a secondary amine, or a mixture thereof. Deblocking of the R' position is commonly referred to as front end deprotection.
[00201] In an embodiment, the nucleobase blocking groups, if present, are cleavable after the assembly of the respective nucleic acid with an acidic reagent. In another embodiment, one or more of the nucleobase blocking groups is cleavable under neither acidic nor basic conditions, e.g.
cleavable with fluoride salts or hydrofluoric acid complexes. In yet another embodiment, one or more of the nucleobase blocking groups are cleavable after the assembly of the respective nucleic acid in the presence of base or a basic solvent, and wherein the nucleobase blocking group is stable to the conditions of the front end deprotection step with amines.
[00202] In some embodiments, blocking groups for nucleobases are not required.
In other embodiments, blocking groups for nucleobases are required. In yet other embodiments, certain nucleobases require blocking group while other nucleobases do not require blocking groups. In embodiments where the nucleobases are blocked, the blocking groups are either completely or partially removed under conditions appropriate to remove the blocking group at the front end. For example, R' can denote ORE, wherein Ra is acyl, and Ba denotes guanine blocked with an acyl group including, but not limited to isobutyryl, acetyl or 4-(tert-butylphenoxy)acetyl. The acyl groups at R1 and Ba will be removed or partially removed during the same deblocking step.
Reagents Condensing reagent [00203] The condensing reagent (CR) useful in the methods of the invention has one of the following general formulae:

Z1~L Z2-~L Z4-O-L ~L or Z8-P9 L
z Z6 Z

wherein Z', Z2, Z3, Z4, Z5, Z6, Z7, Z8, and Z9 are independently selected from alkyl, aminoalkyl, cycloalkyl, heterocyclic, cycloalkylalkyl, heterocycloalkyl, aryl, heteroaryl, alkyloxy, aryloxy, or heteroaryloxy, or wherein any of Z2 and Z3, Z5 and Z6, Z7 and Z8, Z8 and Z9, Z9 and Z7, or Z7 and Z8 and Z9 are taken together to form a 3 to 20 membered alicyclic or heterocyclic ring; Q- is a counter anion; and L is a leaving group.
[00204] In some embodiments, the counter ion of the condensing reagent CR is Cl-, Br , BF4 , PF6 , TfO-, TfzN-, AsF6 , 004, or SbF6 , wherein Tf is CF3 SO2. In some embodiments, the leaving group of the condensing reagent CR is F, Cl, Br, I, 3-nitro-1,2,4-triazole, imidazole, alkyltriazole, tetrazole, pentafluorobenzene, or 1-hydroxybenzotriazole.
[00205] Examples of condensing agents that can be used in the process include, and are not limited to, pentafluorobenzoyl chloride, carbonyldiimidazole (CDI), 1-mesitylenesulfonyl-3-nitrotriazole (MSNT), 1-ethyl-3-(3'-dimethylaminopropyl)carbodiimide hydrochloride (EDCI-HC1), benzotriazole-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (PyBOP), N,N'-bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCl), 2-(1H-7-azabenzotriazol-l-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU), and O-benzotriazole-N,N,N',N'-tetramethyluronium hexafluorophosphate (HBTU), DIPCDI; NN'-bis(2-oxo-3-oxazolidinyl)phosphinic bromide (BopBr), 1, 3 -dimethyl-2-(3 -nitro-1,2,4-triazol-1-yl)-2-pyrrolidin-1-yl-1,3,2-diazaphospholidinium hexafluorophosphate (MNTP), 3-nitro-1,2,4-triazol-l-yl-tris(pyrrolidin-l-yl)phosphonium hexafluorophosphate (PyNTP), bromotripyrrolidinophosphonium hexafluorophosphate (PyBrOP); O-(benzotriazol-l-yl)-N,N,N',N'-tetramethyluronium tetrafluoroborate (TBTU); and tetramethylfluoroformamidinium hexafluorophosphate (TFFH). In certain embodiments, the counter ion of the condensing reagent CR
is Cl-, Br , BF4, PF6 , TfO-, TfzN-, AsF6 , C104 , or SbF6 , wherein Tf is CF3SO2.
[00206] In other embodiments of the invention, the condensing reagent is 1-(2,4,6-triisopropylbenzenesulfonyl)-5-(pyridin-2-yl) tetrazolide, pivaloyl chloride, bromotrispyrrolidinophosphonium hexafluorophosphate, NN'-bis(2-oxo-3-oxazolidinyl) phosphinic chloride (BopCl), or 2-chloro-5,5-dimethyl-2-oxo-1,3,2-dioxaphosphinane. In one embodiment, the condensing reagent is NN'-bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCl). Other known condensing reagents have been described (see e.g., WO/2006/066260).
[00207] In other embodiments, the condensing reagent is 1,3 -dimethyl-2-(3 -nitro-1,2,4-triazol-l-yl)-2-pyrrolidin- 1-yl-1,3,2-diazaphospholidinium hexafluorophosphate (MNTP), or 3-nitro-1,2,4-triazol-l-yl-tris(pyrrolidin-l-yl)phosphonium hexafluorophosphate (PyNTP).

01 N 0 PF6 N N O N-P-NuO C P"+ CN_(b02 N \W UU 0 \ N~-NO2 N

BopCI MNTP PyNTP
Chiral reagent [00208] In the methods of the present invention, chiral reagents are used to confer stereoselectivity in the production of X-phosphonate linkages. Many different chiral auxiliaries may be used in this process which are compounds of Formula 3-I where W1 and W2 are any of -0-, -5-, or -NG5-, which are capable of reacting with the H-phosphonate starting material, a compound of Formula 2 to form the chiral intermediate, as shown in structure III of Schemes 5 and 6.

H- W, W2-H
~U1 U3.,/G4 G3 U21r 12 G1 G
Formula 3-I
[00209] Ui and U3 are carbon atoms which are bonded to U2 if present, or to each other if r is 0, via a single, double or triple bond. U2 is -C-, -CG'-, -CG8G8-, -NGg-, -N-, -0-, or -S-where r is an integer of 0 to 5 and no more than two heteroatoms are adjacent. When any one of U2 is C, a triple bond must be formed between a second instance of U2, which is C, or to one of Ui or U3. Similarly, when any one of U2 is CG8, a double bond is formed between a second instance of U2 which is -CG8- or -N-, or to one of Ui or U3.
[00210] For example, in some embodiments, -Ui-(U2),U3- is -CG3G4-CG'G2-. In some embodiments, -Ui-(U2)rU3- is -CG3= CG'-. In some embodiments, -Ui-(U2),U3- is -C=C-. In some embodiments, -Ui-(U2)r U3- is -CG3=C G'-CG'G2-. In some embodiments, -Ui-(U2),U3- is -CG3G4-O-CG'G2-.
In some embodiments, -Ui-(U2),U3- is -CG 3G4-NGg-CG'G2-. In some embodiments, -Ui-(U2),U3- is -CG3G4-N-CGz-In some embodiments, -Ui-(U2),U3- is -CG3G4-N=C G'-CG'G2 .
[00211] G', G2, G3, G4, G5, and G8 are independently hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, hetaryl, or aryl, or two of G', G2, G3, G4, and G5 are G6 taken together form a saturated, partially unsaturated or unsaturated carbocyclic or heteroatom-containing ring of up to about 20 ring atoms which is monocyclic or polycyclic, and is fused or unfused. In some embodiments, the ring so formed is substituted by oxo, thioxo, alkyl, alkenyl, alkynyl, heteroaryl, or aryl moieties. In some embodiments, when the ring formed by taking two G6 together is substituted, it is substituted by a moiety which is bulky enough to confer stereoselectivity during the reaction.
[00212] For example, in some embodiments, the ring formed by taking two of G6 together is cyclopentyl, pyrrolyl, cyclopropyl, cyclohexenyl, cyclopentenyl, tetrahydropyranyl, or piperazinyl.
[00213] In some embodiments of the invention, the chiral reagent is a compound of Formula 3.

Formula 3 [00214] In some embodiments of Formula 3, Wi and W2 are independently -NG5-, -0-, or -S-; G', G2, G3, G4, and Gs are independently hydrogen, alkyl, aralkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, hetaryl, or aryl, or two of G1, G2, G3, G4, and Gs are G6 taken together form a saturated, partially unsaturated or unsaturated carbocyclic or heteroatom-containing ring of up to about 20 ring atoms which is monocyclic or polycyclic, fused or unfused, and no more than four of G1, G2, G3, G4, and Gs are G6. Similarly to the compounds of Formula 3', any of G1, G2, G3, G4, or Gs are substituted by oxo, thioxo, alkyl, alkenyl, alkynyl, heteroaryl, or aryl moieties. In some embodiments, such substitution induces stereoselectivity in X-phosphonate production.
[00215] In some embodiments of the invention, the chiral reagent has one of the following Formulae:

G4 /G1 G4 /G1 G4 '/iG1 G4 'iG1 G4 /G1 G4 iiG1 Formulae 3-A 3-B 3-C 3-D 3-E 3-F
[00216] In some embodiments, the chiral reagent is an aminoalcohol. In some other embodiments, the chiral reagent is an aminothiol. In yet other embodiments, the chiral reagent is an aminophenol. In some embodiments, the chiral reagent is (S)- and (R)-2-methylamino-1-phenylethanol, (1R, 2S)-ephedrine, or (1R, 2S)-2-methylamino- 1,2-diphenylethanol.
[00217] In other embodiments of the invention the chiral reagent is a compound of one of the following formulae:

H CY C)Me Ph C PhMe Formula 0 Formula P Formula Q Formula R

[00218] The choice of chiral reagent, for example, the isomer represented by Formula 0 or its stereoisomer, Formula P, permits the specific control of the chirality at phosphorus. Thus either a RP or SP
configuration can be selected in each synthesis cycle, permitting control of the overall three dimensional structure of the nucleic acid product. In some embodiments of the invention, a nucleic acid product has all RP stereocenters. In some embodiments of the invention, a nucleic acid product has all SP stereocenters. In some embodiments, the selection of RP and SP centers is made to confer a specific three dimensional superstructure to the nucleic acid chain.
Stereochemistry of Oligonucleoside Phosphorothioate Linkages [00219] Oligonucleoside phosphorothioates have shown therapeutic potential (Stein et at., Science (1993), 261:1004-12; Agrawal et at., Antisence Res. and Dev. (1992), 2:261-66; Bayever et at., Antisense Res. and Dev. (1993), 3:383-390). Oligonucleoside phosphorothioates prepared without regard to the sterochemistry of the phosphorothioate exist as a mixture of 2n diastereomers, where n is the number of internucleotide phosphorothioates linkages. The chemical and biological properties of these diastereomeric phosphorothioates can be distinct. For example, Wada et at (Nucleic Acids Symposium Series No. 51 p.
119-120; doi:10.1093/nass/nrmO60) found that stereodefined-(Rp)-(Ups)9U/(Ap)9A
duplex showed a higher Tm value than that of natural-(Up)9U/(Ap)9A and stereodefined-(Sp)-(Ups)9U did not form a duplex. In another example, in a study by Tang et al., (Nucleosides Nucleotides (1995), 14:985-990) stereopure Rp-oligodeoxyribonucleoside phosphorothioates were found to possess lower stability to nucleases endogenous to human serum that the parent oligodeoxyribonucleoside phosphorothioates with undefined phosphorous chirality.
Nucleobases and Modified Nucleobases [00220] The nucleobase Ba in Formula 1 is a natural nucleobase or a modified nucleobase derived from natural nucleobases. Examples include, but are not limited to, uracil, thymine, adenine, cytosine, and guanine having their respective amino groups protected by acyl protecting groups, 2-fluorouracil, 2-fluorocytosine, 5-bromouracil, 5-iodouracil, 2,6-diaminopurine, azacytosine, pyrimidine analogs such as pseudoisocytosine and pseudouracil and other modified nucleobases such as 8-substituted purines, xanthine, or hypoxanthine (the latter two being the natural degradation products). The modified nucleobases disclosed in Chiu and Rana, RNA, 2003, 9, 1034-1048, Limbach et at. Nucleic Acids Research, 1994, 22, 2183-2196 and Revankar and Rao, Comprehensive Natural Products Chemistry, vol. 7, 313, are also contemplated as Ba moieties of Formula 1.
[00221] Compounds represented by the following general formulae are also contemplated as modified nucleobases:

' ! 0 HN I (N

ReN~N N" N
N H I 0"

RR 0 Rio N~ N N/\N
Rio HN N R9 ~J !
R1QNN/ `N N% N N
N N

[00222] In the formulae above, R8 is a linear or branched alkyl, aryl, aralkyl, or aryloxylalkyl group having 1 to 15 carbon atoms, including, by way of example only, a methyl, isopropyl, phenyl, benzyl, or phenoxymethyl group; and each of R9 and R10 represents a linear or branched alkyl group having 1 to 4 carbon atoms.
[00223] Modified nucleobases also include expanded-size nucleobases in which one or more benzene rings has been added. Nucleic base replacements described in the Glen Research catalog (www.glenresearch.com); Krueger AT et al, Acc. Chem. Res., 2007, 40, 141-150;
Kool, ET, Acc. Chem.
Res., 2002, 35, 936-943; Benner S.A., et al., Nat. Rev. Genet., 2005, 6, 553-543; Romesberg, F.E., et al., Curr. Opin. Chem. Biol., 2003, 7, 723-733; Hirao, I., Curr. Opin. Chem. Biol., 2006, 10, 622-627, are contemplated as useful for the synthesis of the nucleic acids described herein. Some examples of these expanded-size nucleobases are shown below:
NH, 0 N~ N 'NH
NH2_ O
</ NHZ
N 'N NH

NH2 HN"""NH HNN
NH
Ni H
0 NH, HN' 'NH N

[00224] Herein, modified nucleobases also encompass structures that are not considered nucleobases but are other moieties such as, but not limited to, corrin- or porphyrin-derived rings. Porphyrin-derived base replacements have been described in Morales-Rojas, H and Kool, ET, Org. Lett., 2002, 4, 4377-4380.
Shown below is an example of a porphyrin-derived ring which can be used as a base replacement:

y ,J

[00225] Other modified nucleobases also include base replacements such as those shown below:

[00226] Modified nucleobases which are fluorescent are also contemplated. Non-limiting examples of these base replacements include phenanthrene, pyrene, stillbene, isoxanthine, isozanthopterin, terphenyl, terthiophene, benzoterthiophene, coumarin, lumazine, tethered stillbene, benzo-uracil, and naphtho-uracil, as shown below:

I N J
J N' N r/ Y NH
. ~, .~
N J~N""O O" NN~ NH2 ~s~ -vsl mss' ;psi ~s~ ~Si O~,0,~~ HN ' O O

a'Nx [00227] The modified nucleobases can be unsubstituted or contain further substitutions such as heteroatoms, alkyl groups, or linking moieties connected to fluorescent moieties, biotin or avidin moieties, or other protein or peptides. Modified nucleobases also include certain 'universal bases' that are not nucleobases in the most classical sense, but function similarly to nucleobases. One representative example of such a universal base is 3-nitropyrrole.
[00228] In addition to nucleosides of structure IV or IX, other nucleosides can also be used in the process disclosed herein and include nucleosides that incorporate modified nucleobases, or nucleobases covalently bound to modified sugars. Some examples of nucleosides that incorporate modified nucleobases include 4-acetylcytidine; 5-(carboxyhydroxylmethyl)uridine; 2'-O-methylcytidine; 5-carboxymethylaminomethyl-2-thiouridine; 5-carboxymethylaminomethyluridine; dihydrouridine; 2'-O-methylpseudouridine; beta,D-galactosylqueosine; 2'-O-methylguanosine; N6-isopentenyladenosine; 1-methyladenosine; 1-methylpseudouridine; 1-methylguanosine; 1-methylinosine; 2,2-dimethylguanosine; 2-methyladenosine; 2-methylguanosine; N7-methylguanosine; 3-methyl-cytidine; 5-methylcytidine; N6-methyladenosine; 7-methylguanosine; 5-methylaminoethyluridine; 5-methoxyaminomethyl-2-thiouridine; beta,D-mannosylqueosine; 5-methoxycarbonylmethyluridine; 5-methoxyuridine; 2-methylthio-N6-isopentenyladenosine; N-((9-beta,D-ribofuranosyl-2-methylthiopurine-6-yl)carbamoyl)threonine; N-((9-beta,D-ribofuranosylpurine-6-yl)-N-methylcarbamoyl)threonine; uridine-5-oxyacetic acid methylester;
uridine-5-oxyacetic acid (v); pseudouridine; queosine; 2-thiocytidine; 5-methyl-2-thiouridine; 2-thiouridine;
4-thiouridine; 5-methyluridine; 2'-O-methyl-5-methyluridine; and 2'-O-methyluridine.
[00229] In some embodiments, nucleosides include 6'-modified bicyclic nucleoside analogs that have either (R) or (S)-chirality at the 6'-position and include the analogs described in US Patent No. 7,399,845. In other embodiments, nucleosides include 5'-modified bicyclic nucleoside analogs that have either (R) or (S)-chirality at the 5'-position and include the analogs described in US Patent Application Publication No.
20070287831.
[00230] In some embodiments, the nucleobases or modified nucleobases comprises biomolecule binding moieties such as antibodies, antibody fragments, biotin, avidin, streptavidin, receptor ligands, or chelating moieties. In other embodiments, Ba is 5-bromouracil, 5-iodouracil, or 2,6-diaminopurine. In yet other embodiments, Ba is modified by substitution with a fluorescent or biomolecule binding moiety. In some embodiments, the substituent on Ba is a fluorescent moiety. In other embodiments, the substituent on Ba is biotin or avidin.
Modified sugars of the nucleotide/nucleoside.
[00231] The most common naturally occurring nucleotides are ribose sugars linked to the nucleobases adenosine (A), cytosine (C), guanine (G), and thymine (T) or uracil (U). Also contemplated are modified nucleotides wherein the phosphate group or the modified phosphorous atom moieties in the nucleotides can be linked to various positions of the sugar or modified sugar. As non-limiting examples, the phosphate group or the modified phosphorous-atom moiety can be linked to the 2', 3', 4' or 5' hydroxyl moiety of a sugar or modified sugar. Nucleotides that incorporate the modified nucleobases described above can also be used in the process disclosed herein. In some embodiments, nucleotides or modified nucleotides comprising an unprotected -OH moiety are used in the process disclosed herein.
[00232] In addition to the ribose moiety described in Schemes 1-4b, other modified sugars can also be incorporated in the nucleic acids disclosed herein. In some embodiments, the modified sugars contain one or more substituents at the 2' position including one of the following: F; CF3, CN, N3, NO, NO2, 0-, S-, or N-alkyl ; 0-, S-, or N-alkenyl; 0-, S- or N-alkynyl; or O-alkyl-O-alkyl, O-alkyl-N-alkyl or N-alkyl-O-alkyl wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted Ci-Cio alkyl or C2-Cio alkenyl and alkynyl. Examples of substituents include, and are not limited to, O(CH2)õ
OCH3, and O(CH2)õNH2, wherein n is from 1 to about 10, MOE, DMAOE, DMAEOE. Also contemplated herein are modified sugars described in WO 2001/088198; and Martin et al., Hely. Chim. Acta, 1995, 78, 486-504. In some embodiments, modified sugars comprise substituted silyl groups, an RNA
cleaving group, a reporter group, a fluorescent label, an intercalator, a group for improving the pharmacokinetic properties of a nucleic acid, or a group for improving the pharmacodynamic properties of a nucleic acid, and other substituents having similar properties. The modifications may be made at the at the 2', 3', 4', 5', or 6' positions of the sugar or modified sugar, including the 3' position of the sugar on the 3'-terminal nucleotide or in the 5' position of the 5'-terminal nucleotide.
[00233] Modified sugars also include sugar mimetics such as cyclobutyl or cyclopentyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, US Patent Nos.: 4,981,957;
5,118,800; 5,319,080 ; and 5,359,044. Some modified sugars that are contemplated include:

Q
R~ Ba Ri Se Ba Ri Ba Ri1 I ,Ba ls::4 R3 R2 R3 R2 R3 R2 and I R3 R2 Q = Me, Et, i-Pr, [00234] Other non-limiting examples of modified sugars include glycerol, which form glycerol nucleic acid (GNA) analogues. One example of a GNA analogue is shown below and is described in Zhang, R et al., J.
Am. Chem. Soc., 2008, 130, 5846-5847; Zhang L, et at., J. Am. Chem. Soc., 2005, 127, 4174-4175 and Tsai CH et at., PNAS, 2007, 14598-14603:

O
O=P-O Ba i X
O Ba i O=f'-O

X

H
[00235] wherein X is as defined herein. Another example of a GNA derived analogue, flexible nucleic acid (FNA) based on the mixed acetal aminal of formyl glycerol, is described in Joyce GF et al., PNAS, 1987, 84, 4398-4402 and Heuberger BD and Switzer C, J. Am. Chem. Soc., 2008, 130, 412-413, and is shown below:
i O=P-0 Ba X

O

Ba X O~

O

[00236] Other non-limiting examples of modified sugars include hexopyranosyl (6' to 4'), pentopyranosyl (4' to 2'), pentopyranosyl (4' to 3'), or tetrofuranosyl (3' to 2') sugars.
Hexopyranosyl (6' to 4') sugars contemplated include:

X.P O O Ba X,P O O Ba X,P OOH OH Ba OH OH OH ^""T

O O
n n X.P- O Ba X-PDOO O Ba -1-- I,I õ H
OH
[00237] Pentopyranosyl (4' to 2') sugars contemplated include:

O _P\IJ "J'0 OO~ _Oj-Ba O O Ba OQ~~,Ba HO j-Ba OH O'O,P\X OH P,X H O'P=X O-PAX
I Q

[00238] Pentopyranosyl (4' to 3') sugars contemplated include:
IJjIj OQ^\~Ba Ba X p'O OH
O X-D O OH
T

vw [00239] Tetrofuranosyl (3' to 2') sugars contemplated include: IJjIj Ba 0 O 0 Ba-O
I Q
X P'O
O X'P-O

vw I
vw [00240] Other modified sugars contemplated include:
"JIJ

OO O Ba O Ba O
O Ba II-Ba ii ~~- ii X O-P.X x4-0 X-P-O X.PO O Ba 0 O 0 Oi OH
~`O 1-0 0 1-0 0 koo O O O
P~ O O O P'00 O . P~ O -~-o -~, X O -~~-Ba X B X Ba X O O Ba I I I OH ! H

[00241] Further contemplated are the sugar mimetics illustrated below wherein X is selected from S, Se, CH2, N-Me, N-Et or N-iPr.

``'O HO ~ O

X.P O X.P O ~X X-P O0~
p X Ba p Ba p O,Ba OH OH ss OH ! H OH
F'O FAO
O O
X. PO +-X-~-Ba X PO O X Ba OH H

O X-Ba X P Ba 0 ~ ~ _ ~ - B a Hp -X. p Ba OH p-I X O DI~X H p-P.X p PX
T OH Q p 0 Ba p OO Ba p-\,X BaX
O Ba O Q
Xp,p OH P- OH X P-O P

Q
gy`p /--14\1 X X X Ba Ba p O Ba irBa ~-~
X
O-PAX X P-O X-P-p XP\ppO Ba X.P- p X X.P~-p X X.P~ p X X.P~ p X
p Ba p Ba X'0 -~~-Ba p 04 Ba I I I OH ! H

[00242] The modified sugars and sugar mimetics can be prepared by methods known in the art, including, but not limited to: A. Eschenmoser, Science (1999), 284:2118; M. Bohringer et at, Helv. Chim. Acta (1992), 75:1416-1477; M. Egli et al, J. Am. Chem. Soc. (2006), 128(33):10847-56; A.
Eschenmoser in Chemical Synthesis: Gnosis to Prognosis, C. Chatgilialoglu and V. Sniekus, Ed., (Kluwer Academic, Netherlands, 1996), p.293; K.-U. Schoning et at, Science (2000), 290:1347-1351; A.
Eschenmoser et at, Helv. Chim. Acta (1992), 75:218; J. Hunziker et at, Helv. Chim. Acta (1993), 76:259; G. Otting et at, Helv. Chim. Acta (1993), 76:2701; K. Groebke et at, Helv. Chim. Acta (1998), 81:375; and A.
Eschenmoser, Science (1999), 284:2118.
Blocking groups [00243] In the reactions described, it is necessary in certain embodiments to protect reactive functional groups, for example hydroxy, amino, thiol or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Protecting groups are used to block some or all reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. In one embodiment, each protective group is removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal. In some embodiments, protective groups are removed by acid, base, and/or hydrogenolysis. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used in certain embodiments to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and/or Fmoc groups, which are base labile. In other embodiments, carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butylcarbamate or with carbamates that are both acid and base stable but hydrolytically removable.
[00244] In another embodiment, hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc. In another embodiment, carboxylic acid reactive moieties are protected by conversion to simple ester compounds, or they are, in yet another embodiment, blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups are blocked with fluoride labile silyl or carbamate blocking groups.
[00245] Allyl blocking groups are useful in the presence of acid- and base-protecting groups since the former are stable and can be subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked hydroxy groups can be deprotected with a Pd(O)-catalyzed reaction in the presence of acid labile t-butylcarbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and cannot react. Once released from the resin, the functional group is available to react.
[00246] Typically blocking/protecting groups useful in the synthesis of the compounds described herein are, by way of example only:
O
0 ~S H3C i2 y \ I S' \ I 5 .
O
auvl Bn Cbz allot Me H3C\ ICH3 O
H3C-" (H3C)3C1 (H3C)3C-Si Me3si,,AO L j Et Lbbou TBDMS S Teoc O
O_1L_/
/O / K
_TA (C6H5)3C_SS
(CH3)3C
O ~ H3C~/

Boc pMBE n'i l ace 1 Emoc [00247] Representative protecting groups useful to protect nucleotides during synthesis include base labile protecting groups and acid labile protecting groups. Base labile protecting groups are used to protect the exocyclic amino groups of the heterocyclic nucleobases. This type of protection is generally achieved by acylation. Three commonly used acylating groups for this purpose are benzoyl chloride, phenoxyacetic anhydride, and isobutyryl chloride. These protecting groups are stable to the reaction conditions used during nucleic acid synthesis and are cleaved at approximately equal rates during the base treatment at the end of synthesis.
[00248] In some embodiments, the 5'-protecting group is trityl, monomethoxy trityl, dimethoxytrityl, trimethoxytrityl, 2-chlorotrityl, DATE, TBTr, 9-phenylxanthine-9-yl (Pixyl), or 9-(p-methoxyphenyl)xanthine-9-yl (MOX).
[00249] In some embodiments, thiol moieties are incorporated in the compounds of Formula 1, 2, 4, or 5 and are protected. In some embodiments, the protecting groups include, but are not limited to, pixyl, trityl, benzyl, p-methoxybenzyl (PMB), or tent-butyl (t-Bu).
[00250] Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure.
METHODS OF USE OF THE NUCLEIC ACID PRODRUGS COMPRISING A CHIRAL X-PHOSPHONATE MOIETY
[00251] The stereodefined oligonucleotide prodrug comprising a chiral phosphorous or phosphorothioate moiety which are obtained by the methods of the invention are useful in a number of areas for applications due to a combination of stability, defined chirality and ease of synthesis.
Broadly, the compounds synthesized by this method are useful as therapeutics, diagnostic probes and reagents, synthetic tools for producing other oligonucleotide products, and nanostructure materials suitable for a variety of new materials and computing applications.
[00252] The stereodefined oligonucleotide prodrug of the invention have improved serum stability over that of natural DNA/RNA equivalents, and in particular, stereodefined oligonucleotide prodrug of the class of phosphorothioates. Further, the SP isomer is more stable than the RP isomer.
In some embodiments, the level of serum stability is modulated by the introduction of either all SP
centers or SP centers at selected positions to confer resistance to degradation. In other embodiments, introduction of selectable RP and/or Sp stereocenters can provide for specific base pairing association with an endogenous or exogenous target thus protecting the target from metabolism or enhancing a particular biological reaction.
[00253] RNase H activation is also modulated by the presence of the stereocontrolled phosphorothioate nucleic acid analogs, with natural DNA/RNA being more susceptible than the RP
stereoisomer which in turn is more susceptible than the corresponding Sp isomer.
[00254] Improved duplex stability towards RNA is seen with RP phosphorothioate oligonucleotides having greater duplex stability than corresponding SP oligonucleotides which in turn demonstrates higher stability than that of natural DNA/RNA. Improved duplex stability towards DNA is seen with SP having greater duplex stability than RP which has more stability than that of natural DNA/RNA. (P. Guga, Curr. Top Med.
Chem., 2007, 7, 695-713).
[00255] These molecules may be useful as therapeutic agents, in a number of particular applications. They can be incorporated into oligonucleotides which also contain the standard DNA/RNA nucleosides, or they may be synthesized as entire sequences of the stereocontrolled oligonucleotides of the invention. Some categories of therapeutic agents include but are not limited to antisense oligonucleotides, antigene oligonucleotides which form triple helix with targeted sequences to repress transcription of undesired genes and modulate protein expression and/or activity, decoy oligonucleotides, DNA
vaccines, aptamers, ribozymes, deoxyribozymes (DNAzymes or DNA enzymes), siRNAs, microRNAs, ncRNAs (non-coding RNAs), and P-modified prodrugs. Modulation encompasses indirectly or directly increasing or decreasing the activity of a protein or inhibition or promotion of the expression of a protein. These nucleic acid compounds can be used to control cell proliferation, viral replication, or any other cell signaling process.
[00256] In one example, the field of siRNA therapeutics has a need for oligonucleotide species that can afford increased stability against RNase activity, in order to improve the duration of action over that seen with siRNA composed of natural nucleosides. Additionally, A-form helix formation appears to be more indicative of success at entering RNAi than the presence of specific native elements on the oligonucleotide.
Both of these requirements can be afforded by the use of the stereocontrolled oligonucleotides of the invention may provide enhanced stability (Y-L Chiu, T.M. Rana RNA, 2003, 9,1034-1048).
Methods of Treatment [00257] The nucleic acids described herein are useful as therapeutic agents against various disease states, including use as antiviral agents. The nucleic acids can be used as agents for treatment of diseases through modulation of DNA and/or RNA activity. In some embodiments, the nucleic acids can be used for inhibiting specific gene expression. For example, the nucleic acids can be complementary to a specific target messenger RNA (mRNA) sequence. They can be used to inhibit viral replication of myriad viruses.
Exemplary virus families include orthomyxoviruses, pox viruses, herpes viruses, papilloma viruses, picornaviruses, flaviviruses, retroviruses, hepatitis viruses, paramyxoviruses, reoviruses, parvoviruses, filoviruses, coronaviruses, arenaviruses, rhabdoviruses and adenoviruses.
Additional virus families are known and are also contemplated herein. Other examples include uses as antisense compounds against HIV
RNA or other retroviral RNA or for hybridizing to HIV mRNA encoding the tat protein, or to the TAR
region of HIV mRNA. In some embodiments, the nucleic acids mimic the secondary structure of the TAR
region of HIV mRNA, and by doing so bind the tat protein. In an embodiment, the nucleic acids is used to inhibit expression of a target protein by contacting a cell with a compound of Formula 1 wherein the expression of other proteins in the cell are not inhibited or are minimally inhibited. In some embodiment, target protein inhibition occurs in vivo in a mammal. In other embodiments, a therapeutically effective amount of a compound of Formula 1 is administered for inhibiting the expression of a target protein.

[00258] Other examples of proteins where expression can be modulated include Jun N-terminal kinase (JNK) proteins, diacylglycerol acyltransferase I, apolipoprotein B, glucagon receptor, Aurora B, acyl CoA
cholesterol acyltransferase-2, c-reactive protein, STAT (signal transducers and activators of transcription) family of proteins, and MDR P-glycoprotein. The nucleic acids can be used to inhibit protein phosphatase lB (PTPIB) expression, RNA-dependent RNA viral polymerase. The nucleic acids can be used to induce events such as apoptosis in cancer cells or to make a cell more susceptible to apoptosis. The nucleic acids can be used to modulate activities of proteins. For example, it can help modulate RNase H activity targeting multidrug resistance (MDR) RNA molecules.
[00259] In another aspect, the present invention provides methods of treating a disease mediated by undesired gene expression in a subject (e.g., mammals, such as humans) in need of such treatment. By "diseases" is meant diseases, or disease symptoms. The method includes administering to the subject an effective amount of a non-racemic pro-oligonucleotide of the present invention.
[00260] Examples of diseases mediated by undesired gene expression include cancer (e.g., leukemia, tumors, and metastases), allergy, asthma, obesity, inflammation (e.g., inflammatory diseases such as inflammatory airways disease), hypercholesterolemia, hematological disorders, severe acute respiratory syndrome (SARS), obstructive airways disease, asthma, autoimmune diseases, retroviral diseases such as AIDS or HIV, other viral infections, intrauterine infections, metabolic diseases, infection (e.g., bacterial, viral, yeast, fungal), CNS diseases, brain tumors, degenerative neural diseases, cardiovascular diseases, and diseases associated with angiogenesis, neovascularization, and vasculogenesis.
[00261] In an exemplary embodiment, the compounds are useful for treating cancer, including pancreatic cancer, and other diseases or disorders involving abnormal cell proliferation.
[00262] Located in the upper abdomen (in the retroperitoneum), the pancreas is a dual-function gland of the digestive and endocrine system. In certain instances, the pancreas functions as an endocrine gland (e.g., producing several important hormones). In certain instances, the pancreas functions as an exocrine gland (e.g., secreting fluids containing digestive enzymes that pass to the small intestine).
[00263] Pancreatic cancer is the fourth most common cause of cancer death in the US (after lung, colon and breast), comprising 6% of all cancer-related deaths. In 2008, an estimated 37,680 new cases of pancreatic cancer will have been diagnosed in the US, with 34,290 deaths. Incidence of the disease, rises linearly after age 50, with the only definitive risk factor being cigarette smoking (smokers are four times more likely to develop the disease than non-smokers). Invasive pancreatic cancer is almost always fatal. The collective median survival time of all patients is 4-6 months. Relative 1-year survival is 24%; the overall 5-year survival rate < 5%.
[00264] Pancreatic cancer is asymptomatic in its early stage and often remains undiagnosed for several months (less than one third of patients being diagnosed within 2 months of the onset symptoms). In certain instances, the delayed diagnosis results in (either partially or fully) metastasis of the cancerous cells to the liver or lymph nodes.

[00265] Currently, surgery (resectioning of the pancreas) is the primary and only curative therapy for pancreatic cancer. However, only 15-25% of tumors are resectable at the time of diagnosis and only 10-20%
of patients undergoing surgery survive more than two years. Once tumor infiltration occurs and other tissues have been affected, surgery is no longer possible.
[00266] In certain instances, diabetes mellitus or pancreatitis predisposes an individual to develop a proliferative disorder of a plurality of pancreatic cells. In certain instances, individuals are at an increased risk of developing a proliferative disorder of a plurality of pancreatic cells due to a hereditary syndrome selected from the group consisting of hereditary nonpolyposis colorectal cancer (HNPCC) and familial adenomatous polyposis (FAP). In certain instances, individuals are at an increased risk of developing a proliferative disorder of a plurality of pancreatic cells due to a mutation in a gene selected from the group consisting of MSH2, MSH6, MLH1, and APC.
[00267] Ideally, effective treatment of pancreatic cancer should (i) control the primary tumor mass, both initially and subsequently, and (ii) treat the metastatic tumor cells.
Chemoprevention (the administration of agents such as drugs, biologics, nutrients and the like) slows the progression of, reverses, or inhibits carcinogenesis, thereby lowering the risk of developing invasive or clinically significant disease.
[00268] Disclosed herein, in certain embodiments, is a method of treating pancreatic cancer. As used herein, "pancreatic cancer" includes forms of cancer of the pancreas. In some embodiments, the pancreatic cancer is metastatic pancreatic cancer. In some embodiments, the pancreatic cancer is a carcinoma, sarcoma, cancer, or combinations thereof. In some embodiments, a pancreatic cancer to be treated includes sporadic and hereditary pancreatic cancers. In some embodiments, the pancreatic cancer is duct cell carcinoma, acinar cell carcinoma, papillary mucinous carcinoma, signet ring carcinoma, adenosquamous carcinoma, undifferentiated carcinoma, mucinous carcinoma, giant cell carcinoma, small cell carcinoma, cystcancer, serous cystcancer, mucinous cystcancer, unclassified pancreatic cancer, pancreatoblastoma, or combinations thereof.
[00269] In some embodiments, an individual in need of treatment for pancreatic cancer presents with a localized tumor of the pancreas. In some embodiments, an individual in need of treatment for pancreatic cancer presents with a negative regional lymph node biopsy. In some embodiments, an individual in need of treatment for pancreatic cancer presents with a positive regional lymph node biopsy. In some embodiments, an individual in need of treatment for pancreatic cancer presents with a nodal negative pancreatic tumor (e.g., node-negative). In some embodiments, an individual in need of treatment for pancreatic cancer presents with a nodal positive tumor (e.g., node-positive).
[00270] In some embodiments, the pancreatic cancer in an individual in need of treatment for pancreatic cancer has metastasized to other locations in the body. In some embodiments, the pancreatic cancer has metastasized to a location selected from the group consisting of lymph node, stomach, bile duct, liver, bone, ovary, peritoneum and brain.

[00271] In some embodiments, cancer cells or precancerous cells are identified by histological typing or grading of a tissue sample (e.g., a biopsy sample). In some embodiments, cancer cells or precancerous cells are identified through the use of appropriate molecular markers.
[00272] In some embodiments, the pancreatic cancer in an individual in need of treatment for pancreatic cancer is staged according to the American Joint Committee on Cancer (AJCC) TNM classification system, where the tumor (T) has been assigned a stage of Tx, Ti, T2, T3, T4; and where the regional lymph nodes (N) have been assigned a stage of NX, NO, Ni; and where distant metastasis (M) has been assigned a stage of MX, MO, or M1. In some embodiments, the pancreatic cancer in an individual in need of treatment for pancreatic cancer is staged as Stage 0, I, IA, IB, II, IIA, IIB, III, and IV
pancreatic cancer. In some embodiments, the pancreatic cancer in an individual in need of treatment for pancreatic cancer is staged as Grade GX (e.g., grade cannot be assessed), Grade 1, Grade 2, Grade 3 or Grade 4.
[00273] More specific examples of cancers treated with the compounds of the present invention include breast cancer, lung cancer, melanoma, colorectal cancer, bladder cancer, ovarian cancer, prostate cancer, renal cancer, squamous cell cancer, glioblastoma, Kaposi's sarcoma, multiple myeloma, and leukemia.
Evaluation and Treatment of Cancer [00274] The term "tumor cell antigen" is defined herein as an antigen that is present in higher quantities on a tumor cell or in body fluids than unrelated tumor cells, normal cells, or in normal body fluid. The antigen presence may be tested by any number of assays known to those skilled in the art and include without limitation negative and/or positive selection with antibodies, such as an ELISA assay, a radioimmunoassay, or by Western Blot.
[00275] "Apoptosis inducing agent" is defined herein to induce apoptosis/programmed cell death, and include, for example, anticancer agents and treatments wherein cells (e.g., tumor cells) are induced to undergo programmed cell death. Exemplary apoptosis inducing agents are described in more detail below.
[00276] The terms "apoptosis" or "programmed cell death," refers to the physiological process by which unwanted or useless cells are eliminated during development and other normal biological processes.
Apoptosis is a mode of cell death that occurs under normal physiological conditions and the cell is an active participant in its own demise ("cellular suicide"). It is most often found during normal cell turnover and tissue homeostasis, embryogenesis, induction and maintenance of immune tolerance, development of the nervous system and endocrine-dependent tissue atrophy. Cells undergoing apoptosis show characteristic morphological and biochemical features. These features include chromatin aggregation, nuclear and cytoplasmic condensation, partition of cytoplasm and nucleus into membrane bound vesicles (apoptotic bodies), which contain ribosomes, morphologically intact mitochondria and nuclear material. In vivo, these apoptotic bodies are rapidly recognized and phagocytized by macrophages, dendritic cells or adjacent epithelial cells. Due to this efficient mechanism for the removal of apoptotic cells in vivo no inflammatory response is elicited. In vitro, the apoptotic bodies as well as the remaining cell fragments ultimately swell and finally lyse. This terminal phase of in vitro cell death has been termed "secondary necrosis." Apoptosis can be measured by methods known to those skilled in the art like DNA
fragmentation, exposure of Annexin V, activation of caspases, release of cytochrome c, etc. A cell that has been induced to die is termed herein as an "apoptotic cell."
[00277] Apoptosis can also be tested using a standard Annexin V Apoptosis Assay: NIH:OVCAR-3 cells are grown in 6-well plates (NUNC) and irradiated or treated with an antagonist (or in combination with another anti-cancer drug) for 4-48 hours, washed and stained with Annexin V-FITC (BD-Pharmingen) for 1 hour.
Cells are analyzed by flow cytometry (Becton-Dickinson, CellQuest), counterstained with Propidium Iodide and analyzed again in the flow cytometer.
[00278] Patients can be assessed with respect to symptoms at one or more multiple time points including prior to, during, and after treatment regimens. Treatment can result in improving the subject's condition and can be assessed by determining if one or more of the following factors has occurred: decreased tumor size, decreased cell proliferation, decreased numbers of cells, decreased neovascularization, increased apoptosis, or decreased survival of at least a portion of the tumor cells. One or more of these occurrences may, in some cases, result in partial or total elimination of the cancer and prolongation of survival of the patient.
Alternatively, for terminal stage cancers, treatment may result in stasis of disease, better quality of life and/or prolongation of survival.
Methods of Assaying Cell Migration [00279] Assays for cell migration have been described in the literature, e.g., by Brooks, et al., J. Clin. Invest 1997, 99:1390-1398 and methods for measuring cell migration are known to those of skill in the art. In one method for measuring cell migration described herein, membranes from transwell migration chambers are coated with substrate, the transwells washed, and non-specific binding sites blocked with BSA. Tumor cells from sub-confluent cultures are harvested, washed, and resuspended in migration buffer in the presence or absence of assay antibodies. After the tumor cells are allowed to migrate to the underside of the coated transwell membranes, the cells remaining on the top-side of the membrane are removed and cells that migrate to the under-side are stained with crystal violet. Cell migration is then quantified by direct cell counts per microscopic field.
Methods of Assaying Tumor Growth [00280] Tumor growth can be assayed by methods known to those of skill in the art, e.g., the SCID mouse model, the nude mouse model, and BALB/c mice with syngeneic tumors. SCID mouse models for tumor growth are carried out as follows: subconfluent human M21 melanoma cells (or any desired tumor cell type) are harvested, washed, and resuspended in sterile PBS (20 x 106 per mL). SCID
mice are injected subcutaneously with 100 L of M21 human melanoma cell (2 x 106) suspension.
Three days after tumor cell injection, mice are either untreated or treated intraperitoneally with an antagonist in the desired dose ranges.
The mice are treated daily for 24 days. Tumor size is measured with calipers and the volume estimated using the formula V = (L x W2)/2, where V is equal to the volume, L is equal to the length, and W is equal to the width.

[00281] Alternatively, nude mouse models, SCID mouse models and/or BALB/c syngeneic mouse models can also be utilized to assess tumor growth and inhibition thereof by the humanized anti-endoglin antibodies or antigen-binding fragments described herein.
Methods of Assaying Cell Proliferation [00282] Cell proliferation can be assayed by methods known to those of skill in the art. As described herein, subconfluent human endothelial cells (HUVECs) can be resuspended in proliferation buffer containing low (5.0%) serum in the presence or absence of CM (25 L) from ECV or ECVL cells, and endothelial cells allowed to proliferate for 24 hours. Proliferation can be quantified by measuring mitochondrial dehydrogenase activity using a commercially available WST-1 assay kit (Chemicon). Also, as described herein, proliferation can be quantified by measuring 3H incorporation using standard methods. (She et al., Int. J. Cancer, 108: 251-257 (2004)).
[00283] Other methods of assessing cell proliferation are known in the art and are contemplated herein.
Further non-limiting examples are described in more detail in the examples.
[00284] One would understand that classification and staging systems described herein represent one means to assess treatment of cancers described herein; additionally, other staging schemes are known in the art and may be used in connection with the methods described herein. By way of example only, the TNM
classification of malignant tumors may be used as a cancer staging system to describe the extent of cancer in a patient's body. T describes the size of the tumor and whether it has invaded nearby tissue, N describes regional lymph nodes that are involved, and M describes distant metastasis.
TNM is maintained by the International Union Against Cancer (UICC) and is used by the American Joint Committee on Cancer (AJCC) and the International Federation of Gynecology and Obstetrics (FIGO). One would understand that not all tumors have TNM classifications such as, for example, brain tumors. Generally, T (a,is,(0), 1-4) is measured as the size or direct extent of the primary tumor. N (0-3) refers to the degree of spread to regional lymph nodes: NO means that tumor cells are absent from regional lymph nodes, Ni means that tumor cells spread to the closest or small numbers of regional lymph nodes, N2 means that tumor cells spread to an extent between Ni and N3; N3 means that tumor cells spread to most distant or numerous regional lymph nodes. M (0/1) refers to the presence of metastasis: MO means that no distant metastasis are present; M1 means that metastasis has occurred to distant organs (beyond regional lymph nodes). Other parameters may also be assessed. G (1-4) refers to the grade of cancer cells (i.e., they are low grade if they appear similar to normal cells, and high grade if they appear poorly differentiated). R (0/1/2) refers to the completeness of an operation (i.e., resection-boundaries free of cancer cells or not). L (0/1) refers to invasion into lymphatic vessels. V (0/1) refers to invasion into vein. C (1-4) refers to a modifier of the certainty (quality) of V.
[00285] Provided herein are methods for degrading, inhibiting the growth of or killing cancer cells comprising contacting the cells with an amount of a compound described herein effective to degrade, inhibit the growth of or kill cancer cells.

[00286] Provided herein are methods of inhibiting tumor size increase, reducing the size of a tumor, reducing tumor proliferation or preventing tumor proliferation in an individual comprising administering to said individual an effective amount of a compound described herein to inhibit tumor size increase, reduce the size of a tumor, reduce tumor proliferation or prevent tumor proliferation.
Treatment of tumors in some cases includes stasis of symptoms, that is, by treating the patient, the cancer does not worsen and survival of the patient is prolonged.
[00287] Patients may be assessed with respect to symptoms at one or more multiple time points including prior to, during, and after treatment regimens. Treatment can result in improving the subject's condition and can be assessed by determining if one or more of the following events has occurred: decreased tumor size, decreased tumor cell proliferation, decreased numbers of cells, decreased neovascularization and/or increased apoptosis. One or more of these occurrences may, in some cases, result in partial or total elimination of the cancer and prolongation of survival of the patient.
Alternatively, for terminal stage cancers, treatment may result in stasis of disease, better quality of life and/or prolongation of survival. Other methods of assessing treatment are known in the art and contemplated herein.
[00288] In an exemplary embodiment, the pro-oligonucleotide compounds of the invention are administered to a subject such as a mammal (e.g., a human), suffering from a medical disorder, e.g., a cancer, or non-malignant conditions characterized by the presence of a class of unwanted cells.
[00289] Primary outcome measures may be assessed for patients treated using the methods described herein and include, for example, progression-free survival. In one embodiment, an increase in progression free survival is observed in an amount of by about 2-fold, 5-fold, 10-fold, 20 fold, 50 fold or more compared to lack of treatment. In another embodiment, an increase in progression free survival is increased survival by about 3 months, about 6 months, about 9 months, about 12 months, about 18 months, about 2 years, about 3 years, about 4 years, about 5 years or more compared to lack of treatment.
[00290] Secondary outcome measures may also be assessed and include duration of response, time to tumor progression, overall survival, serious and non-serious adverse events. For example, a treatment may prevent progression of the disease (i.e., stasis) or may result in an improvement.
Alternately, or in addition, other goals can be measured with respect to one or more of the following: decreased tumor burden, decreased neovascularization, reduced side effects, decreased adverse reactions, and/or increased patient compliance.
[00291] Other specific examples of diseases or disorders for which treatment by the compounds or compositions of the invention are useful for treatment or prevention include, but are not limited to transplant rejection (e.g., kidney, liver, heart, lung, islet cells, pancreas, bone marrow, cornea, small bowel, skin allografts or xenografts and other transplants), graft vs. host disease, osteoarthritis, rheumatoid arthritis, multiple sclerosis, diabetes, diabetic retinopathy, inflammatory bowel disease (e.g., Crohn's disease, ulcerative colitis, and other bowel diseases), renal disease, cachexia, septic shock, lupus, myasthenia gravis, psoriasis, dermatitis, eczema, seborrhea, Alzheimer's disease, Parkinson's disease, stem cell protection during chemotherapy, ex vivo selection or ex vivo purging for autologous or allogeneic bone marrow transplantation, ocular disease, retinopathies (e.g., macular degeneration, diabetic retinopathy, and other retinopathies), corneal disease, glaucoma, infections (e.g., bacterial, viral, or fungal), heart disease, including, but not limited to, restenosis.
Activation of RNAse L
[00292] The 2'-5' oligoadenylate (2-5A)/RNase L pathway is one of the enzymatic pathways induced by interferon. Rnase L is activated after binding to 5'-phosphoroylated fragments of 2'-5' adenylic acid. These fragments of 2'-5' adenylic acid (2-5A) are produced under the control of 2'-5' oligo(A) synthetase. This pathway is part of the innate immune system and has an important role in preventing viral infection. 2-5A-Induced cleavage of single-stranded RNA results in apoptosis. Biostable phosphorothioate analogs of 2-5A
have been shown to be potent activators of Rnase L (Xianh et al., Cancer Research (2003), 63:6795-6801).
In this study, the 2-5A analogs induced Rnase L activity and caused apoptosis in cultures of late-stage, metastatic human prostate cancer cell lines DU145, PC3 and LNCaP.
[00293] Sustained activation of RNase L triggers a mitochondrial pathway of apoptosis that eliminates virus-infected cells as well as cancerous/tumor cells. RNase L can inhibit fibrosarcoma growth, prostate cancer growth, colorectal cancer growth and pancreatic cancer growth. Given the common role of RNase L in different cancers, it is contemplated that the invention described herein can be use for the treatment of any type of cancer. Silverman, RH, Cytokine Growth Factor Rev, 18(5-6): 381-388 (2007); Bisbal, C. and Silverman, RH, Biochimie. 89(6-7): 789-798 (2007). By way of example, downregulation of RNase L refers to any reduction in expression levels of the gene or genes encoding RNase L, silencing of the gene or genes encoding RNase L, reduction in the levels of expression/translation of the proteins comprising RNase L, redudction in the amount of RNase L present within a cell, and/or any reduction in activity of RNase L as compared to a predetermined level of RNase L in an exemplary healthy population. Alternatively any reduction in RNase L levels as described herein can be indicative of downregulation of RNase L.
In one exemplary embodiment, the compounds described herein are useful for the treatment of diseases having downregulated RNase L. In another embodiment, the disease associated with downregulated RNase L is cancer. In further embodiments, the cancer is pancreatic cancer, prostate cancer, or colorectal cancer.
Alternatively, the compounds described herein are useful for the treatment of disease having upregulated RNase L. In one exemplary embodiment, the disease having upregulated RNase L
is chronic fatigue syndrome. Additional diseases having upregulated RNase L are known in the art and contemplated herein.
[00294] When used as therapeutics, the nucleic acid described herein is administered as a pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a nucleic acid comprising a chiral X-phosphonate moiety of Formula 1, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable inactive ingredient selected from pharmaceutically acceptable diluents, pharmaceutically acceptable excipients, and pharmaceutically acceptable carriers. In another embodiment, the pharmaceutical composition is formulated for intravenous injection, oral administration, buccal administration, inhalation, nasal administration, topical administration, ophthalmic administration or otic administration. In further embodiments, the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a suspension, a gel, a colloid, a dispersion, a suspension, a solution, an emulsion, an ointment, a lotion, an eye drop or an ear drop.
Pharmaceutical Compositions and Administration [00295] In another aspect, the present invention provides a pharmaceutical composition comprising a non-racemic pro-oligonucleotide in admixture with a pharmaceutically acceptable excipient. One of skill in the art will recognize that the pharmaceutical compositions include the pharmaceutically acceptable salts of the non-racemic pro-oligonucleotides described above.
Compounds for enhancing and targeting delivery [00296] The pro-oligonucleotides described herein can be delivered using a variety of delivery strategies, including conjugates of oligonucleotides with various ligands, as well as use of nanocarrier approaches. Any nucleic acid delivery strategies are contemplated for use with the pro-oligonucleotides described herein. The choice between exemplary delivery strategies, including but not limited to, chemical conjugates, cationic lipid/liposomal transfer vessicles and supramolecular nanocarriers depends on the therapeutic context, and methods for determining the optimal delivery modality are known in the art and further contemplated herein.
Cell Penetrating Compounds ("CPCs') [00297] Numerous compounds are known to act as carriers for cargo such as nucleic acids and facilitate entry of the nucleic acid in a cell in an in vivo setting. Exemplary carriers are described in Dietz et al., Molecular & Cellular Neuroscience, 27(2): 85-131 (2004) which is incorporated herein by reference. The prototypical CPCs derived from the Tat and antennepedia transcriptional regulators have been joined by a large number of new moieties. As an example, CPCs that are peptides can be relatively short (9-30 amino acids) polycationic peptides rich in argine and lysine, or membrane-interactive hydrophobic sequences. CPCs can be linked by recombinant DNA techniques or chemically coupled to peptides, oligonucleotides or nanocarriers, which then comprise the `cargo' for the CPC.
Cell Targeting Ligands ("CTLs') [00298] Another strategy is to deliver oligonucleotides by use of a CTL that binds with high affinity to a cell surface receptor that is capable of undergoing efficient internalization.
Potential ligands include antibodies, polypeptides derived from phage display libraries, and small organic molecules. Additional cell-targeting ligands are known in the art, or will be developed, and are contemplated for use with the invention described herein. Because various receptors are often preferentially expressed on particular cell types, this approach offers the possibility of improved selectivity for the oligonucleotide reagents. Exemplary receptor targets include, but are not limited to, lipoprotein receptors (such as those in the liver), integrins, receptor tyrosine kinases, and the G-protein coupled receptor (GPCR) superfamily.
Nanocarriers [00299] A variety of supramolecular nanocarriers can be used to deliver nucleic acids. Exemplary nanocarriers include, but are not limited to liposomes, cationic polymer complexes and various polymeric.

Complexation of nucleic acids with various polycations is another approach for intracellular delivery; this includes use of PEGlyated polycations, polyethyleneamine (PEI) complexes, cationic block co-polymers, and dendrimers. Several cationic nanocarriers, including PEI and polyamidoamine dendrimers help to release contents from endosomes. Other approaches include use of polymeric nanoparticles, polymer micelles, quantum dots and lipoplexes.
[00300] Additional nucleic acid delivery strategies are known in addition to the exemplary delivery strategies described herein.
[00301] In therapeutic and/or diagnostic applications, the compounds of the invention can be formulated for a variety of modes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remington, The Science and Practice of Pharmacy, (20th ed.
2000).
[00302] The compounds according to the invention are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 100 mg per day are examples of dosages that may be used. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
[00303] Pharmaceutically acceptable salts are generally well known to those of ordinary skill in the art, and may include, by way of example but not limitation, acetate, benzenesulfonate, besylate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, carnsylate, carbonate, citrate, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, mucate, napsylate, nitrate, pamoate (embonate), pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, or teoclate. Other pharmaceutically acceptable salts may be found in, for example, Remington, The Science and Practice of Pharmacy (20th ed. 2000). Preferred pharmaceutically acceptable salts include, for example, acetate, benzoate, bromide, carbonate, citrate, gluconate, hydrobromide, hydrochloride, maleate, mesylate, napsylate, pamoate (embonate), phosphate, salicylate, succinate, sulfate, or tartrate.
[00304] Depending on the specific conditions being treated, such agents may be formulated into liquid or solid dosage forms and administered systemically or locally. The agents may be delivered, for example, in a timed- or sustained- low release form as is known to those skilled in the art.
Techniques for formulation and administration may be found in Remington, The Science and Practice of Pharmacy (20th ed. 2000).
Suitable routes may include oral, buccal, by inhalation spray, sublingual, rectal, transdermal, vaginal, transmucosal, nasal or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intra-articullar, intra-sternal, intra-synovial, intra-hepatic, intralesional, intracranial, intraperitoneal, intranasal, or intraocular injections or other modes of delivery.

[00305] For injection, the agents of the invention may be formulated and diluted in aqueous solutions, such as in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For such transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
[00306] Use of pharmaceutically acceptable inert carriers to formulate the compounds herein disclosed for the practice of the invention into dosages suitable for systemic administration is within the scope of the invention. With proper choice of carrier and suitable manufacturing practice, the compositions of the present invention, in particular, those formulated as solutions, may be administered parenterally, such as by intravenous injection. The compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration. Such carriers enable the compounds of the invention to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject (e.g., patient) to be treated.
[00307] For nasal or inhalation delivery, the agents of the invention may also be formulated by methods known to those of skill in the art, and may include, for example, but not limited to, examples of solubilizing, diluting, or dispersing substances such as, saline, preservatives, such as benzyl alcohol, absorption promoters, and fluorocarbons.
[00308] Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
[00309] In addition to the active ingredients, these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. The preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
[00310] Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethyl-cellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone). If desired, disintegrating agents may be added, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[00311] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol (PEG), and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dye-stuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[00312] Pharmaceutical preparations that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols (PEGs). In addition, stabilizers may be added.
[00313] Depending upon the particular condition, or disease state, to be treated or prevented, additional therapeutic agents, which are normally administered to treat or prevent that condition, may be administered together with the inhibitors of this invention. For example, chemotherapeutic agents or other anti-proliferative agents may be combined with the inhibitors of this invention to treat proliferative diseases and cancer. Examples of known chemotherapeutic agents include, but are not limited to, adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, and platinum derivatives.
[00314] Other examples of agents the non-racemic pro-oligonucleotide of this invention may also be combined with include, without limitation, anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, riluzole, and anti-Parkinsonian agents; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, anti-leukemic agents, and growth factors; agents for treating diabetes such as insulin, insulin analogues, alpha glucosidase inhibitors, biguanides, and insulin sensitizers;
and agents for treating immunodeficiency disorders such as gamma globulin.
[00315] These additional agents may be administered separately, as part of a multiple dosage regimen, from the non-racemic pro-oligonucleotide-containing composition. Alternatively, these agents may be part of a single dosage form, mixed together with the non-racemic pro-oligonucleotide in a single composition.
[00316] The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations.

EXAMPLES
Example 1: The synthesis of (SP)- 1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tert-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] is illustrated in Scheme A.
Scheme A

~~ R ~--~ TBDPSO-I_rt ¾a H HN
0.x'N-P-N~ O 1~I-"-yi TBDPS[-,I/0 a o CI IOI Ph Y BcpCI P.^ L-2 pyridine Th3 POP,pp01 OTBDPS pyridine H OHDBHU
it OTBDPS
Th HO Ba 0 a TBDPSO-1¾a F C~ ^ S
TBDPS ~I I

1y O 3 "0 CF3COIm DTD
Cl "a pyridine NJ^~
pyridine Ph Ph OTBS

TBDPSa TBDPSa conc NH3 S .O 55'C, 12 h S` 100 F3C~ 0 0o O - rt 7a 0 1.O-, Ra N III~~~
-Ph OTBS OTBS
4tt [00317] 8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl phosphonate (1t) (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL).
N,N'-Bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCl; 500 mol) is added and the mixture is stirred for min. A solution of amino alcohol (L-2) (100 mol) is repeatedly coevaporated with dry pyridine and dissolved in dry pyridine (1 mL). The aminoalcohol solution is added to the reaction mixture dropwise via syringe, and the mixture is stirred for 5 min under argon. 3'-O-(tent-butyldimethylsilyl)thymidine 3t is dried using repeated coevaporations with dry pyridine and dissolved in 100 mol pyridine. The above mixture is added via cannula to the solution of 3'-O-(tert-butyldimethylsilyl)thymidine 3t in dry (100 mol) pyridine.
After 5 min, N-trifluoroacetyl imidazole (CF3COIm; 200 mol) is added. After an additional 30 s, N,N'-dimethylthiuram disulfide (DTD; 120 mol) is added. Following an additional 3 min, the mixture is dried in vacuum. Concentrated NH3 (10 mL) is added to the residue, and the mixture is heated for 12 h at 55 C. The mixture is then allowed to cool to room temperature and then concentrated to dryness under reduced pressure. The mixture is diluted with CHC13 (5 mL), and washed with 0.2 M
phosphate buffer (pH 7.0, 5 mL). The aqueous layers are back-extracted with CHC13 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure. The residue is purified by PTLC.

The product is dissolved in CHC13 (5 mL), washed with 0.2 M 1,8-diazabicyclo[5.4.0]undec-7-enium bicarbonate buffer (5 mL) and back-extracted with CHC13 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness to afford (SP)-4tt.
Example 2. The synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-deoxyadenosin-3'-y13'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(Se)-4at].
[00318] (SP)-4at is obtained from 1,8-diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)- deoxyadenosin-3'-yl phosphonate (1a) instead of It , using the reaction steps described in Example 1 and Scheme A for (SP)-4tt.
Example 3. The synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-deoxycytidin-3'-y13'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(Se)-4ct].
[00319] (SP)-4ct is obtained from 1,8-diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-deoxycytidin-3'-yl phosphonate (1c) instead of It, using the reaction steps described in Example 1 and Scheme A for (SP)-4tt.
Example 4. The synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)deoxyguanosin-3'-y13'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(Se)-4gt].
[00320] (SP)-4gt is obtained from 1,8-diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)deoxyguanosin-3'-yl phosphonate (1g) instead of It, using the reaction steps described in Example 1 and Scheme A for (SP)-4tt.
Example 5. The synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tert-butyldiphenylsilyl)thymidin-3'-y13'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(Rp)-4tt].
[00321] (RP)-4tt is produced via the transformations described in Example 1 and Scheme A for the synthesis of (SP)-4tt using the amino alcohol D-2 as a chiral reagent, instead of L-2.

HO N
Ph' Example 6. The synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)deoxyadenosin-3'-y13'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(Re)-4at].
[00322] (RP)-4at is produced via the transformations described in Example 2 using compound la and the amino alcohol D-2 as a chiral reagent, instead of L-2.
Example 7. The synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)deoxycytidin-3'-y13'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(Re)-4ct].

[00323] (RP)-4ct is produced via the transformations described above in Example 3 using compound lc and the amino alcohol D-2 as a chiral reagent, instead of L-2.
Example 8. The synthesis of (RP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)deoxyguanosin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(RP)-4gt].
[00324] (RP)-4gt is produced via the transformations described above in Example 4 using compound lg and the amino alcohol D-2 as a chiral reagent, instead of L-2.
Scheme B

II R II TBDPSOa H _ ....
N-P1V O ~-,/ry(JVv~1 TBDPSO,/OIga O CI O Me"
Y BopCI /F\O (aR,2S)-6 Pyridine H 'O HDBU Pyridine BaT IF`OrTBDPS
it 7 OTBDPS
BDBa HO Ba DPS a TBDPS Ba TBDPSO a TB
OTBS
O 3t j0 1% TFAICHzCIz H, oO
O"P" N-~\ pyridine H Qf,~ O O~y sa Mc,`'h j Ph O Oa OI TBS
7tt Example 9. Synthesis of (RP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(RP)-7tt] as described in Scheme B.
[00325] It (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N,N'-Bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCl; 500 mol) is added, and the mixture is stirred for 5 min. To the mixture, a solution of amino alcohol (((xR, 2S)-6) (100 mol), which has been dried by coevaportions with dry pyridine and dissolved in dry pyridine (1 mL), is added dropwise via syringe, and the mixture is stirred for 5 min under argon. 3'-O-(tent-butyldimethylsilyl)thymidine is dried using repeated coevaporations with dry pyridine and dissolved in 100 mol pyridine. The above mixture is added via cannula to the solution of 3'-O-(tert-butyldimethylsilyl)thymidine 3t in dry (100 mol) pyridine.
After 15 min, the mixture is concentrated under reduced pressure. The residue is diluted with CH2C12 (5 mL), and washed with saturated NaHCO3 (3 x 5 mL). The combined aqueous layers are back-extracted with with CH2C12 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to ca. 1 mL under reduced pressure. The residue is added dropwise via a syringe to a stirred 1% trifluoroacetic acid (TFA) solution in dry CH2C12 (20 mL) at 0 C. After an additional 5 min, the mixture is diluted with dry CH2C12 (100 mL), and washed with saturated NaHCO3 aqueous solutions (2 x 100 mL). The combined aqueous layers are back-extracted with CH2C12 (2 x 100 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure to afford crude (RP)-7tt.

Example 10. Synthesis of (Rp)-6-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)deoxyadenosin-3'-y13'-O-(tert-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(Rp)-7at].
[00326] Crude (RP)-7at is produced as described in Example 9 using la instead of It.
Example 11. Synthesis of (Rp)-4-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)deoxycytidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(Rp)-7ct].
[00327] Crude (RP)-7ct is produced as described in Example 9 using lc instead of It.
Example 12. Synthesis of (Rp)-2-N-phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)deoxyguanosin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(Rp)-7gt].
[00328] Crude (RP)-7gt is produced as described in Example 9 using Ig instead of It.
Example 13. Synthesis of (SP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-y13'-O-(tert-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(SP)-7tt].
[00329] Crude (SP)-7tt is produced as described in Example 9 using ((xS, 2R)-6 instead of ((xR, 2S)-6 as a chiral reagent.
Example 14. Synthesis of (Sp)-6-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)deoxyadenosin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(Sp)-7at].
[00330] Crude (SP)-7at is produced as described in Example 9 using compound la and ((xS, 2R)-6 instead of ((xR, 2S)-6 as a chiral reagent.
Example 15. Synthesis of (Sp)-4-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)deoxycytidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(Sp)-7ct].
[00331] Crude (SP)-7ct is produced as described in Example 9 using compound lc and ((xS, 2R)-6 instead of ((xR, 2S)-6 as a chiral reagent.
Example 16. Synthesis of (Sp)-2-N-phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)deoxyguanosin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(Sp)-7gt].
[00332] Crude (SP)-7gt is produced as described in Example 9 using compound Ig instead of It and compound ((xS, 2R)-6 instead of compound ((xR, 2S)-6 as a chiral reagent.
Scheme C

TBDPSO- _rt ~a H
0, N-P-N~ l~l--lyf TBDPS a 0 0 OTBS Ph BopCI TBSQ~P~ L-2 Oe0 OTBS pyridine Ba pyridine oiI"Or-"- TBDPS
H'p\O HDBU
8u TBSO
OTBDPS
Ba O a TBDPSO a HO TBDPSO a NH

Q OTBS 9u PiO, OTTBSCF3COIm DTD
O'P~N pyridine H w0 y4a J' CaPh pyridine ph TBSO OTBS
TBDPS a oono N H TBDPS a (3:1, , v/v) v/v S~,O OTBS rt, 12 h S-.. O OTBS
F3C(~N( Cf "sO a O"P"' 0^ Ra ~Ph ~IVOI~
TBSO OTBS TBSO OTBS
IOuu Example 17. Synthesis of (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)uridin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Sp)-l Ouu].
[00333] 1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)uridin-3'-yl phosphonate (8u) (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N,N'-Bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCl; 500 mol) is added, and the mixture is stirred for 5 min. To the mixture, a solution of amino alcohol (L-2) (100 mol), which has been dried by repeated coevaportions with dry pyridine and dissolved in dry pyridine (1 mL), is added dropwise via syringe, and the mixture is stirred for 5 min under argon. 2',3'-O-bis(tent-butyldimethylsilyl)uridine 9u is dried by repeated coevaporations with dry pyridine and dissolved in 100 mol pyridine. Then the above mixture is added via cannula into the solution of 2',3'-O-bis(tert-butyldimethylsilyl)uridine 9u (100 mol). After 10 min, N-trifluoroacetyl imidazole (CF3COIm; 200 mol) is added. After an additional 30 s, N,N'-dimethylthiuram disulfide (DTD; 120 mol) is added. After an additional 3 min, the mixture is dried in vacuum. To the residue, conc NH3-EtOH (3:1, v/v, 10 mL) is added, and the mixture is stirred for 12 h, and then concentrated to dryness under reduced pressure. Then, the mixture is diluted with CHC13 (5 mL), and washed with 0.2 M phosphate buffer (pH 7.0, 5 mL). The aqueous layers are back-extracted with CHC13 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure. The residue is purified by PTLC. The product is dissolved in CHC13 (5 mL), washed with 0.2 M 1,8-diazabicyclo[5.4.0]undec-7-enium bicarbonate buffer (5 mL) and back-extracted with CHC13 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness to afford (SP)-1 Ouu.
Example 18. Synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)adenosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Sp)-lOau].
[00334] (SP)-lOau is produced as described in Example 17 using 1,8-diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)adenosin-3'-yl phosphonate (8a) instead of 8u.
Example 19. Synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)- 2'-O-(tent-butyldimethylsilyl)cytidin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Se)-10cu].
[00335] (SP)-lOcu is produced as described in Example 17 using 1,8-diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)cytidin-3'-yl phosphonate (8c) instead of 8u.
Example 20. Synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)- 2'-O-(tent-butyldimethylsilyl)guanosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Sp)-lOgu].
[00336] (SP)-10gu is produced as described in Example 17 using 1,8-diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)guanosin-3'-yl phosphonate (8g) instead of 8u.
Example 21. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)uridin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Re)-l Ouu].
[00337] (Rp)-1Ouu is produced as described in Example 17 using chiral reagent D-2 instead of chiral reagent L-2.
Example 22. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)adenosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Rp)-lOau].
[00338] (RP)-lOau is produced as described in Example 17 using 8a instead of 8u and chiral reagent D-2 instead of chiral reagent L-2.
Example 23. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)cytidin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Re)-10cu].
[00339] (RP)-IOcu is produced as described in Example 17 using 8c instead of 8u and chiral reagent D-2 instead of chiral reagent L-2.

Example 24. Synthesis of (RP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)guanosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-ylphosphorothioate [(RP)-IOgu].
[00340] (RP)-IOgu is produced as described in Example 17 using 8g instead of 8u and chiral reagent D-2 instead of chiral reagent L-2.
Scheme D

TBDPSo-j,,4a H H
TBDPS a o CI 101 OTBS McPh BopCI TB P.4 (aR, 2S)-6 S~
O. O OTBS pyridine Ba 0, OTBDPS Pyridine H O HDBU
8u TBSO
OTBDPS
Ba Hp~~yBa TBDPSO~ya ~--/ TBDPSO-y4o a TBDPSO a ~-/ TBSO OTBS
~/
OIOTBS 9u PLOOTBS 1% TFNCH2O2 H, i0 OTBS
O'P~N pyridine H a O~~'O a /N Me Me""' CÃ Ph" I I
Ph TBSO OTBS TBSO OTBS
12uu Example 25. Synthesis of (RP)-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)uridin-3'-yl 2',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(RP)-12uu].
[00341] 8u (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N,N'-Bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCI; 500 mol) is added, and the mixture is stirred for 5 min. To the mixture, a solution of amino alcohol (((xR, 2S)-6) (100 mol), which is dried by coevaportions with dry pyridine and dissolved in dry pyridine (1 mL), is added dropwise via syringe, and the mixture is stirred for 5 min under argon. Then the mixture is added via cannula into a solution of 9u (100 mol), which is prepared by repeated coevaporations with dry pyridine and dissolution in pyridine. After 15 min, the mixture is concentrated under reduced pressure.
The residue is diluted with CH2C12 (5 mL), and washed with saturated NaHCO3 (3 x 5 mL). The combined aqueous layers are back-extracted with with CH2C12 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to ca. 1 mL under reduced pressure. The residue is added dropwise via a syringe to a stirred 1%
trifluoroacetic acid (TFA) solution in dry CH2C12 (20 mL) at 0 C. After an additional 5 min, the mixture is diluted with dry CH2C12 (100 mL), and washed with saturated NaHCO3 aqueous solutions (2 x 100 mL).
The combined aqueous layers are back-extracted with CH2C12 (2 x 100 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure to afford crude (Rp)-12uu, which is analyzed by 31P NMR.
Example 26. Synthesis of (Rp)-6-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)adenosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(RP)-12au].

[00342] Crude (RP)-12au is produced as described in Example 25 using 8a instead of 8u.
Example 27. Synthesis of (Rp)-4-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)cytidin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(RP)-12cu].
[00343] Crude (RP)-12cu is produced as described in Example 25 using 8c instead of 8u.
Example 28. Synthesis of (Rp)-2-N-Phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)guanosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-y1H-phosphonate [(RP)-12gu].
[00344] Crude (RP)-12gu is produced as described in Example 25 using 8g instead of 8u.
Example 29. Synthesis of (SP)-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tent-butyldimethylsilyl)uridin-3'-yl 2',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(SP)-12uu].
[00345] Crude (SP)-12uu is produced as described in Example 25 using chiral reagent (a.S, 2R)-6 instead of chiral reagent (aR, 2S)-6.
Example 30. Synthesis of (Sp)-6-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)adenosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(SP)-12au].
[00346] Crude (SP)-12au is produced as described in Example 25 using 8a instead of 8u and chiral reagent (a.S, 2R)-6 instead of chiral reagent (aR, 2S)-6.
Example 31. Synthesis of (Sp)-4-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)cytidin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(SP)-12cu].
[00347] Crude (SP)- 12cu is produced as described in Example 25 using 8c instead of 8u and chiral reagent (a.S, 2R)-6 instead of chiral reagent (a.R, 2S)-6.
Example 32. Synthesis of (Sp)-2-N-Phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)-2'-O-(tert-butyldimethylsilyl)guanosin-3'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-y1H-phosphonate [(SP)-12gu].
[00348] Crude (SP)-12gu is produced as described in Example 25 using 8g instead of 8u and chiral reagent (a.S, 2R)-6 instead of chiral reagent (aR, 2S)-6.

p Scheme E

H Q HJV~ HOa n II ~~ P
TBDPS Y CI
O-1 _~ ¾a O BopCI 0 TBSO OTBS
rIF'-I~(J L-2 9u TBSO O.<O pyridine pyridine pyridine HDBUOr H
13u F C"Rx N^N ~~NH TBDPS Ba 3 concNH3-EtOH
0 (3:1, v/v) CF3COIm DTD rt, 12 h TBSO
S. Ao pyridine O.PK"p-j, ga TBSIO^IOOTBS
14uu Example 33. Synthesis of (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)uridin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(SP)-14uu].

[00349] 1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)uridin-3'-yl phosphonate (13u) (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N,N'-Bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCl; 500 mol) is added, and the mixture is stirred for 5 min. To the mixture, a solution of amino alcohol (L-2) (100 mol), which has been dried by repeated coevaportions with dry pyridine and dissolved in dry pyridine (1 mL), is added dropwise via syringe, and the mixture is stirred for 5 min under argon. 2',3'-O-bis(tent-butyldimethylsilyl)uridine 9u is dried by repeated coevaporations with dry pyridine and dissolved in 100 mol pyridine. Then the above mixture is added via cannula into the solution of 2',3'-O-bis(tert-butyldimethylsilyl)uridine 9u (100 mol). After 10 min, N-trifluoroacetyl imidazole (CF3COIm; 200 mol) is added. After an additional 30 s, N,N'-dimethylthiuram disulfide (DTD; 120 mol) is added. After an additional 3 min, the mixture is dried in vacuum. To the residue, conc NH3-EtOH (3:1, v/v, 10 mL) is added, and the mixture is stirred for 12 h, and then concentrated to dryness under reduced pressure. Then, the mixture is diluted with CHC13 (5 mL), and washed with 0.2 M phosphate buffer (pH 7.0, 5 mL). The aqueous layers are back-extracted with CHC13 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure. The residue is purified by PTLC. The product is dissolved in CHC13 (5 mL), washed with 0.2 M 1,8-diazabicyclo[5.4.0]undec-7-enium bicarbonate buffer (5 mL) and back-extracted with CHC13 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness to afford (SP)- l4uu.
Example 34. Synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)adenosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Sp)-14au].
[00350] (SP)-l4au is produced as described in Example 33 using 1,8-diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)adenosin-2'-yl phosphonate (13a) instead of 13u.
Example 35. Synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)- 3'-O-(tent-butyldimethylsilyl)cytidin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Se)-14cu].
[00351] (SP)-l4cu is produced as described in Example 33 using 1,8-diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)cytidin-2'-yl phosphonate (13c) instead of 13u.
Example 36. Synthesis of (Se)-1,8-Diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)- 3'-O-(tent-butyldimethylsilyl)guanosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Sp)-14gu].
[00352] (SP)-14gu is produced as described in Example 33 using 1,8-diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)guanosin-2'-yl phosphonate (13g) instead of 13u.

Example 37. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)uridin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Re)-14uu].
[00353] (Rp)-14uu is produced as described in Example 33 using chiral reagent D-2 instead of chiral reagent L-2.
Example 38. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 6-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)adenosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-ylphosphorothioate [(Rp)-l4au].
[00354] (RP)-l4au is produced as described in Example 33 using 13a instead of 13u and chiral reagent D-2 instead of chiral reagent L-2.
Example 39. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 4-N-benzoyl-5'-O-(tert-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)cytidin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl phosphorothioate [(Re)-14cu].
[00355] (RP)- l4cu is produced as described in Example 33 using 13c instead of 13u and chiral reagent D-2 instead of chiral reagent L-2.
Example 40. Synthesis of (Re)-1,8-Diazabicyclo[5.4.0]undec-7-enium 2-N-phenoxyacetyl-5'-O-(tert-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)guanosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-ylphosphorothioate [(Rp)-l4gu].
[00356] (RP)-14gu is produced as described in Example 33 using 13g instead of 13u and chiral reagent D-2 instead of chiral reagent L-2.
Scheme F

Y F-\ R HO~o~a N-j-N IR 1--( O
TBDPSO IO a CI O Ph TBSO OTBS
BopCI (aR, 2S)-6 9u TBSO G ,< 0 pyridine pyridine pyridine HDBU'- Cr H
13u TBDPSO O a TBSO
1% TFA/CH2CI2 H,, oO a TBSO OTBS
15uu Example 41. Synthesis of (RP)-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)uridin-2'-yl 2',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(Re)-15uu].
[00357] 13u (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N,N'-Bis(2-oxo-3-oxazolidinyl)phosphinic chloride (BopCI; 500 mol) is added, and the mixture is stirred for 5 min. To the mixture, a solution of amino alcohol (((xR, 2S)-6) (100 mol), which is dried by coevaportions with dry pyridine and dissolved in dry pyridine (1 mL), is added dropwise via syringe, and the mixture is stirred for 5 min under argon. Then the mixture is added via cannula into a solution of 9u (100 mol), which is prepared by repeated coevaporations with dry pyridine and dissolution in pyridine. After 15 min, the mixture is concentrated under reduced pressure.
The residue is diluted with CH2C12 (5 mL), and washed with saturated NaHCO3 (3 x 5 mL). The combined aqueous layers are back-extracted with with CH2C12 (2 x 5 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to ca. 1 mL under reduced pressure. The residue is added dropwise via a syringe to a stirred 1%
trifluoroacetic acid (TFA) solution in dry CH2C12 (20 mL) at 0 C. After an additional 5 min, the mixture is diluted with dry CH2C12 (100 mL), and washed with saturated NaHCO3 aqueous solutions (2 x 100 mL).
The combined aqueous layers are back-extracted with CH2C12 (2 x 100 mL). The combined organic layers are dried over Na2SO4, filtered, and concentrated to dryness under reduced pressure to afford crude (Rp)-15uu, which is analyzed by 31P NMR.
Example 42. Synthesis of (Rp)-6-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)adenosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(Re)-15au].
[00358] Crude (RP)-15au is produced as described in Example 41 using 13a instead of 13u.
Example 43. Synthesis of (Rp)-4-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)cytidin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(Re)-15cu].
[00359] Crude (RP)-15cu is produced as described in Example 41 using 13c instead of 13u.
Example 44. Synthesis of (Rp)-2-N-Phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)guanosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-y1H-phosphonate [(Re)-15gu].
[00360] Crude (RP)-15gu is produced as described in Example 41 using 13g instead of 13u.
Example 45. Synthesis of (SP)-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tent-butyldimethylsilyl)uridin-2'-yl 2',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(Se)-15uu].
[00361] Crude (SP)-15uu is produced as described in Example 41 using chiral reagent (a.S, 2R)-6 instead of chiral reagent (aR, 2S)-6.
Example 46. Synthesis of (Sp)-6-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)adenosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(Se)-15au].
[00362] Crude (SP)-15au is produced as described in Example 41 using 13a instead of 13u and chiral reagent (a.S, 2R)-6 instead of chiral reagent (aR, 2S)-6.
Example 47. Synthesis of (Sp)-4-N-Benzoyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)cytidin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-yl H-phosphonate [(Se)-15cu].
[00363] Crude (SP)-15cu is produced as described in Example 41 using 13c instead of 13u and chiral reagent (a.S, 2R)-6 instead of chiral reagent (a.R, 2S)-6.
Example 48. Synthesis of (Sp)-2-N-Phenoxyacetyl-5'-O-(tent-butyldiphenylsilyl)-3'-O-(tert-butyldimethylsilyl)guanosin-2'-y12',3'-O-bis(tent-butyldimethylsilyl)uridin-5'-y1H-phosphonate [(Se)-15gu].

[00364] Crude (SP)-15gu is produced as described in Example 41 using 13g instead of 13u and chiral reagent (a.S, 2R)-6 instead of chiral reagent (aR, 2S)-6.
Scheme G: Synthesis of S-acyl-2-thioethyl nucleic acid prodrugs.

TBDPSO a TBDPSO a H NCS i..p^/SAc A~~sP`w OO a CH3CN 0a pyridine O O, OTBS OTBS
At HO a Et3N3 HF p`cS
=O
O 0-10a 16tt OH
Example 49. Synthesis of the S-acyl-2-thioethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(Re)-16tt] as described in Scheme G.
[00365] (RP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(RP)-7tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with S-acetyl-2-thioethanol (100 mol) in dry (100 mol) pyridine. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-16tt.
Example 50. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Re)-16at].
[00366] Crude (RP)-16at is produced as described in Example 49 using (RP)-7at instead of (RP)-7tt.
Example 51. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Re)-16ct].
[00367] Crude (RP)-16ct is produced as described in Example 49 using (RP)-7ct instead of (RP)-7tt.
Example 52. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Re)-16gt].
[00368] Crude (RP)-16gt is produced as described in Example 49 using (RP)-7g instead of (RP)-7tt.
Example 53. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(Se)-16tt].
[00369] Crude (SP)-16tt is produced as described in Example 49 using (SP)-7tt instead of (RP)-7tt.

Example 54. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Se)-16at].
[00370] Crude (SP)-16at is produced as described in Example 49 using (SP)-7at instead of (RP)-7tt.
Example 55. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Se)-16ct].
[00371] Crude (SP)-16ct is produced as described in Example 49 using (SP)-7ct instead of (RP)-7tt.
Example 56. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Se)-16gt].
[00372] Crude (SP)-16gt is produced as described in Example 49 using (SP)-7gt instead of (RP)-7tt.
Example 57. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphonate [(Re)-16uu].
[00373] Crude (RP)-16uu is produced as described in Example 49 using (RP)-12uu instead of (RP)-7tt.
Example 58. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Rp)-l6au].
[00374] Crude (RP)-16au is produced as described in Example 49 using (RP)-12au instead of (RP)-7tt.
Example 59. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Re)-16cu].
[00375] Crude (RP)-16cu is produced as described in Example 49 using (RP)-12cu instead of (RP)-7tt.
Example 60. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Re)-16gu].
[00376] Crude (RP)-16gu is produced as described in Example 49 using (RP)-12gu instead of (RP)-7tt.
Example 61. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphonate [(Se)-16uu].
[00377] Crude (SP)-16uu is produced as described in Example 49 using (SP)-12uu instead of (RP)-7tt.
Example 62. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Sp)-16au].
[00378] Crude (SP)-16au is produced as described in Example 49 using (SP)-12au instead of (RP)-7tt.
Example 63. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Se)-16cu].
[00379] Crude (SP)-16cu is produced as described in Example 49 using (SP)-12au instead of (RP)-7tt.
Example 64. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Se)-16gu].
[00380] Crude (SP)-16gu is produced as described in Example 49 using (SP)-12gu instead of (RP)-7tt.
Example 65. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphonate [(Re)-17uu].
[00381] Crude (SP)-17uu is produced as described in Example 49 using (SP)-15uu instead of (RP)-7tt.

Example 66. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Rp)-17au].
[00382] Crude (RP)-17au is produced as described in Example 49 using (SP)-15au instead of (RP)-7tt.
Example 67. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Re)-17cu].
[00383] Crude (RP)-17cu is produced as described in Example 49 using (SP)-15cu instead of (RP)-7tt.
Example 68. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl H-phosphonate [(Re)-17gu].
[00384] Crude (RP)-17gu is produced as described in Example 49 using (SP)-15gu instead of (RP)-7tt.
Example 69. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphonate [(Se)-17uu].
[00385] Crude (SP)-17uu is produced as described in Example 49 using (SP)-15uu instead of (RP)-7tt.
Example 70. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Sp)-17au].
[00386] Crude (SP)-17au is produced as described in Example 49 using (SP)-15au instead of (RP)-7tt.
Example 71. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Se)-17cu].
[00387] Crude (SP)-17cu is produced as described in Example 49 using (SP)-15cu instead of (RP)-7tt.
Example 72. Synthesis of the S-acyl-2-thioethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphonate [(Se)-17gu].
[00388] Crude (SP)-17gu is produced as described in Example 49 using (SP)-15gu instead of (RP)-7tt.
Scheme H: Synthesis of acyloxy pronucleotides.

TBDPSOea TTB`DPO SO-1 0a H NCS )OOH O
O OOa CH2CI2 O Ola OTBS OTBS
At HOa Et3N 3 HF
AO
O a 18tt IOOHH
Example 73. Synthesis of the acyloxy pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(Re)-18tt] as described in Scheme H.
[00389] (RP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(RP)-7tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with hydroxymethyl acetate, (100 mol) in dry (100 mol) methylene chloride. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-18tt.
Example 74. Synthesis of the acyloxy pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(RP)-18at].
[00390] Crude (RP)-18at is produced as described in Example 73 using (RP)-7at instead of (RP)-7tt.
Example 75. Synthesis of the acyloxy pronucleotide of (RP)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(RP)-18ct].
[00391] Crude (RP)-18ct is produced as described in Example 73 using (RP)-7ct instead of (RP)-7tt.
Example 76. Synthesis of the acyloxy pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(RP)-18gt].
[00392] Crude (RP)-18gt is produced as described in Example 73 using (RP)-7g instead of (RP)-7tt.
Example 77. Synthesis of the acyloxy pronucleotide of (SP)- thymidin-3'-yl thymidin-5'-yl phosphonate [(SP)-18tt].
[00393] Crude (SP)-18tt is produced as described in Example 73 using (SP)-7tt instead of (RP)-7tt.
Example 78. Synthesis of the acyloxy pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-18at].
[00394] Crude (SP)-18at is produced as described in Example 73 using (SP)-7at instead of (RP)-7tt.
Example 79. Synthesis of the acyloxy pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(SP)-18ct].
[00395] Crude (SP)-18ct is produced as described in Example 73 using (SP)-7ct instead of (RP)-7tt.
Example 80. Synthesis of the acyloxy pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(SP)-18gt].
[00396] Crude (SP)-18gt is produced as described in Example 73 using (SP)-7gt instead of (RP)-7tt.
Example 81. Synthesis of the acyloxy pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphonate [(RP)-18uu].
[00397] Crude (RP)-18uu is produced as described in Example 73 using (RP)-12uu instead of (RP)-7tt.
Example 82. Synthesis of the acyloxy pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(RP)-18au].
[00398] Crude (RP)-18au is produced as described in Example 73 using (RP)-12au instead of (RP)-7tt.
Example 83. Synthesis of the acyloxy pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(RP)-18cu].
[00399] Crude (RP)-18cu is produced as described in Example 73 using (RP)-12cu instead of (RP)-7tt.

Example 84. Synthesis of the acyloxy pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Re)-18gu].
[00400] Crude (RP)-18gu is produced as described in Example 73 using (RP)-12gu instead of (RP)-7tt.
Example 85. Synthesis of the acyloxy pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphonate [(Se)-18uu].
[00401] Crude (SP)-18uu is produced as described in Example 73 using (SP)-12uu instead of (RP)-7tt.
Example 86. Synthesis of the acyloxy pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Se)-18au].
[00402] Crude (SP)-18au is produced as described in Example 73 using (SP)-12au instead of (RP)-7tt.
Example 87. Synthesis of the acyloxy pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Se)-18cu].
[00403] Crude (SP)-18cu is produced as described in Example 73 using (SP)-12au instead of (RP)-7tt.
Example 88. Synthesis of the acyloxy pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Se)-18gu].
[00404] Crude (SP)-18gu is produced as described in Example 73 using (SP)-12gu instead of (RP)-7tt.
Example 89. Synthesis of the acyloxy pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphonate [(Re)-19uu].
[00405] Crude (SP)-19uu is produced as described in Example 73 using (SP)-15uu instead of (RP)-7tt.
Example 90. Synthesis of the acyloxy pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Re)-19au].
[00406] Crude (RP)-19au is produced as described in Example 73 using (SP)-15au instead of (RP)-7tt.
Example 91. Synthesis of the acyloxy pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Re)-19cu].
[00407] Crude (RP)-19cu is produced as described in Example 73 using (SP)-15cu instead of (RP)-7tt.
Example 92. Synthesis of the acyloxy pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphonate [(Re)-19gu].
[00408] Crude (RP)-19gu is produced as described in Example 73 using (SP)-15gu instead of (RP)-7tt.
Example 93. Synthesis of the acyloxy pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphonate [(Se)-19uu].
[00409] Crude (SP)-19uu is produced as described in Example 73 using (SP)-15uu instead of (RP)-7tt.
Example 94. Synthesis of the acyloxy pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Se)-19au].
[00410] Crude (SP)-19au is produced as described in Example 73 using (SP)-15au instead of (RP)-7tt.
Example 95. Synthesis of the acyloxy pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Se)-19cu].
[00411] Crude (SP)-19cu is produced as described in Example 73 using (SP)-15cu instead of (RP)-7tt.

Example 96. Synthesis of the acyloxy pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphonate [(Se)-19gu].
[00412] Crude (SP)-19gu is produced as described in Example 73 using (SP)-15gu instead of (RP)-7tt.
Scheme I: Synthesis of thioacyloxy pronucleotides.

TBDPSO a TTBBDPSO-1 0a OO,0a O"Oa S>e OTBS OTBS
4tt HOa Et3N 3 HF - ~~
S> jO
O Ia 20t OH
Example 97. Synthesis of the thioacyloxy pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-20tt] as described in Scheme I.
[00413] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry methylene chloride (1 mL). The mixture is treated with chloromethyl acetate, prepared by the method of Bodor et at. J.
Org. Chem. (1983), 48:5280, (100 mol) in dry (100 mol) methylene chloride. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A
0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL).
The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M
ammonium acetate buffer (pH 7.0)] to afford (RP)-20tt.
Example 98. Synthesis of the thioacyloxy pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-20at].
[00414] Crude (RP)-20at is produced as described in Example 97 using (RP)-4at instead of (RP)-4tt.
Example 99. Synthesis of the thioacyloxy pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-20ct].
[00415] Crude (RP)-20ct is produced as described in Example 97 using (RP)-4ct instead of (RP)-4tt.
Example 100. Synthesis of the thioacyloxy pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-20gt].
[00416] Crude (RP)-20gt is produced as described in Example 97 using (RP)-4g instead of (RP)-4tt.
Example 101. Synthesis of the thioacyloxy pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-20tt].
[00417] Crude (SP)-20tt is produced as described in Example 97 using (SP)-4tt instead of (RP)-4tt.

Example 102. Synthesis of the thioacyloxy pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-20at].
[00418] Crude (SP)-20at is produced as described in Example 97 using (SP)-4at instead of (RP)-4tt.
Example 103. Synthesis of the thioacyloxy pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-20ct].
[00419] Crude (SP)-20ct is produced as described in Example 97 using (SP)-4ct instead of (RP)-4tt.
Example 104. Synthesis of the thioacyloxy pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-20gt].
[00420] Crude (SP)-20gt is produced as described in Example 97 using (SP)-4gt instead of (RP)-4tt.
Example 105. Synthesis of the thioacyloxy pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-20uu].
[00421] Crude (RP)-20uu is produced as described in Example 97 using (RP)-1 Ouu instead of (RP)-4tt.
Example 106. Synthesis of the thioacyloxy pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-20au].
[00422] Crude (RP)-20au is produced as described in Example 97 using (RP)-lOau instead of (RP)-4tt.
Example 107. Synthesis of the thioacyloxy pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-20cu].
[00423] Crude (RP)-20cu is produced as described in Example 97 using (RP)-lOcu instead of (RP)-4tt.
Example 108. Synthesis of the thioacyloxy pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-20gu].
[00424] Crude (RP)-20gu is produced as described in Example 97 using (RP)-IOgu instead of (RP)-4tt.
Example 109. Synthesis of the thioacyloxy pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-20uu].
[00425] Crude (SP)-20uu is produced as described in Example 97 using (SP)-1 Ouu instead of (RP)-4tt.
Example 110. Synthesis of the thioacyloxy pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-20au].
[00426] Crude (SP)-20au is produced as described in Example 97 using (SP)-1Oau instead of (RP)-4tt.
Example 111. Synthesis of the thioacyloxy pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-20cu].
[00427] Crude (SP)-20cu is produced as described in Example 97 using (SP)-1Oau instead of (RP)-4tt.
Example 112. Synthesis of the thioacyloxy pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-20gu].
[00428] Crude (SP)-20gu is produced as described in Example 97 using (SP)-IOgu instead of (RP)-4tt.
Example 113. Synthesis of the thioacyloxy pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(RP)-2luu].
[00429] Crude (SP)-2luu is produced as described in Example 97 using (SP)-14uu instead of (RP)-4tt.

Example 114. Synthesis of the thioacyloxy pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-21au].
[00430] Crude (RP)-2lau is produced as described in Example 97 using (SP)-14au instead of (RP)-4tt.
Example 115. Synthesis of the thioacyloxy pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(RP)-21cu].
[00431] Crude (RP)-21cu is produced as described in Example 97 using (SP)-14cu instead of (RP)-4tt.
Example 116. Synthesis of the thioacyloxy pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-21gu].
[00432] Crude (RP)-21gu is produced as described in Example 97 using (SP)-14gu instead of (RP)-4tt.
Example 117. Synthesis of the thioacyloxy pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(SP)-21uu].
[00433] Crude (SP)-2luu is produced as described in Example 97 using (SP)-14uu instead of (RP)-4tt.
Example 118. Synthesis of the thioacyloxy pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-21au].
[00434] Crude (SP)-2lau is produced as described in Example 97 using (SP)-14au instead of (RP)-4tt.
Example 119. Synthesis of the thioacyloxy pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(SP)-21cu].
[00435] Crude (SP)-21cu is produced as described in Example 97 using (SP)-14cu instead of (RP)-4tt.
Example 120. Synthesis of the thioacyloxy pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-21gu].
[00436] Crude (SP)-2lgu is produced as described in Example 97 using (SP)-14gu instead of (RP)-4tt.
Scheme J: Synthesis of 2-carboalkoxyethyl pronucleotides.

TBDPSO-10a TBDPSO a S,O Me0" W Me0 O O,0a O Oa OTBS OTBS
4tt HOa -10 Et3N 3 HF
Me0 S> jO
O Ia 22t OH
Example 121. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-22tt] as described in Scheme J.
[00437] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry methylene chloride (1 mL). The mixture is treated with methyl acrylate (100 mol) in dry (100 mol) methylene chloride.
After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-22tt.
Example 122. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-22at].
[00438] Crude (RP)-22at is produced as described in Example 121 using (RP)-4at instead of (RP)-4tt.
Example 123. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-22ct].
[00439] Crude (RP)-22ct is produced as described in Example 121 using (RP)-4ct instead of (RP)-4tt.
Example 124. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-22gt].
[00440] Crude (RP)-22gt is produced as described in Example 121 using (RP)-4g instead of (RP)-4tt.
Example 125. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-22tt].
[00441] Crude (SP)-22tt is produced as described in Example 121 using (SP)-4tt instead of (RP)-4tt.
Example 126. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-22at].
[00442] Crude (SP)-22at is produced as described in Example 121 using (SP)-4at instead of (RP)-4tt.
Example 127. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-22ct].
[00443] Crude (SP)-22ct is produced as described in Example 121 using (SP)-4ct instead of (RP)-4tt.
Example 128. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-22gt].
[00444] Crude (SP)-22gt is produced as described in Example 121 using (SP)-4gt instead of (RP)-4tt.
Example 129. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-22uu].
[00445] Crude (RP)-22uu is produced as described in Example 121 using (RP)-l Ouu instead of (RP)-4tt.
Example 130. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-22au].
[00446] Crude (RP)-22au is produced as described in Example 121 using (RP)-lOau instead of (RP)-4tt.
Example 131. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-22cu].
[00447] Crude (RP)-22cu is produced as described in Example 121 using (RP)-lOcu instead of (RP)-4tt.

Example 132. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-22gu].
[00448] Crude (RP)-22gu is produced as described in Example 121 using (RP)-l Ogu instead of (RP)-4tt.
Example 133. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-22uu].
[00449] Crude (SP)-22uu is produced as described in Example 121 using (SP)-lOuu instead of (RP)-4tt.
Example 134. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-22au].
[00450] Crude (SP)-22au is produced as described in Example 121 using (SP)-lOau instead of (RP)-4tt.
Example 135. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-22cu].
[00451] Crude (SP)-22cu is produced as described in Example 121 using (SP)-lOau instead of (RP)-4tt.
Example 136. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-22gu].
[00452] Crude (SP)-22gu is produced as described in Example 121 using (SP)-lOgu instead of (RP)-4tt.
Example 137. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-23uu].
[00453] Crude (SP)-23uu is produced as described in Example 121 using (SP)-14uu instead of (RP)-4tt.
Example 138. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-23au].
[00454] Crude (RP)-23au is produced as described in Example 121 using (SP)-14au instead of (RP)-4tt.
Example 139. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-23cu].
[00455] Crude (RP)-23cu is produced as described in Example 121 using (SP)-14cu instead of (RP)-4tt.
Example 140. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-23gu].
[00456] Crude (RP)-23gu is produced as described in Example 121 using (SP)-14gu instead of (RP)-4tt.
Example 141. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-23uu].
[00457] Crude (SP)-23uu is produced as described in Example 121 using (SP)-14uu instead of (RP)-4tt.
Example 142. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-23au].
[00458] Crude (SP)-23au is produced as described in Example 121 using (SP)-14au instead of (RP)-4tt.
Example 143. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-23cu].
[00459] Crude (SP)-23cu is produced as described in Example 121 using (SP)-14cu instead of (RP)-4tt.

Example 144. Synthesis of the 2-carboalkoxyethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-23gu].
[00460] Crude (SP)-23gu is produced as described in Example 121 using (SP)-14gu instead of (RP)-4tt.
Scheme K: Synthesis of disulfide pronucleotides.

n_ ~a TBDPSO a TBDPSO, Y"y ~~I
S >e EtSSEt S -S. O
O O a EtOH O"O,0a OTBS OTBS
4tt HO a -10 Et3N3HF
S-S Ao O a 24tt IOOHH
Example 145. Synthesis of the disulfide pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-24tt] as described in Scheme K.
[00461] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry ethanol (1 mL). The mixture is treated with diethyl disulfide (200 mol) in dry (100 mol) ethanol. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A
0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL).
The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M
ammonium acetate buffer (pH 7.0)] to afford (RP)-24tt.
Example 146. Synthesis of the disulfide pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-24at].
[00462] Crude (RP)-24at is produced as described in Example 145 using (RP)-4at instead of (RP)-4tt.
Example 147. Synthesis of the disulfide pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-24ct].
[00463] Crude (RP)-24ct is produced as described in Example 145 using (RP)-4ct instead of (RP)-4tt.
Example 148. Synthesis of the disulfide pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-24gt].
[00464] Crude (RP)-24gt is produced as described in Example 145 using (RP)-4g instead of (RP)-4tt.
Example 149. Synthesis of the disulfide pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-24tt].
[00465] Crude (SP)-24tt is produced as described in Example 145 using (SP)-4tt instead of (RP)-4tt.

Example 150. Synthesis of the disulfide pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-24at].
[00466] Crude (SP)-24at is produced as described in Example 145 using (SP)-4at instead of (RP)-4tt.
Example 151. Synthesis of the disulfide pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-24ct].
[00467] Crude (SP)-24ct is produced as described in Example 145 using (SP)-4ct instead of (RP)-4tt.
Example 152. Synthesis of the disulfide pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-24gt].
[00468] Crude (SP)-24gt is produced as described in Example 145 using (SP)-4gt instead of (RP)-4tt.
Example 153. Synthesis of the disulfide pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-24uu].
[00469] Crude (RP)-24uu is produced as described in Example 145 using (RP)-l Ouu instead of (RP)-4tt.
Example 154. Synthesis of the disulfide pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-24au].
[00470] Crude (RP)-24au is produced as described in Example 145 using (RP)-lOau instead of (RP)-4tt.
Example 155. Synthesis of the disulfide pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-24cu].
[00471] Crude (RP)-24cu is produced as described in Example 145 using (RP)-lOcu instead of (RP)-4tt.
Example 156. Synthesis of the disulfide pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-24gu].
[00472] Crude (RP)-24gu is produced as described in Example 145 using (RP)-l Ogu instead of (RP)-4tt.
Example 157. Synthesis of the disulfide pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-24uu].
[00473] Crude (SP)-24uu is produced as described in Example 145 using (SP)-lOuu instead of (RP)-4tt.
Example 158. Synthesis of the disulfide pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-24au].
[00474] Crude (SP)-24au is produced as described in Example 145 using (SP)-lOau instead of (RP)-4tt.
Example 159. Synthesis of the disulfide pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-24cu].
[00475] Crude (SP)-24cu is produced as described in Example 145 using (SP)-lOau instead of (RP)-4tt.
Example 160. Synthesis of the disulfide pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-24gu].
[00476] Crude (SP)-24gu is produced as described in Example 145 using (SP)-lOgu instead of (RP)-4tt.
Example 161. Synthesis of the disulfide pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-25uu].
[00477] Crude (SP)-25uu is produced as described in Example 145 using (SP)-14uu instead of (RP)-4tt.

Example 162. Synthesis of the disulfide pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-25au].
[00478] Crude (RP)-25au is produced as described in Example 145 using (SP)-14au instead of (RP)-4tt.
Example 163. Synthesis of the disulfide pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-25cu].
[00479] Crude (RP)-25cu is produced as described in Example 145 using (SP)-14cu instead of (RP)-4tt.
Example 164. Synthesis of the disulfide pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-25gu].
[00480] Crude (RP)-25gu is produced as described in Example 145 using (SP)-14gu instead of (RP)-4tt.
Example 165. Synthesis of the disulfide pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-25uu].
[00481] Crude (SP)-25uu is produced as described in Example 145 using (SP)-14uu instead of (RP)-4tt.
Example 166. Synthesis of the disulfide pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-25au].
[00482] Crude (SP)-25au is produced as described in Example 145 using (SP)-14au instead of (RP)-4tt.
Example 167. Synthesis of the disulfide pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-25cu].
[00483] Crude (SP)-25cu is produced as described in Example 145 using (SP)-14cu instead of (RP)-4tt.
Example 168. Synthesis of the disulfide pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-25gu].
[00484] Crude (SP)-25gu is produced as described in Example 145 using (SP)-14gu instead of (RP)-4tt.
Scheme L: Synthesis of thioacetal pronucleotides.

TBDPSOa Me0`^/OAc Ac TBDPSO a Me0 _ S ~O TMSOTf '00 0 wO a CH2CI2 MeO 0 10, õ ga OTBS OTBS
4tt AcO~ HOa Et3N 3 HF
S;jO
MeO O 0- Ba 26tt OH
Example 169. Synthesis of the thioacetal pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-26tt] as described in Scheme L.
[00485] 3,3-Dimethoxypropyl acetate (100 mol) is added to a solution of trimethylsilyltriflate (100 mol) in methylene chloride (1 mL) at -78 C. After stirring at -78 C for 30 min, (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is added in dry methylene chloride (1 mL). The mixture is allowed to slowly warm to room temperature. After 1 hour, the mixture is
129 concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-26tt.
Example 170. Synthesis of the thioacetal pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-26at].
[00486] Crude (RP)-26at is produced as described in Example 169 using (RP)-4at instead of (RP)-4tt.
Example 171. Synthesis of the thioacetal pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-26ct].
[00487] Crude (RP)-26ct is produced as described in Example 169 using (RP)-4ct instead of (RP)-4tt.
Example 172. Synthesis of the thioacetal pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-26gt].
[00488] Crude (RP)-26gt is produced as described in Example 169 using (RP)-4g instead of (RP)-4tt.
Example 173. Synthesis of the thioacetal pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-26tt].
[00489] Crude (SP)-26tt is produced as described in Example 169 using (SP)-4tt instead of (RP)-4tt.
Example 174. Synthesis of the thioacetal pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-26at].
[00490] Crude (SP)-26at is produced as described in Example 169 using (SP)-4at instead of (RP)-4tt.
Example 175. Synthesis of the thioacetal pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-26ct].
[00491] Crude (SP)-26ct is produced as described in Example 169 using (SP)-4ct instead of (RP)-4tt.
Example 176. Synthesis of the thioacetal pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-26gt].
[00492] Crude (SP)-26gt is produced as described in Example 169 using (SP)-4gt instead of (RP)-4tt.
Example 177. Synthesis of the thioacetal pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-26uu].
[00493] Crude (RP)-26uu is produced as described in Example 169 using (RP)-1 Ouu instead of (RP)-4tt.
Example 178. Synthesis of the thioacetal pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-26au].
[00494] Crude (RP)-26au is produced as described in Example 169 using (RP)-1Oau instead of (RP)-4tt.
Example 179. Synthesis of the thioacetal pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-26cu].
[00495] Crude (RP)-26cu is produced as described in Example 169 using (RP)-IOcu instead of (RP)-4tt.
130 Example 180. Synthesis of the thioacetal pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-26gu].
[00496] Crude (RP)-26gu is produced as described in Example 169 using (RP)-IOgu instead of (RP)-4tt.
Example 181. Synthesis of the thioacetal pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-26uu].
[00497] Crude (SP)-26uu is produced as described in Example 169 using (SP)-lOuu instead of (RP)-4tt.
Example 182. Synthesis of the thioacetal pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-26au].
[00498] Crude (SP)-26au is produced as described in Example 169 using (SP)-lOau instead of (RP)-4tt.
Example 183. Synthesis of the thioacetal pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-26cu].
[00499] Crude (SP)-26cu is produced as described in Example 169 using (SP)-lOau instead of (RP)-4tt.
Example 184. Synthesis of the thioacetal pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-26gu].
[00500] Crude (SP)-26gu is produced as described in Example 169 using (SP)-lOgu instead of (RP)-4tt.
Example 185. Synthesis of the thioacetal pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-27uu].
[00501] Crude (SP)-27uu is produced as described in Example 169 using (SP)-14uu instead of (RP)-4tt.
Example 186. Synthesis of the thioacetal pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-27au].
[00502] Crude (RP)-27au is produced as described in Example 169 using (SP)-14au instead of (RP)-4tt.
Example 187. Synthesis of the thioacetal pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-27cu].
[00503] Crude (RP)-27cu is produced as described in Example 169 using (SP)-14cu instead of (RP)-4tt.
Example 188. Synthesis of the thioacetal pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-27gu].
[00504] Crude (RP)-27gu is produced as described in Example 169 using (SP)-14gu instead of (RP)-4tt.
Example 189. Synthesis of the thioacetal pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-27uu].
[00505] Crude (SP)-27uu is produced as described in Example 169 using (SP)-14uu instead of (RP)-4tt.
Example 190. Synthesis of the thioacetal pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-27au].
[00506] Crude (SP)-27au is produced as described in Example 169 using (SP)-14au instead of (RP)-4tt.
Example 191. Synthesis of the thioacetal pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-27cu].
[00507] Crude (SP)-27cu is produced as described in Example 169 using (SP)-14cu instead of (RP)-4tt.
131 Example 192. Synthesis of the thioacetal pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-27gu].
[00508] Crude (SP)-27gu is produced as described in Example 169 using (SP)-14gu instead of (RP)-4tt.
Scheme M: Synthesis of C3 enol ester pronucleotides.

TBDPSO a Ac TBDPSO a CI~OAc S >e i0 OOa O"Oa DMF
OTBS OTBS
4tt AcO,,. HOa Et3N 3 HF
Sj 0 p OHY'y 28tt Example 193. Synthesis of the C3 enol ester pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-28tt] as described in Scheme M.
[00509] To a solution of (E)-3-chloroprop-l-enyl acetate (100 mol) in DMF (1 mL) is added (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol).
After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-28tt.
Example 194. Synthesis of the C3 enol ester pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-28at].
[00510] Crude (RP)-28at is produced as described in Example 193 using (RP)-4at instead of (RP)-4tt.
Example 195. Synthesis of the C3 enol ester pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-28ct].
[00511] Crude (RP)-28ct is produced as described in Example 193 using (RP)-4ct instead of (RP)-4tt.
Example 196. Synthesis of the C3 enol ester pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-28gt].
[00512] Crude (RP)-28gt is produced as described in Example 193 using (RP)-4g instead of (RP)-4tt.
Example 197. Synthesis of the C3 enol ester pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-28tt].
[00513] Crude (SP)-28tt is produced as described in Example 193 using (SP)-4tt instead of (RP)-4tt.
Example 198. Synthesis of the C3 enol ester pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-28at].
132 [00514] Crude (SP)-28at is produced as described in Example 193 using (SP)-4at instead of (RP)-4tt.
Example 199. Synthesis of the C3 enol ester pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-28ct].
[00515] Crude (SP)-28ct is produced as described in Example 193 using (SP)-4ct instead of (RP)-4tt.
Example 200. Synthesis of the C3 enol ester pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-28gt].
[00516] Crude (SP)-28gt is produced as described in Example 193 using (SP)-4gt instead of (RP)-4tt.
Example 201. Synthesis of the C3 enol ester pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-28uu].
[00517] Crude (RP)-28uu is produced as described in Example 193 using (RP)-1 Ouu instead of (RP)-4tt.
Example 202. Synthesis of the C3 enol ester pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-28au].
[00518] Crude (RP)-28au is produced as described in Example 193 using (RP)-1Oau instead of (RP)-4tt.
Example 203. Synthesis of the C3 enol ester pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-28cu].
[00519] Crude (RP)-28cu is produced as described in Example 193 using (RP)-IOcu instead of (RP)-4tt.
Example 204. Synthesis of the C3 enol ester pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-28gu].
[00520] Crude (RP)-28gu is produced as described in Example 193 using (RP)-IOgu instead of (RP)-4tt.
Example 205. Synthesis of the C3 enol ester pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-28uu].
[00521] Crude (SP)-28uu is produced as described in Example 193 using (SP)-lOuu instead of (RP)-4tt.
Example 206. Synthesis of the C3 enol ester pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-28au].
[00522] Crude (SP)-28au is produced as described in Example 193 using (SP)-lOau instead of (RP)-4tt.
Example 207. Synthesis of the C3 enol ester pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-28cu].
[00523] Crude (SP)-28cu is produced as described in Example 193 using (SP)-lOau instead of (RP)-4tt.
Example 208. Synthesis of the C3 enol ester pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-28gu].
[00524] Crude (SP)-28gu is produced as described in Example 193 using (SP)-lOgu instead of (RP)-4tt.
Example 209. Synthesis of the C3 enol ester pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-29uu].
[00525] Crude (SP)-29uu is produced as described in Example 193 using (SP)-14uu instead of (RP)-4tt.
Example 210. Synthesis of the C3 enol ester pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-29au].
133 [00526] Crude (RP)-29au is produced as described in Example 193 using (SP)-14au instead of (RP)-4tt.
Example 211. Synthesis of the C3 enol ester pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-29cu].
[00527] Crude (RP)-29cu is produced as described in Example 193 using (SP)-14cu instead of (RP)-4tt.
Example 212. Synthesis of the C3 enol ester pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-29gu].
[00528] Crude (RP)-29gu is produced as described in Example 193 using (SP)-14gu instead of (RP)-4tt.
Example 213. Synthesis of the C3 enol ester pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-29uu].
[00529] Crude (SP)-29uu is produced as described in Example 193 using (SP)-14uu instead of (RP)-4tt.
Example 214. Synthesis of the C3 enol ester pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-29au].
[00530] Crude (SP)-29au is produced as described in Example 193 using (SP)-14au instead of (RP)-4tt.
Example 215. Synthesis of the C3 enol ester pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-29cu].
[00531] Crude (SP)-29cu is produced as described in Example 193 using (SP)-14cu instead of (RP)-4tt.
Example 216. Synthesis of the C3 enol ester pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-29gu].
[00532] Crude (SP)-29gu is produced as described in Example 193 using (SP)-14gu instead of (RP)-4tt.
Scheme N: Synthesis of C4 enol ester pronucleotides.

TBDPSO a Ac0 TBDPSO a ~OAc CI
S >e i0 OOa O"Oa DMF
OTBS OTBS
4tt Ac-V__\_ HO2 Et3N 3 HF
S> jO
O Ia 30tt OH
Example 217. Synthesis of the C4 enol ester pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-30tt] as described in Scheme N.
[00533] To a solution of (E)-4-chlorobut-l-enyl acetate (100 mol) in DMF (1 mL) is added (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol).
After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced
134 pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-30tt.
Example 218. Synthesis of the C4 enol ester pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-30at].
[00534] Crude (RP)-30at is produced as described in Example 217 using (RP)-4at instead of (RP)-4tt.
Example 219. Synthesis of the C4 enol ester pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-30ct].
[00535] Crude (RP)-30ct is produced as described in Example 217 using (RP)-4ct instead of (RP)-4tt.
Example 220. Synthesis of the C4 enol ester pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-30gt].
[00536] Crude (RP)-30gt is produced as described in Example 217 using (RP)-4g instead of (RP)-4tt.
Example 221. Synthesis of the C4 enol ester pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-30tt].
[00537] Crude (SP)-30tt is produced as described in Example 217 using (SP)-4tt instead of (RP)-4tt.
Example 222. Synthesis of the C4 enol ester pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-30at].
[00538] Crude (SP)-30at is produced as described in Example 217 using (SP)-4at instead of (RP)-4tt.
Example 223. Synthesis of the C4 enol ester pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-30ct].
[00539] Crude (SP)-30ct is produced as described in Example 217 using (SP)-4ct instead of (RP)-4tt.
Example 224. Synthesis of the C4 enol ester pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-30gt].
[00540] Crude (SP)-30gt is produced as described in Example 217 using (SP)-4gt instead of (RP)-4tt.
Example 225. Synthesis of the C4 enol ester pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(RP)-30uu].
[00541] Crude (RP)-30uu is produced as described in Example 217 using (RP)-10uu instead of (RP)-4tt.
Example 226. Synthesis of the C4 enol ester pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-30au].
[00542] Crude (RP)-30au is produced as described in Example 217 using (RP)-10au instead of (RP)-4tt.
Example 227. Synthesis of the C4 enol ester pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-30cu].
[00543] Crude (RP)-30cu is produced as described in Example 217 using (RP)-10cu instead of (RP)-4tt.
Example 228. Synthesis of the C4 enol ester pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-30gu].
[00544] Crude (RP)-30gu is produced as described in Example 217 using (RP)-10gu instead of (RP)-4tt.
135 Example 229. Synthesis of the C4 enol ester pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(SP)-30uu].
[00545] Crude (SP)-30uu is produced as described in Example 217 using (SP)-lOuu instead of (RP)-4tt.
Example 230. Synthesis of the C4 enol ester pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-30au].
[00546] Crude (SP)-30au is produced as described in Example 217 using (SP)-10au instead of (RP)-4tt.
Example 231. Synthesis of the C4 enol ester pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-30cu].
[00547] Crude (SP)-30cu is produced as described in Example 217 using (SP)-10au instead of (RP)-4tt.
Example 232. Synthesis of the C4 enol ester pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(SP)-30gu].
[00548] Crude (SP)-30gu is produced as described in Example 217 using (SP)-lOgu instead of (RP)-4tt.
Example 233. Synthesis of the C4 enol ester pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(RP)-3luu].
[00549] Crude (SP)-3 luu is produced as described in Example 217 using (SP)-14uu instead of (RP)-4tt.
Example 234. Synthesis of the C4 enol ester pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-3 l au].
[00550] Crude (RP)-31 au is produced as described in Example 217 using (SP)-14au instead of (RP)-4tt.
Example 235. Synthesis of the C4 enol ester pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(RP)-31cu].
[00551] Crude (RP)-31cu is produced as described in Example 217 using (SP)-14cu instead of (RP)-4tt.
Example 236. Synthesis of the C4 enol ester pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-31gu].
[00552] Crude (RP)-31gu is produced as described in Example 217 using (SP)-14gu instead of (RP)-4tt.
Example 237. Synthesis of the C4 enol ester pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(SP)-3 luu].
[00553] Crude (SP)-3 luu is produced as described in Example 217 using (SP)-14uu instead of (RP)-4tt.
Example 238. Synthesis of the C4 enol ester pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-3 l au].
[00554] Crude (SP)-3 l au is produced as described in Example 217 using (SP)-l4au instead of (RP)-4tt.
Example 239. Synthesis of the C4 enol ester pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(SP)-31cu].
[00555] Crude (SP)-3lcu is produced as described in Example 217 using (SP)-l4cu instead of (RP)-4tt.
Example 240. Synthesis of the C4 enol ester pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-31gu].
[00556] Crude (SP)-31gu is produced as described in Example 217 using (SP)-14gu instead of (RP)-4tt.
136 Scheme 0: Synthesis of protected 2'-5'-A3 H-phosphonate (MeOC6H4)3C\ O O A

(MeOC6H4)3 O A Scheme 46 TBSO TFA

H. i0 TBSO 0.0 HO-/O~IA OAP "O O~IA
HDBU'- Cr H ~J ~--/

9a HO A (MeOC6H4)3O A

Scheme 46 TFA

H. oO H. i0 OiP ~,0 A couple with 32 H. oO -0 TBSO OTBS
Example 241. The synthesis of a 5'-O-(methoxytrityl) protected 2'-5'-A3 H-phosphonate is illustrated in Scheme O-a.
[00557] 5'-O-(Methoxytrityl) protected compound 32 is coupled with 9a as described in Scheme 6, Example 41. The resulting H-phosphonate 33 is subjected to 5'-O-(methoxytrityl) deprotection by treatment with 1%
TFA in CH2C12 to give 5'-OH compound 34. Coupling of 34 with 32, as described in Scheme 6, Example 41, gives the H-phosphonate trinucleotide 35. Deprotection of the 5'-OH group with 1% TFA in CH2C12 to gives H-phosphonate trinucleotide 36.
137 Scheme O-b: Synthesis of a 2'-5'-A3 S-acetyl-2-thioethyl pronucleotide HO A

H, .O H, *0 /,~/SAc O 'O A Otl A NCS HO
CF3CN pyridine TBSO TBSO
A O"O O
H ;P a A

TBSO OTBS TBSO OTBS
Ik_"'_O' '0 J'S~ .0 _YS
II_j/ O A ~y Sv /% O A

/ g TBS Et3N3 HF
`5-'*~'O' .O = S/~~O ID
O

g TBSO g HO
/ \s' '- .O " `S^/ .0 O "' A OP` A
38 O-~I I~~~I( 39 TBSO OTBS HO OH
Example 242. Synthesis a 2'-5'-A3 S-acetyl-2-thioethyl pronucleotide is illustrated in Scheme O-b.
[00558] 5'-OH H-phosphonate trinucleotide compound 36 is converted to the S-acetyl-2-thioethyl prodrug by the method of Eldrup, as described in US 7,202,224. To 36 (1 mmol) is added 1H-tetrazole (1.1 mmol) and the mixture dried overnight over P205. To this mixture is added dry acetonitrile (10 mL) followed by bis(S-acetyl-2-thioethyl)N,N-diisopropylphosphoramidite (1.1 mmol) and the resulting mixture is stirred at room temperature for 2 hours. The solvent is removed, the residue cooled to -40 C
and a solution of m-CPBA
(1.0 mmol) in dichloromethane (10 mL) is added. After stirring at room temperature for 1 hour, aq.
NaHSO3 is added and the organic layer separated and product 37 is isolated by chromatography.
[00559] Compound 37 is converted into the final product 39 following the procedure of Scheme 7, Example 49. Compound 37 (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with S-acetyl-2-thioethanol (100 mol) in dry (100 mol) pyridine.
After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford 39.
138 Scheme P: Synthesis of trimethylammoniumethyl nucleic acid prodrugs.
T a ~/O OG O
Hk ~O W-S HOv Ol -0 Cf IeR,, 0~_~
0_1~_ CH3CN pyridine OTBS OTBS
At a Et3N3 HF Fe. 0 e 0_1~_ 40tt CIS
Example 243. Synthesis of the trimethylammoniumethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-16tt] as described in Scheme P.
[00560] (RP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(RP)-7tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with 1-(2-hydroxy)-ethyl-trimethylammonium chloride (100 mol) in dry (100 mol) pyridine. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M
ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-40tt.
Example 244. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Rp)-40at].
[00561] Crude (RP)-40at is produced as described in Example 243 using (RP)-7at instead of (RP)-7tt.
Example 245. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Rp)-40ct].
[00562] Crude (RP)-40ct is produced as described in Example 49 using (RP)-7ct instead of (RP)-7tt.
Example 246. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Rp)-40gt].
[00563] Crude (RP)-40gt is produced as described in Example 49 using (RP)-7g instead of (RP)-7tt.
Example 247. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(SP)-40tt].
[00564] Crude (SP)-40tt is produced as described in Example 49 using (SP)-7tt instead of (RP)-7tt.
139 Example 248. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-40at].
[00565] Crude (SP)-40at is produced as described in Example 49 using (SP)-7at instead of (RP)-7tt.
Example 249. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Sp)-40ct].
[00566] Crude (SP)-40ct is produced as described in Example 49 using (SP)-7ct instead of (RP)-7tt.
Example 250. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-40gt].
[00567] Crude (SP)-40gt is produced as described in Example 49 using (SP)-7gt instead of (RP)-7tt.
Example 251. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphonate [(Rp)-40uu].
[00568] Crude (RP)-40uu is produced as described in Example 49 using (RP)-12uu instead of (RP)-7tt.
Example 252. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Rp)-40au].
[00569] Crude (RP)-40au is produced as described in Example 49 using (RP)-12au instead of (RP)-7tt.
Example 253. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Rp)-40cu].
[00570] Crude (RP)-16cu is produced as described in Example 49 using (RP)-12cu instead of (RP)-7tt.
Example 254. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Rp)-40gu].
[00571] Crude (RP)-40gu is produced as described in Example 49 using (RP)-12gu instead of (RP)-7tt.
Example 255. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphonate [(Sp)-40uu].
[00572] Crude (SP)-40uu is produced as described in Example 49 using (SP)-12uu instead of (RP)-7tt.
Example 256. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Sp)-40au].
[00573] Crude (SP)-40au is produced as described in Example 49 using (SP)-12au instead of (RP)-7tt.
Example 257. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Sp)-40cu].
[00574] Crude (SP)-40cu is produced as described in Example 49 using (SP)-12au instead of (RP)-7tt.
Example 258. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Sp)-40gu].
[00575] Crude (SP)-40gu is produced as described in Example 49 using (SP)-12gu instead of (RP)-7tt.
Example 259. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphonate [(RP)-41uu].
[00576] Crude (RP)-4luu is produced as described in Example 49 using (RP)-15uu instead of (RP)-7tt.
140 Example 260. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Rp)-4lau].
[00577] Crude (RP)-41 au is produced as described in Example 49 using (SP)-15au instead of (RP)-7tt.
Example 261. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Rp)-4lcu].
[00578] Crude (RP)-4lcu is produced as described in Example 49 using (SP)-15cu instead of (RP)-7tt.
Example 262. Synthesis of the trimethylammoniumethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl H-phosphonate [(RP)-41gu].
[00579] Crude (RP)-4lgu is produced as described in Example 49 using (SP)-15gu instead of (RP)-7tt.
Example 263. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphonate [(SP)-41uu].
[00580] Crude (SP)-4luu is produced as described in Example 49 using (SP)-15uu instead of (RP)-7tt.
Example 264. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Sp)-4lau].
[00581] Crude (SP)-41 au is produced as described in Example 49 using (SP)-15au instead of (RP)-7tt.
Example 265. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Sp)-4lcu].
[00582] Crude (SP)-4lcu is produced as described in Example 49 using (SP)-15cu instead of (RP)-7tt.
Example 266. Synthesis of the trimethylammoniumethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphonate [(SP)-41gu].
[00583] Crude (SP)-4lgu is produced as described in Example 49 using (SP)-15gu instead of (RP)-7tt.
Scheme Q: Synthesis of alkylhydroxamate nucleic acid prodrugs.

T a /K/19 a N:.S Ho,-\/CCN(OMeWe 0 0. O
CH3CN py-idine OTBS OTBS
At i a Et3N3 HF
O O~0 42tt OH
Example 267. Synthesis of the alkylhydroxamate nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-42tt] as described in Scheme Q.
[00584] (RP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(RP)-7tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved
141 in dry pyridine (1 mL). N-chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with N-methoxy-N-methyl-3-hydroxypropionamide (100 mol) in dry (100 mol) pyridine.
After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M
ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-42tt.
Example 268. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Rp)-42at].
[00585] Crude (RP)-42at is produced as described in Example 267 using (RP)-7at instead of (RP)-7tt.
Example 269. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Rp)-42ct].
[00586] Crude (RP)-42ct is produced as described in Example 267 using (RP)-7ct instead of (RP)-7tt.
Example 270. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Rp)-42gt].
[00587] Crude (RP)-42gt is produced as described in Example 267 using (RP)-7g instead of (RP)-7tt.
Example 271. Synthesis of the alkylhydroxamate pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphonate [(SP)-42tt].
[00588] Crude (SP)-42tt is produced as described in Example 267 using (SP)-7tt instead of (RP)-7tt.
Example 272. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-42at].
[00589] Crude (SP)-42at is produced as described in Example 267 using (SP)-7at instead of (RP)-7tt.
Example 273. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Sp)-42ct].
[00590] Crude (SP)-42ct is produced as described in Example 267 using (SP)-7ct instead of (RP)-7tt.
Example 274. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-42gt].
[00591] Crude (SP)-42gt is produced as described in Example 267 using (SP)-7gt instead of (RP)-7tt.
Example 275. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphonate [(Rp)-42uu].
[00592] Crude (RP)-42uu is produced as described in Example 267 using (RP)-12uu instead of (RP)-7tt.
Example 276. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Rp)-42au].
[00593] Crude (RP)-42au is produced as described in Example 267 using (RP)-12au instead of (RP)-7tt.
142 Example 277. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Rp)-42cu].
[00594] Crude (RP)-42cu is produced as described in Example 267 using (RP)-12cu instead of (RP)-7tt.
Example 278. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Rp)-42gu].
[00595] Crude (RP)-42gu is produced as described in Example 267 using (RP)-12gu instead of (RP)-7tt.
Example 279. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphonate [(Sp)-42uu].
[00596] Crude (SP)-42uu is produced as described in Example 267 using (SP)-12uu instead of (RP)-7tt.
Example 280. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Sp)-42au].
[00597] Crude (SP)-42au is produced as described in Example 267 using (SP)-12au instead of (RP)-7tt.
Example 281. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Sp)-42cu].
[00598] Crude (SP)-42cu is produced as described in Example 267 using (SP)-12au instead of (RP)-7tt.
Example 282. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Sp)-42gu].
[00599] Crude (SP)-42gu is produced as described in Example 267 using (SP)-12gu instead of (RP)-7tt.
Example 283. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphonate [(Rp)-43uu].
[00600] Crude (RP)-43uu is produced as described in Example 267 using (RP)-15uu instead of (RP)-7tt.
Example 284. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Rp)-43au].
[00601] Crude (RP)-43au is produced as described in Example 267 using (SP)-15au instead of (RP)-7tt.
Example 285. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Rp)-43cu].
[00602] Crude (RP)-43cu is produced as described in Example 267 using (SP)-15cu instead of (RP)-7tt.
Example 286. Synthesis of the alkylhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl H-phosphonate [(Rp)-43gu].
[00603] Crude (RP)-43gu is produced as described in Example 267 using (SP)-15gu instead of (RP)-7tt.
Example 287. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphonate [(Sp)-43uu].
[00604] Crude (SP)-43uu is produced as described in Example 267 using (SP)-15uu instead of (RP)-7tt.
Example 288. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Sp)-43au].
[00605] Crude (SP)-43au is produced as described in Example 267 using (SP)-15au instead of (RP)-7tt.
143 Example 289. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Sp)-43cu].
[00606] Crude (SP)-43cu is produced as described in Example 267 using (SP)-15cu instead of (RP)-7tt.
Example 290. Synthesis of the alkylhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphonate [(Sp)-43gu].
[00607] Crude (SP)-43gu is produced as described in Example 267 using (SP)-15gu instead of (RP)-7tt.
Scheme R: Synthesis of acylhydroxamate nucleic acid prodrugs.

T a 0=~ i K/b a H^/CON AOMe d f O 0. O
Ftõ - - d F~' O'V~
CH3CN pyridine OTBS OTBS

At iK/19 a Et3N3 HF _r~
O O `0. O
C, 'C'V~
44tt OH
Example 291. Synthesis of the acylhydroxamate nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-44tt] as described in Scheme R.
[00608] (RP)-5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tent-butyldimethylsilyl)thymidin-5'-yl H-phosphonate [(RP)-7tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with N-acyloxy-N-methyl-3-hydroxypropionamide (100 mol) in dry (100 mol) pyridine.
After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L).
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M
ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-44tt.
Example 292. Synthesis of the acylhydroxamate pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Rp)-44at].
[00609] Crude (RP)-40at is produced as described in Example 291 using (RP)-7at instead of (RP)-7tt.
Example 293. Synthesis of the acylhydroxamate pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Rp)-44ct].
[00610] Crude (RP)-44ct is produced as described in Example 291 using (RP)-7ct instead of (RP)-7tt.
144 Example 294. Synthesis of the acylhydroxamate pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Rp)-44gt].
[00611] Crude (RP)-44gt is produced as described in Example 291 using (RP)-7g instead of (RP)-7tt.
Example 295. Synthesis of the acylhydroxamate pronucleotide of (Sp)- thymidin-3'-yl thymidin-5'-yl phosphonate [(SP)-44tt].
[00612] Crude (SP)-44tt is produced as described in Example 291 using (SP)-7tt instead of (RP)-7tt.
Example 296. Synthesis of the acylhydroxamate pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-44at].
[00613] Crude (SP)-44at is produced as described in Example 291 using (SP)-7at instead of (RP)-7tt.
Example 297. Synthesis of the acylhydroxamate pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphonate [(Sp)-44ct].
[00614] Crude (SP)-44ct is produced as described in Example 291 using (SP)-7ct instead of (RP)-7tt.
Example 298. Synthesis of the acylhydroxamate pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphonate [(Sp)-44gt].
[00615] Crude (SP)-44gt is produced as described in Example 291 using (SP)-7gt instead of (RP)-7tt.
Example 299. Synthesis of the acylhydroxamate pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphonate [(Rp)-44uu].
[00616] Crude (RP)-44uu is produced as described in Example 291 using (RP)-12uu instead of (RP)-7tt.
Example 300. Synthesis of the acylhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Rp)-44au].
[00617] Crude (RP)-44au is produced as described in Example 291 using (RP)-12au instead of (RP)-7tt.
Example 301. Synthesis of the acylhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Rp)-44cu].
[00618] Crude (RP)-44cu is produced as described in Example 291 using (RP)-12cu instead of (RP)-7tt.
Example 302. Synthesis of the acylhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Rp)-44gu].
[00619] Crude (RP)-40gu is produced as described in Example 291 using (RP)-12gu instead of (RP)-7tt.
Example 303. Synthesis of the acylhydroxamate pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphonate [(Sp)-44uu].
[00620] Crude (SP)-44uu is produced as described in Example 291 using (SP)-12uu instead of (RP)-7tt.
Example 304. Synthesis of the acylhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphonate [(Sp)-44au].
[00621] Crude (SP)-44au is produced as described in Example 291 using (SP)-12au instead of (RP)-7tt.
Example 305. Synthesis of the acylhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphonate [(Sp)-44cu].
[00622] Crude (SP)-44cu is produced as described in Example 291 using (SP)-12au instead of (RP)-7tt.
145 Example 306. Synthesis of the acylhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphonate [(Sp)-44gu].
[00623] Crude (SP)-44gu is produced as described in Example 291 using (SP)-12gu instead of (RP)-7tt.
Example 307. Synthesis of the acylhydroxamate pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphonate [(Rp)-45uu].
[00624] Crude (RP)-45uu is produced as described in Example 291 using (RP)-15uu instead of (RP)-7tt.
Example 308. Synthesis of the acylhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Rp)-45au].
[00625] Crude (RP)-45au is produced as described in Example 291 using (SP)-15au instead of (RP)-7tt.
Example 309. Synthesis of the acylhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Rp)-45cu].
[00626] Crude (RP)-45cu is produced as described in Example 291 using (SP)-15cu instead of (RP)-7tt.
Example 310. Synthesis of the acylhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl H-phosphonate [(Rp)-45gu].
[00627] Crude (RP)-45gu is produced as described in Example 291 using (SP)-15gu instead of (RP)-7tt.
Example 311. Synthesis of the acylhydroxamate pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphonate [(Sp)-45uu].
[00628] Crude (SP)-45uu is produced as described in Example 291 using (SP)-15uu instead of (RP)-7tt.
Example 312. Synthesis of the acylhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphonate [(Sp)-45au].
[00629] Crude (SP)-45au is produced as described in Example 291 using (SP)-15au instead of (RP)-7tt.
Example 313. Synthesis of the acylhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphonate [(Sp)-45cu].
[00630] Crude (SP)-45cu is produced as described in Example 291 using (SP)-15cu instead of (RP)-7tt.
Example 314. Synthesis of the acylhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphonate [(Sp)-45gu].
[00631] Crude (SP)-45gu is produced as described in Example 291 using (SP)-15gu instead of (RP)-7tt.
Scheme S: Synthesis of thiotrialkylammoniumethyl pronucleotides.
146 TBDP'~" q TBDPS
O D+ Me3f~1~ j ~
-SI PC, O cf OTBS OTBS
4tt HCr~
Et3N3 HF

e 'II

46tt OH
Example 315. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-46tt] as described in Scheme S.
[00632] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry methylene chloride (1 mL). The mixture is treated with vinyltrimethylammonium chloride (100 mol) in dry (100 mol) methylene chloride. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-46tt.
Example 316. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-46at].
[00633] Crude (RP)-46at is produced as described in Example 315 using (RP)-4at instead of (RP)-4tt.
Example 317. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-46ct].
[00634] Crude (RP)-46ct is produced as described in Example 315 using (RP)-4ct instead of (RP)-4tt.
Example 318. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-46gt].
[00635] Crude (RP)-46gt is produced as described in Example 315 using (RP)-4g instead of (RP)-4tt.
Example 319. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Se)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-46tt].
[00636] Crude (SP)-46tt is produced as described in Example 315 using (SP)-4tt instead of (RP)-4tt.
Example 320. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-46at].
147 [00637] Crude (SP)-46at is produced as described in Example 315 using (SP)-4at instead of (RP)-4tt.
Example 321. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-46ct].
[00638] Crude (SP)-46ct is produced as described in Example 315 using (SP)-4ct instead of (RP)-4tt.
Example 322. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-46gt].
[00639] Crude (SP)-46gt is produced as described in Example 315 using (SP)-4gt instead of (RP)-4tt.
Example 323. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-46uu].
[00640] Crude (RP)-46uu is produced as described in Example 315 using (RP)-10uu instead of (RP)-4tt.
Example 324. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-46au].
[00641] Crude (RP)-46au is produced as described in Example 315 using (RP)-10au instead of (RP)-4tt.
Example 325. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-46cu].
[00642] Crude (RP)-46cu is produced as described in Example 315 using (RP)-10cu instead of (RP)-4tt.
Example 326. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-46gu].
[00643] Crude (RP)-46gu is produced as described in Example 315 using (RP)-10gu instead of (RP)-4tt.
Example 327. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-46uu].
[00644] Crude (SP)-46uu is produced as described in Example 315 using (SP)-10uu instead of (RP)-4tt.
Example 328. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-46au].
[00645] Crude (SP)-46au is produced as described in Example 315 using (SP)-10au instead of (RP)-4tt.
Example 329. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-46cu].
[00646] Crude (SP)-46cu is produced as described in Example 315 using (SP)-10au instead of (RP)-4tt.
Example 330. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-46gu].
[00647] Crude (SP)-46gu is produced as described in Example 315 using (SP)-10gu instead of (RP)-4tt.
Example 331. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-47uu].
[00648] Crude (SP)-47uu is produced as described in Example 315 using (SP)-14uu instead of (RP)-4tt.
Example 332. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-47au].
148 [00649] Crude (RP)-47au is produced as described in Example 315 using (SP)-14au instead of (RP)-4tt.
Example 333. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-47cu].
[00650] Crude (RP)-47cu is produced as described in Example 315 using (SP)-14cu instead of (RP)-4tt.
Example 334. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-47gu].
[00651] Crude (RP)-47gu is produced as described in Example 315 using (SP)-14gu instead of (RP)-4tt.
Example 335. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-47uu].
[00652] Crude (SP)-47uu is produced as described in Example 315 using (SP)-14uu instead of (RP)-4tt.
Example 336. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-47au].
[00653] Crude (SP)-47au is produced as described in Example 315 using (SP)-14au instead of (RP)-4tt.
Example 337. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-47cu].
[00654] Crude (SP)-47cu is produced as described in Example 315 using (SP)-14cu instead of (RP)-4tt.
Example 338. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-47gu].
[00655] Crude (SP)-47gu is produced as described in Example 315 using (SP)-14gu instead of (RP)-4tt.
Scheme T: Synthesis of thio N-alkylhydroxamate pronucleotides.

TBDP'~" nnL.-I rf XVb nnL- / C'V

-5,F~' 0 O S, Fe. 0 OTBS OTBS
4tt rv Et3N3 HF
0 eFICO
48tt OH
Example 339. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-48tt] as described in Scheme T.
[00656] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry methylene chloride (1 mL). The mixture is treated with N,O-dimethylacrylamide (100 mol) in dry (100 mol) methylene chloride. After 1 hour, the
149 mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M
ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-48tt.
Example 340. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-48at].
[00657] Crude (RP)-48at is produced as described in Example 339 using (RP)-4at instead of (RP)-4tt.
Example 341. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-48ct].
[00658] Crude (RP)-48ct is produced as described in Example 339 using (RP)-4ct instead of (RP)-4tt.
Example 342. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-48gt].
[00659] Crude (RP)-48gt is produced as described in Example 339 using (RP)-4g instead of (RP)-4tt.
Example 343. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-48tt].
[00660] Crude (SP)-48tt is produced as described in Example 339 using (SP)-4tt instead of (RP)-4tt.
Example 344. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-48at].
[00661] Crude (SP)-48at is produced as described in Example 339 using (SP)-4at instead of (RP)-4tt.
Example 345. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-48ct].
[00662] Crude (SP)-48ct is produced as described in Example 339 using (SP)-4ct instead of (RP)-4tt.
Example 346. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-48gt].
[00663] Crude (SP)-48gt is produced as described in Example 339 using (SP)-4gt instead of (RP)-4tt.
Example 347. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-48uu].
[00664] Crude (RP)-48uu is produced as described in Example 339 using (RP)-10uu instead of (RP)-4tt.
Example 348. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-48au].
[00665] Crude (RP)-48au is produced as described in Example 339 using (RP)-10au instead of (RP)-4tt.
Example 349. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-48cu].
[00666] Crude (RP)-48cu is produced as described in Example 339 using (RP)-10cu instead of (RP)-4tt.
150 Example 350. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-48gu].
[00667] Crude (RP)-48gu is produced as described in Example 339 using (RP)-10gu instead of (RP)-4tt.
Example 351. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-48uu].
[00668] Crude (SP)-48uu is produced as described in Example 339 using (SP)-10uu instead of (RP)-4tt.
Example 352. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-48au].
[00669] Crude (SP)-48au is produced as described in Example 339 using (SP)-10au instead of (RP)-4tt.
Example 353. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-48cu].
[00670] Crude (SP)-48cu is produced as described in Example 339 using (SP)-10au instead of (RP)-4tt.
Example 354. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-48gu].
[00671] Crude (SP)-48gu is produced as described in Example 339 using (SP)-10gu instead of (RP)-4tt.
Example 355. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-49uu].
[00672] Crude (SP)-49uu is produced as described in Example 339 using (SP)-14uu instead of (RP)-4tt.
Example 356. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-49au].
[00673] Crude (RP)-49au is produced as described in Example 339 using (SP)-14au instead of (RP)-4tt.
Example 357. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-49cu].
[00674] Crude (RP)-49cu is produced as described in Example 339 using (SP)-14cu instead of (RP)-4tt.
Example 358. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-49gu].
[00675] Crude (RP)-49gu is produced as described in Example 339 using (SP)-14gu instead of (RP)-4tt.
Example 359. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-49uu].
[00676] Crude (SP)-49uu is produced as described in Example 339 using (SP)-14uu instead of (RP)-4tt.
Example 360. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-49au].
[00677] Crude (SP)-49au is produced as described in Example 339 using (SP)-14au instead of (RP)-4tt.
Example 361. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-49cu].
[00678] Crude (SP)-49cu is produced as described in Example 339 using (SP)-14cu instead of (RP)-4tt.
151 Example 362. Synthesis of the thio N-alkylhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-49gu].
[00679] Crude (SP)-49gu is produced as described in Example 339 using (SP)-14gu instead of (RP)-4tt.
Scheme U: Synthesis of thio N-acetoxyhydroxamate pronucleotides.

TBDP'~" Mew Nr O4c K/L_ OTBS OTBS
4tt rv Et3N3 HF
O SPAO
Mt OH
Example 363. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-50tt] as described in Scheme U.
[00680] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(tent-butyldiphenylsilyl)thymidin-3'-yl 3'-O-(tert-butyldimethylsilyl)thymidin-5'-yl phosphorothioate [(SP)-4tt] (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry methylene chloride (1 mL). The mixture is treated with N-methyl-N-acetoxy-acrylamide (100 mol) in dry (100 mol) methylene chloride. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-50tt.
Example 364. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-50at].
[00681] Crude (RP)-50at is produced as described in Example 363 using (RP)-4at instead of (RP)-4tt.
Example 365. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-50ct].
[00682] Crude (RP)-50ct is produced as described in Example 363 using (RP)-4ct instead of (RP)-4tt.
Example 366. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Rp)-50gt].
[00683] Crude (RP)-50gt is produced as described in Example 363 using (RP)-4g instead of (RP)-4tt.
152 Example 367. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(SP)-50tt].
[00684] Crude (SP)-50tt is produced as described in Example 363 using (SP)-4tt instead of (RP)-4tt.
Example 368. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-6-N-benzoyl-deoxyadenosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-50at].
[00685] Crude (SP)-50at is produced as described in Example 363 using (SP)-4at instead of (RP)-4tt.
Example 369. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-4-N-benzoyl-deoxycytidin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-50ct].
[00686] Crude (SP)-50ct is produced as described in Example 363 using (SP)-4ct instead of (RP)-4tt.
Example 370. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-2-N-phenoxyacetyl-deoxyguanosin-3'-yl thymidin-5'-yl phosphorothioate [(Sp)-50gt].
[00687] Crude (SP)-50gt is produced as described in Example 363 using (SP)-4gt instead of (RP)-4tt.
Example 371. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-50uu].
[00688] Crude (RP)-50uu is produced as described in Example 363 using (RP)-10uu instead of (RP)-4tt.
Example 372. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-50au].
[00689] Crude (RP)-50au is produced as described in Example 363 using (RP)-10au instead of (RP)-4tt.
Example 373. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-50cu].
[00690] Crude (RP)-50cu is produced as described in Example 363 using (RP)-10cu instead of (RP)-4tt.
Example 374. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Rp)-50gu].
[00691] Crude (RP)-50gu is produced as described in Example 363 using (RP)-10gu instead of (RP)-4tt.
Example 375. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-uridin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-50uu].
[00692] Crude (SP)-50uu is produced as described in Example 363 using (SP)-10uu instead of (RP)-4tt.
Example 376. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-50au].
[00693] Crude (SP)-50au is produced as described in Example 363 using (SP)-10au instead of (RP)-4tt.
Example 377. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-50cu].
[00694] Crude (SP)-50cu is produced as described in Example 363 using (SP)-10au instead of (RP)-4tt.
Example 378. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-3'-yl uridin-5'-yl phosphorothioate [(Sp)-50gu].
[00695] Crude (SP)-50gu is produced as described in Example 363 using (SP)-10gu instead of (RP)-4tt.
153 Example 379. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Rp)-5luu].
[00696] Crude (SP)-5 luu is produced as described in Example 363 using (SP)-14uu instead of (RP)-4tt.
Example 380. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-51 au].
[00697] Crude (RP)-5lau is produced as described in Example 363 using (SP)-14au instead of (RP)-4tt.
Example 381. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(RP)-51cu].
[00698] Crude (RP)-5lcu is produced as described in Example 363 using (SP)-14cu instead of (RP)-4tt.
Example 382. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Rp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(RP)-51gu].
[00699] Crude (RP)-5lgu is produced as described in Example 363 using (SP)-14gu instead of (RP)-4tt.
Example 383. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-uridin-2'-yl uridin-5'-yl phosphorothioate [(Sp)-5luu].
[00700] Crude (SP)-5 luu is produced as described in Example 363 using (SP)-14uu instead of (RP)-4tt.
Example 384. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-6-N-Benzoyl-adenosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-51 au].
[00701] Crude (SP)-5lau is produced as described in Example 363 using (SP)-14au instead of (RP)-4tt.
Example 385. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-4-N-Benzoyl-cytidin-2'-yl uridin-5'-yl phosphorothioate [(SP)-51cu].
[00702] Crude (SP)-5lcu is produced as described in Example 363 using (SP)-l4cu instead of (RP)-4tt.
Example 386. Synthesis of the thio N-acetoxyhydroxamate pronucleotide of (Sp)-2-N-Phenoxyacetyl-guanosin-2'-yl uridin-5'-yl phosphorothioate [(SP)-51gu].
[00703] Crude (SP)-5lgu is produced as described in Example 363 using (SP)-14gu instead of (RP)-4tt.
Scheme V: Synthesis of thiotrialkylammoniumethyl pronucleotides.

DMTr 0 a I O O DMTr 0 a ~/I I +
S" IO Me3N /D
O/F=.,,.,0 Me Me a 0 "0 a 52tt DMTrO DMTrO
HO a C130002H 1 S\ 100010 Mw Or--/ O/R "'0 Me a 53tt HO
154 Example 387. Synthesis of the thiotrialkylammoniumethyl pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-53tt] as described in Scheme V.
[00704] (SP)-1,8-Diazabicyclo[5.4.0]undec-7-enium 5'-O-(dimethoxytrity)thymidin-3'-yl 3'-0-(dimethoxytrity)thymidin-5'-yl phosphorothioate [(SP)-52tt] is prepared by the same method used for the preparation of compound 4tt in Example 1 (Scheme A). Compound (SP)-52tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry dimethylformamide (1 mL). The mixture is treated with 2-iodoethyl trimethylammonium iodide (100 mol) in dry DMF (0.5 mL). After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added. The mixture is stirred for 15 h at room temperature. A 0.1 M
ammonium acetate buffer (2.5 mL) is then added to the mixture and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-53tt.
Scheme W: Synthesis of thiotrialkylammoniumethyl pronucleotides.

DMTr O a DMTr O a _S O erg" 1*0 O"O a 0 a 52tt DMTrO DMTrO
HO a C130002H _ S\ ,# o O O a 54tt HO
Example 388. Synthesis of the disulfide pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-54tt] as described in Scheme W.
[00705] Compound (SP)-52tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry ethanol (1 mL). The mixture is treated with p-nitrobenezene sulfenyl chloride (200 mol) in dry (100 mol) ethanol. After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added. The mixture is stirred for 15 h at room temperature. A 0.1 M
ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified
155 by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-54tt.
Scheme X: Synthesis of 2-thiopivalylethyl nucleic acid prodrugs.
e DMTr HO 'S
0-yj a S Me DMTrOa _YOj H,, oO NCS 0 o i0 oR'v a Me ~- O O~ CH3CN pyridine MO Oja ODMTr ODMTr 55tt HO -t,,::J

O 0. ,O
Me OO a Me Me 56tt OH
Example 389. Synthesis of the 2-thiopivalylethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-56tt] as described in Scheme X.
[00706] (RP)-5'-O-(dimethoxytrityl)thymidin-3'-yl 3'-O-(dimethoxytrityl)thymidin-5'-yl H-phosphonate [(RP)-55tt] is prepared by the same method used for the preparation of compound 7tt in Example 8 (Scheme B). Compound (RP)-55tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-Chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with 2-hydroxyethylthiopivalate (100 mol) in dry (100 mol) pyridine.
After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added. The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et2O (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH
7.0)] to afford (RP)-56tt.
156 Scheme Y: Synthesis of 2-carboethoxyethyl nucleic acid prodrugs.
DMTrO_OY~a HO Q p DMTrO O a OD H,, ,O NCS DO O. ~O
oR=,, a '' O O~ CH3CN pyridine O O~
ODMTr ODMTr 55tt Ha DO O. O
"o-yj a 57tt OH
Example 390. Synthesis of the 2-carboethoxyethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-57tt] as described in Scheme Y.
[00707] Compound (RP)-55tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-Chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with ethyl 2-hydroxyethylpropionate (100 mol) in dry (100 mol) pyridine. After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added.
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et2O (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-57tt.
Scheme Z: Synthesis of thio(cyclohexyl)acyloxy pronucleotides.
DMTr O a O^CI DMTr O a O
s._ ~`S
0 OOH "O
a O'~R' O a ODMTr ODMTr 52tt Ha S~ ,O
eR""O a 58tt OH
157 Example 391. Synthesis of the thio(cyclohexyl)acyloxy pronucleotide of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphorothioate [(RP)-58tt] as described in Scheme Z.
[00708] Compound (SP)-52tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry methylene chloride (1 mL). The mixture is treated with chloromethylcyclohexylacetic acetate (100 mol) in dry (100 mol) methylene chloride. After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added. The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M
ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-58tt.
Scheme AA: Synthesis of 2-carboxyethyl nucleic acid prodrugs.
DMTrO1Y a HO o p DMTrO O a OtBu H,~ ,O NCS tBuO `O'~ '0O
ir'R=W a =4 a o O CH3CN pyridine O O
ODMTr ODMTr 55tt HOa HO 0. O

OAR ""O_';~j a 59tt OH
Example 392. Synthesis of the 2-carboxyethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-59tt] as described in Scheme AA.
[00709] Compound (RP)-55tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-Chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with tert-butyl 2-hydroxyethylpropionate (100 mol) in dry (100 mol) pyridine. After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added.
The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et2O (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-59tt.
158 Scheme BB: Synthesis of 2-((2-hydroxyethyl)disulfide)ethyl nucleic acid prodrugs.
TBDPSO,,/.
a TBDPS 0 a S TBDPSO ,J
HO^~
H~ '00 NCS 0~ '*O
iP' a ~'' O O~ CH3CN pyridine TBDPSO-~ O O-j,,,Ja OTBS OTBS
7tt Ha Et3N-3 HF

O P"
HO O
_';~j a 60tt OH
Example 393. Synthesis of the 2-((2-hydroxyethyl)disulfide)ethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-60tt] as described in Scheme BB.
[00710] Compound (RP)-7tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-Chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with 2-((2-(tert-butyldiphenylsilyloxy)ethyl)disulfanyl)ethanol (100 mol) in dry (100 mol) pyridine. After 1 hour, the mixture is concentrated and then dissolved in triethylamine trihydrofluoride (500 L). The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et20 (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-60tt.
Scheme CC: Synthesis of 2-(methanesulfonothioate)ethyl nucleic acid prodrugs.
DMTr a ^/S\ ~~0 DMTrO a HO OS-Me yS
H\ AO NCS 0 0 0\ .00 OAP"'O a Me O O'O a ~O~ CH3CN pyridine _1;Oj ODMTr ODMTr 55tt HO a CI3000ZH _Ir_j 0~* _0. i0 Me `O eF""'0 a 61tt OH
159 Example 394. Synthesis of the 2-(methanesulfonothioate)ethyl nucleic acid prodrug of (Rp)-thymidin-3'-yl thymidin-5'-yl phosphonate [(RP)-61tt] as described in Scheme CC.
[00711] Compound (RP)-55tt (100 mol) is dried by repeated coevaporations with dry pyridine and then dissolved in dry pyridine (1 mL). N-Chlorosuccinimide (0.1 mmol) is added, and the mixture is stirred for 2 hours at 0 C. The mixture is concentrated and dissolved in dry pyridine (1 mL). The above mixture is treated with S-2-hydroxyethyl methanesulfonothioate (100 mol) in dry (100 mol) pyridine. After 1 hour, the mixture is concentrated and then dissolved in CH2C12 (1000 L) and trichloroacetic acid (50 mol) is added. The mixture is stirred for 15 h at room temperature. A 0.1 M ammonium acetate buffer (2.5 mL) is then added to the mixture, and the mixture is washed with Et2O (3 x 3 mL). The combined organic layers are back-extracted with 0.1 M ammonium acetate buffer (3 mL). The combined aqueous layers are then concentrated to dryness under reduced pressure, and the residue is purified by reverse-phase column chromatography [a linear gradient of acetonitrile 0-10% in 0.1 M ammonium acetate buffer (pH 7.0)] to afford (RP)-61tt.
Scheme DD: Synthesis of Prodrug molecules from H-Phosphonate Thymidine dimer [00712] Chemoselectivity and stereospecificity of iodine mediated oxidative couplings using separate diastereomers of dinucleoside H-phosphonate and 0- nucleophiles to prepare phosphotriesters are known in the art (Nucleosides, Nucleotides & Nucleic Acids 2003 Vol. 22, Nos. 5-8, 1467-1469). The products were purified by silica gel chromatography and characterized by 31P and 1H NMR
spectroscopy. The products were further purified by reverse phase HPLC for kinetic studies.

Scheme DD
O
NH NH

H:0NO
N DMTrO 0 DMTrO O;PO N`0 0,P N"~O 0 O

O R-S R-S
HO
DMTrO DMTrO

63a R=SCH2CH2OTBDMS 64a R=SCH2CH2OH
62 63b R=S02CH3 64b R=S02CH3 63c R=SCO(CH3)3 64c R=SCO(CH3)3 i) I2, ACN: Py (3:2), TBDPSCI and 2-((2-((tent-butyl dimethylsilyl)oxy)ethyl)disulfanyl)ethanol for 63a; 2-hydroxyethyl mehanesulfonate for 63b and 63c) 2-Hydroxyethylthiopivalate ii) 3% DCA/DCM
160 Example 395. General Procedure for Synthesis of 63a, 63b and 63c (Scheme DD) [00713] (RP,SP)-5'-O-(4,4'-dimethoxytrityl)thymidin-3'-yl 3'-O-(4,4'-dimethoxytrityl)thymidin-5'-yl H-phosphonate (62) (113.5 mg, 100 mol) was dried under high vacuum overnight and dissolved in ACN (2 ml) and pyridine (2 ml). tert-Butyldiphenylsilyl chloride (52 L, 200 mol) and 12 (76 mg, 300 moles) were added. The reaction mixture was cooled in ice and respective alkylating reagent (1 mmol) dissolved in ACN (2 mL) was added drop wise to reaction mixture. The mixture was stirred for 10 min under argon. TLC
of the crude reaction mixture showed quantitative conversion to product. The solvents were evaporated and residue was dissolved in ethyl acetate and washed with 5% Na2S203, brine and dried over Na2SO4. The ethyl acetate layer was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to afford (RP,SP)-5'-O-(4,4'-dimethoxytrityl)thymidin-3'-yl 3'-O-(4,4'-dimethoxytrityl)thymidin-5'-yl phosphotriester 63a, 63b and 63c in 80-90%
yield.

General Procedure for Synthesis of 64a, 64b and 64c [00714] 3% DCA/DCM was added slowly to DMTr protected triester and reaction was left for stirring at room temperature for 30min. Reaction was quenched with methanol, solvents were evaporated and residue was purified by silica column. In case of compound 63a, TBDMS deprotection occurred simultaneously.
Compounds 64a, 64b and 64c were obtained in quantitative yields.
[00715] Compound 63a: 1H NMR (400 MHz, CDC13) 6 7.58-7.18 (m, 20H), 6.87-6.78 (m, 8H), 6.49-6.27 (m, 2H), 5.11-5.07 (m, 1H), 4.2-4.05 (m, 3H), 3.99-3.87 (m, 1H), 3.85-3.73 (m, 13H), 3.73-3.56 (m, 1H), 3.54-3.26 (m, 2H), 2.94-2.66 (m, 8H), 1.97-1.78(m, 4H), 1.76-1.54 (m, 1H), 1.43-1.3 (m, 3H), 0.94-0.78 (m, 9H), 0.11-0.03 (m, 6H). 31P NMR (162 MHz, CDC13) 6 -1.19, -1.26 (two diastereomers).
[00716] Compound 63b: 1H NMR (400 MHz, CDC13 + trace amount of Py-D5) 6 9.72-9.45 (m, 2H), 7.62-7.18 (m, 20H), 6.90-6.81 (m, 8H), 6.48-6.31 (m, 2H), 5.18-5.10 (m, 1H), 4.31-4.09 (m, 3H), 4.03-3.91 (m, 1H), 3.88-3.74 (m, 15H), 3.74-3.62 (m, 1H), 3.54-3.31 (m, 2H), 2.89-2.76 (m, 4H), 2.67-2.31 (m, 2H), 1.98-1.89 (m, 1H), 1.88, 1.85 (2s, 3H, diastereomers), 1.76-1.64 (m, 1H), 1.39 (s, 3H). 31P NMR (162 MHz, CDC13) 6 -1.24, -1.27 (two diastereomers).
[00717] Compound 63c: 1H NMR (400 MHz, CDC13 + trace amount of Py-D5) 6 7.6-7.16 (m, 20H), 6.9-6.77 (m, 8H), 6.49-6.27 (m, 2H), 5.18-5.06 (m, 1H), 4.32-4.04 (m, 2H), 4.0-3.85 (m 3H), 3.82-3.71 (m, 12H), 3.71-3.57 (m, 1H), 3.55-3.27 (m, 2H), 3.07-2.88 (m, 2H), 2.62-2.24 (m, 2H), 1.97-1.89 (m, 1H), 1.89-1.81 (m, 3H), 1.78-1.59 (m, 3H), 1.45-1.32 (m, 3H), 1.22-1.14 (m, 9H). 31P NMR
(162 MHz, CDC13) 6 -1.23, -1.27 (two diastereomers).
[00718] Compound 64a: 1H NMR (400 MHz, D20) 6 7.52 (s, 1H), 7.42 (s, 1H), 6.28-6.07 (m, 2H), 5.07-4.87 (m, 1H), 4.54-4.38 (m, 1H), 4.37-4.19 (m, 3H), 4.19-3.95 (m, 2H), 3.80-3.50 (m, 4H), 2.99-2.62 (2m, 4H), 2.60-2.17 (m, 4H), 1.85-1.60 (m, 6H). 31P NMR (162 MHz, CD3OD) 6 -1.37, -1.41 (two diastereomers). Calculated mass = 682.66, Observed mass in ESI-ve mode =
681.22 [00719] Compound 64b: 1H NMR (400 MHz, CD3OD) 6 7.74, 7.49 (2s, 2H), 6.28-6.19 (m, 2H), 5.10-5.04 (m, 1H), 4.41-4.23 (m, 4H), 4.19-4.15 (m, 1H), 4.04-3.98 (m, 1H), 3.78-3.69 (m, 4H), 3.00-2.77 (m, 4H),
161 2.55-2.21 (m, 4H), 1.86, 1.83 (2s, 6H). 31P NMR (162 MHz, CD3OD) 6 -1.37, -1.41 (two diastereomers).
Calculated mass = 684.63, Observed mass in ESI+ve mode = 683.14 [00720] Compound 64c: 1H NMR (400 MHz, CD3OD) 6 7.799 (s, 1H), 7.54 (s, 1H), 6.32-6.24 (m, 2H), 5.17-5.07 (m, 1H), 4.48-4.26 (m, 3H), 4.26-4.12 (m, 3H), 4.08-3.99 (m, 1H), 3.21-3.15 (m, 2H), 2.61-2.48 (m, 1H), 2.46-2.16 (m, 3H), 1.9 (s, 3H), 1.87 (s, 3H), 1.27-1.19 (d, 9H). 31P
NMR (162 MHz, CD3OD) 6 -1.53, -1.60 (two diastereomers). Calculated mass = 690.66, Observed mass in ESI-ve mode = 689.53 HPLC purification of 64a, 64b and 64c [00721] Reverse phase purification was carried out using Waters 2525 BGM
combined with 2487 UV
detector, Phenomenex Luna 5u C18 (2) 100A, 250x10 mm column and MassLynx v4.1.
A gradient of Water and acetonitrile was used with flow rate of 5ml/min.
[00722] Gradient used for compound 64a and 64b: 10 to 50% B in 30 min [00723] Gradient used for compound 64c: 20 to 60% B in 30 min [00724] The product peaks were monitored at 254 and 280 nm.
Analytical HPLC conditions [00725] Quantitative analysis was done by reverse-phase HPLC employing an automated Alliance Waters e2695 HPLC instrument in combination with Empower software. An XBridge C18 3.5um, 4.6xlSOmm, Waters part# 186003034A was fitted and detection was done by UV (254nm and 280nm). A gradient elution system was developed (Table 1) enabling the resolution of prodrug, intermediate and the released drug within the same chromatogram; mobile phase A consisting of 20 mM ammonium acetate in water; mobile phase B was acetonitrile.
Table 1 Column temperature: 60 C
Time Flow % A % B Curve 0.01 1.00 99.0 1.0 5.00 1.00 99.0 1.0 1 30.00 1.00 75.0 25.0 6 30.50 1.00 10.0 90.0 6 35.00 1.00 10.0 90.0 1 35.50 1.00 99.0 1.0 6 42.00 1.00 99.0 1.0 1 Example 396. Glutathione assisted prodrug release [00726] To 20 L of 64 in water (2 O.D.), 100 L of lOX PBS, and 630 L of H2O
were mixed. Kept the mixture in a hot plate set at 37 C. 250 L of freshly prepared 20 mM reduced L-glutathione was added to above mixture which gave 5 mM GSH concentration in the reaction mixture which is equal to cytosol concentration. 100 l aliquots were take at time intervals of 10 min, 20 min, 30 min, 40 min, 50 min, 60 min, 1.5 hr, 2 hr and 2.5 hr. Each aliquot was immediately quenched with 400 l of 100 mM citrate buffer (pH 4) and analyzed by reversed-phase HPLC and LC/MS.
162 Scheme EE: Mechanism of Glutathione Cleavage HO T HO HO
O O O
Reductive environment a' GSH 0' .-,o P
30- O I p iS0 I

R O O HSJ O RSSG O
CS
HO HO HO
(Sp+ Rp) (Sp+ Rp) PhOS hodiester 64a, R=SCH2CH2OH Intermediate trapped at pH 4 P
64b, R=SO2CH3 and observed by HPLC

LCMS of reaction mixture of compound 64b + GSH at 20 min time point [00727] Waters Acquity UPLC and SDS were used to characterize the products formed during prodrug release. XBridge c18 3.5um, 4.6xl5Omm,Waters part# 186003034 was used with solvent system A: 5 mM
ammonium formate/water and B: acetonitrile with linear gradient as shown in Table 2.
Table 2 Time Flow % A % B Curve 0.0 1.00 99.0 1.0 5.00 1.00 80 20 6 7 1.00 5 95 6 7.5 1.00 99 1 6 9 1.00 99 1 1 [00728] In Figure 1 is provided a representative HPLC profile of compound 64a + GSH.
[00729] In Figure 2 is provided a representative HPLC profile of compound 64a, a glutathione adduct, and the final product after release from the pro-moeity.
[00730] In Figure 3, compounds 64a and 64b show a pseudo first order kinetics because glutathione concentration is in great excess compared to substrate and thus remains effectively constant during the course of reaction. The curves for depleting starting material and forming product are not mirror images because of accumulation of intermediate which is characterized as Glutathione adduct of dinucleoside triester (see Figure 2 and Figure 4).
Example 397. Carboxyesterase assisted cleavage of compound 64c
163 Scheme FF

HO Hp HO
O
O1"P%p Carboxyesterase O I O P'O OII,P%O
_j- 0 O -jr-O
HS /~
-15 O O fj HO CS
HO HO
O

Compound 64c Phosphodiester [00731] Porcine Liver esterase (Sigma Aldrich, product number: E2884) was a suspension in 3.2M
ammonium sulfate pH=8.0, concentration 36 mg protein/mL and 154 units/mg protein. According to product specifications, one unit will hydrolyse 1 M of ethyl butyrate to butyric acid and ethanol per minute at pH=8.0 at 25 C. Compound 64c (0.1O.D, 5.5 nmoles) in 10 L 1XPBS was incubated at 37 C for 10 min.
Serial dilutions of PLE were made in ten vials with conc in units from 1, 10-1, 10-2, 10-3, 10-4, 10-5, 10-6, 10-7, 10-8, 10-9 each in 10 L 1XPBS. The protein solution was incubated at 37 C for 10 min and then each added to ten vials containing compound 64c. The mixtures were stored at 37 C for 30 min and analysed by analytical HPLC and LCMS. The 64c was completely converted to phosphodiester in vials with protein conc. from 1, 10-1 and 10-2. No side reactions were observed. There was no reaction in vials with protein conc. from 10-6 to 10-9. There was some product seen in the vials containing protein conc. 10-3 and 10-4. This suggests that these concentrations are appropriate to study the kinetics of prodrug release using PLE. Time dependent kinetics will be studied using conc. within the range of 10-3 to 10-4 /-6 nmoles of compound 64c.
[00732] Compound 64c (5 O.D., 2.9 moles) dissolved in 900 L of 1XPBS was incubated at 37 C for 10 min. Porcine Liver Esterase (1U) in 100 L 1xPBS was added to above mixture and was stored at 37 C.
Aliquots of 100 L were withdrawn at 0 min, 15 min and 45 min, quenched with 100 L acetonitrile and samples were cooled in ice-bath. The samples were analyzed by UPLC SQD on XBridge C-18 3.5 m, 4.6 x 150 mm, with solvent system A: 5 mM ammonium formate/water and B: acetonitrile with linear gradient as shown in Table 3. At zero minutes, only compound 64c was observed, at 15 min nearly 50% of the product was formed and reaction was complete at 45 minutes. Thus TpT diester 64c was released by carboxyesterase treatment without detectable accumulation of any intermediates.
Table 3 Time Flow %A %B Curve 0.0 1.00 99.0 1.0 2.0 1.00 99.0 1.0 1 7.0 1.00 60.0 40.0 6 9.0 1.00 5.0 95.00 6
164 Time Flow %A %B Curve 9.5 1.00 99.0 1.0 6 11.0 1.00 99.0 1.0 1 Example 398. Treatment of Pancreatic Cancer [00733] A method for treating a subject having pancreatic cancer comprising administering to the subject a therapeutically effective amount of a composition comprising the 2'-5'-A3 S-acetyl-2-thioethyl pronucleotide of Example 242 is contemplated. Treatment is expected to achieve increased tumor inhibition compared with gemcitabine administered as a single agent or gemcitabine and erlotinib administered in combination.
Example 399. Cell Penetration Assay P32"Labeled Nucleic Acid Drugs [00734] Prepare labeled nucleic acid prodrug and parent drug using [32P]dNTP
radionucleotide (Fisher Scientific, Pittsburgh, PA) to synthesize nucleic acid molecules comprising chiral phosphorous moieties and corresponding parent drugs as described herein.
Cell Culture and Penetration Testing [00735] Select culture of either HeLa (adherent human cervical cancer) cells grown in DMEM-10% FBS or BxPC-3 (adherent human pancreas adenocarcinoma) cells grown in 90% RPMI 1640-10% FBS. For plating of cell cultures, trypsinize cells with 0.05% trypsin-EDTA. Assay for viability and cell counting via standard Trypan Blue in phosphate buffered saline (PBS) staining. Dilute cells and seed at 1x105 cells/well in a 6-well format. Incubate at 37 C in a 5% C02 atmosphere for 16 hours or until cells adhere and grow to at least 80% confluency.
[00736] Add labeled prodrug mixture to prodrug experimental wells to achieve final predetermined range of concentrations (e.g., 1 M, 5 M, and 10 M). Add labeled parent drug mixture to parent drug experimental wells to achieve final predetermined range of concentrations (e.g., 1 M, 5 M, and 10 M). Reserve untreated wells for negative control. Incubate cells with experimental treatments for predetermined ranges of time (e.g., 15 minutes, 1 hour, 4 hours, and 8 hours).
P32 Detection and Determination of Prodrug Penetration [00737] To harvest, wash wells 3 times with serum-free media and apply non-denaturing TRIS-HC1 lysis buffer with 1% Triton X100 (Cell Signaling Technology, Inc., Boston, MA) and sonicate briefly. Collect cytosolic and nuclear fractions via standard collection techniques.
[00738] Measurement of drug penetration is performed using standard radiation detection techniques. For detection via scintillation counter, add 50 L of sample to 5 mL of scintillation cocktail and measure beta-emission via liquid scintillation counting. Aliquots of each sample are assayed via Bradford colorimetric assays to normalize radiation counts by total protein concentration.
Example 400. Functional Cell Penetration Assay Using Reporter Gene
165 Assembly of Fusion Gene Vector and Transfection of Cell Line [00739] When nucleic acid prodrugs are used to inhibit specific gene expression, for example, antisense oligonucleotides or antigene oligonucleotides, a functional penetration assay may be desirable. Culture HeLa (adherent human cervical cancer) cells in DMEM-10% FBS. Clone gene of interest into a commercially available vector such as the Living Colors Fluorescent Protein Vector, Clontech, Mountain View, CA.
Transfection of cells with DNA construct and selection for stable transfectants are performed using standard techniques. The result is constitutive expression of gene of interest and a fluorescent reporter (e.g., the protein AcGFP 1).
Nucleic Acid Drugs Inhibiting Specific Gene Expression [00740] Prepare nucleic acid molecules comprising chiral phosphorous moieties and corresponding parent drugs as described herein to disrupt the vector's gene promoter sequence.
Cell Culture and Penetration Testing [00741] Prepare transfected culture by first trypsinizing cells for plating with 0.05% trypsin-EDTA. Assay for viability and cell counting via standard Trypan Blue in phosphate buffered saline (PBS) staining. Dilute cells and seed at 1x105 cells/well in a 6-well format. Incubate at 37 C in a 5% C02 atmosphere for 16 hours or until cells adhere and grow to at least 80% confluency.
[00742] Fluorescent signal is first detected 8-12 hours after transfection.
Add prodrug mixture to prodrug experimental wells to achieve final predetermined range of concentrations (e.g., 1 M, 5 M, and 10 M).
Add parent drug mixture to parent drug experimental wells to achieve final predetermined range of concentrations (e.g., 1 M, 5 M, and 10 M). Reserve untreated wells for negative control. Incubate cells with experimental treatments for predetermined ranges of time (e.g., 15 minutes, 1 hour, 4 hours, and 8 hours).
Reporter Gene Expression Determination of Prodrug Penetration [00743] To harvest, wash each well 3 times with serum-free media and retrypsinize. Measurement of drug penetration is performed using standard fluorescence detection techniques. For qualitative fluorescence measurement with microscopy and quantitative measurement with flow cytometry, use the wavelength that is excitatory for the fluorescent reporter (e.g., 488 nm for ACGFPI).
[00744] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only.
Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
166

Claims (94)

WHAT IS CLAIMED IS:
1. A nucleic acid prodrug having the following structure:

wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c;
Y1 is O, NR d, S, or Se;
R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);
each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
at least one instance of X is -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
R10 is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl;
Z is S or O;
q is 0, 1, or 3;
w is 1, 2, 3, 4, 5, or 6;
R15 and R16 are independently hydrogen or methyl;
R17 is selected from alkyl, aryl or a CH2CH=CH2;

R18 is selected from N(CH3)2, ; and n is an integer of 1 to about 200.
2. The compound of claim 1 wherein each X-phosphonate moiety of the compound of Formula 1 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
3. The compound of claim 1 wherein each X-phosphonate moiety has a R P
configuration.
4. The compound of claim 1 wherein each X-phosphonate moiety has a S p configuration.
5. The compound of claim 1 wherein each X-phosphonate independently has a R P
configuration or a S P
configuration.
6. The compound of claim 1 wherein R10 is methyl.
7. The compound of claim 1 wherein R11 is methyl.
8. The compound of claim 1 wherein R12 is methyl
9. The compound of claim 1, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
10. The compound of claim 1, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
11. The compound of claim 1, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
12. The compound of claim 1, wherein each X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
13. A nucleic acid prodrug having the following structure:

wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c;
Y1 is O, NR d, S, or Se;
R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);
each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;

at least one X is, ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200.
14. The compound of claim 13 wherein R10 is methyl.
15. The compound of claim 13 wherein R11 is methyl.
16. The compound of claim 13 wherein R12 is methyl
17. The compound of claim 13, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
18. The compound of claim 13, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
19. The compound of claim 13, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
20. The compound of claim 13, wherein at each X moiety of the nucleic acid prodrug is independently selected from ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
21. A pharmaceutical composition comprising a nucleic acid prodrug having the following structure:
wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c;
Y1 is O, NR d, S, or Se;

R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);
each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
wherein at least one X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R1 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
and R12 is hydrogen or alkyl;

R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of: (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
22. The pharmaceutical composition of claim 21 wherein each X-phosphonate moiety of the compound of Formula 1 is more than 98% diastereomerically pure as determined by 31P NMR
spectroscopy or reverse-phase HPLC.
23. The pharmaceutical composition of claim 21 wherein each X-phosphonate moiety has a R P
configuration.
24. The pharmaceutical composition of claim 21 wherein each X-phosphonate moiety has a S P
configuration.
25. The pharmaceutical composition of claim 21 wherein each X-phosphonate independently has a R P
configuration or a S p configuration.
26. The compound of claim 21, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
27. The compound of claim 21, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
28. The compound of claim 21, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
29. The pharmaceutical composition of claim 21 wherein each instance of X is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
30. The pharmaceutical composition of claim 21 wherein R10 is methyl.
31. The pharmaceutical composition of claim 21 wherein R1 is methyl.
32. The pharmaceutical composition of claim 21 wherein R12 is methyl.
33. A method of treating a disease associated with upregulated RNase L by administering a therapeutic amount of a chiral nucleic acid prodrug.
34. The method of claim 33, wherein the disease associated with upregulated RNase L is chronic fatigue syndrome.
35. A method of treating a disease associated with downregulated RNase L by administering a therapeutic amount of a chiral nucleic acid prodrug.
36. The method of claim 35, wherein the disease associated with downregulated RNase L is cancer.
37. The method of claim 36, wherein the cancer with downregulated RNase L is pancreatic cancer.
38. A method of treating cancer comprising administering a therapeutic amount of a nucleic acid prodrug having the following structure:

wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c;
Y1 is O, NR d, S, or Se;
R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);
each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
at least one X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
R10 is an alkyl group having 1 to 4 carbon atoms;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of: (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
39. The method of claim 38, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
40. The method of claim 38, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
41. The method of claim 38, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
42. The method of claim 38, wherein each instance of X is independently selected from wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
43. The method of claim 38, wherein R10 is methyl.
44. The method of claim 389, wherein R11 is methyl.
45. The method of claim 38, wherein R12 is methyl
46. The method of claim 38, wherein each X-phosphonate moiety of the compound of Formula 1 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
47. The method of claim 38, wherein each X-phosphonate moiety has a R P
configuration.
48. The method of claim 38, wherein each X-phosphonate moiety has a S p configuration.
49. The method of claim 38, wherein each X-phosphonate independently has a R P
configuration or a S p configuration.
50. The method of claim 38, wherein the cancer is pancreatic cancer.
51. A nucleic acid prodrug having the following structure:

wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c;
Y1 is O, NR d, S, or Se;
R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);
each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
at least one instance of X is -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;
R10 is an alkyl group having 1 to 4 carbon atoms;
R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
R12 is hydrogen or alkyl; and n is an integer of 1 to about 200.
52. The compound of claim 51 wherein each X-phosphonate moiety of the compound of Formula 2 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
53. The compound of claim 51 wherein each X-phosphonate moiety has a R P
configuration.
54. The compound of claim 51 wherein each X-phosphonate moiety has a S P
configuration.
55. The compound of claim 51 wherein each X-phosphonate independently has a R
P configuration or a S p configuration.
56. The compound of claim 51 wherein R10 is methyl.
57. The compound of claim 51 wherein R11 is methyl.
58. The compound of claim 51 wherein R12 is methyl
59. The compound of claim 51, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
60. The compound of claim 51, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
61. The compound of claim 51, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
62. The compound of claim 51, wherein each X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H,
63. A pharmaceutical composition comprising a nucleic acid prodrug having the following structure:
wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c ;
Y1 is O, NR d, S, or Se;

R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);
each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
wherein at least one X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, ; wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl;
and R12 is hydrogen or alkyl;

R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
wherein the method used to synthesize the nucleic acid prodrug comprises the steps of. (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
64. The pharmaceutical composition of claim 63 wherein each X-phosphonate moiety of the compound of Formula 2 is more than 98% diastereomerically pure as determined by 31P NMR
spectroscopy or reverse-phase HPLC.
65. The pharmaceutical composition of claim 63 wherein each X-phosphonate moiety has a R P
configuration.
66. The pharmaceutical composition of claim 63 wherein each X-phosphonate moiety has a S p configuration.
67. The pharmaceutical composition of claim 63 wherein each X-phosphonate independently has a R P
configuration or a S P configuration.
68. The compound of claim 63, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
69. The compound of claim 63, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
70. The compound of claim 63, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
71. The pharmaceutical composition of claim 63 wherein each instance of X is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
72. The pharmaceutical composition of claim 63 wherein R10 is methyl.
73. The pharmaceutical composition of claim 63 wherein R11 is methyl.
74. The pharmaceutical composition of claim 63 wherein R12 is methyl.
75. A method of treating cancer comprising administering a therapeutic amount of a nucleic acid prodrug having the following structure:

wherein R1 is -OH, -SH, -NR d R d, -N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -P(O)(R e)2, -HP(O)(R e), -OR a or -SR
c;
Y1 is O, NR d, S, or Se;
R a is a blocking group;
R c is a blocking group;
each instance of R d is independently hydrogen, alkyl, alkenyl, alkynyl, aryl, acyl, substituted silyl, carbamate, -P(O)(R e)2, or -HP(O)(R e);

each instance of R e is independently hydrogen, alkyl, aryl, alkenyl, alkynyl, alkyl-Y2-, alkenyl-Y2-, alkynyl-Y2-, aryl-Y2-, or heteroaryl-Y2-, or a cation which is Na+1, Li+1, or K+1;
Y2 is O, NR d, or S;
each instance of R2 is independently hydrogen, -OH, -SH, -NR d R d, -N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1-, alkenyl-Y1-, alkynyl-Y1-, aryl-Y1-, heteroaryl-Y1-, -OR b or -SR c, wherein R b is a blocking group;
each instance of Ba is independently a blocked or unblocked adenine, cytosine, guanine, thymine, uracil or modified nucleobase;
at least one X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl;
R3 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid; and n is an integer of 1 to about 200;
R10 is an alkyl group having 1 to 4 carbon atoms;

wherein the method used to synthesize the nucleic acid prodrug comprises the steps of: (1) reacting a molecule comprising an achiral H-phosponate moiety and a nucleoside comprising a 5'-OH moiety to form a condensed intermediate; and (2) converting the condensed intermediate to the nucleic acid prodrug comprising a chiral X-phosphonate moiety.
76. The method of claim 75, wherein at least 25% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
77. The method of claim 75, wherein at least 50% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
78. The method of claim 75, wherein at least 90% of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, ;wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
79. The method of claim 75, wherein each instance of X is independently selected from wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
80. The method of claim 75, wherein R10 is methyl.
81. The method of claim 75 wherein R11 is methyl.
82. The method of claim 75, wherein R12 is methyl
83. The method of claim 75, wherein each X-phosphonate moiety of the compound of Formula 2 is more than 98% diastereomerically pure as determined by 31P NMR spectroscopy or reverse-phase HPLC.
84. The method of claim 75, wherein each X-phosphonate moiety has a R P
configuration.
85. The method of claim 75, wherein each X-phosphonate moiety has a S P
configuration.
86. The method of claim 75, wherein each X-phosphonate independently has a R P
configuration or a S P
configuration.
87. The method of claim 75, wherein the cancer is pancreatic cancer.
88. The method of claim 75 or claim 87, wherein the compound has the following formula:
wherein each A is adenine and each R11 is independently selected from alkyl, aryl, heteroaryl, heterocyclyl, and cycloalkyl.
89. The method of claim 88, wherein the compound has the following formula
90. A compound or its pharmaceutically acceptable salt having the following formula:

wherein each A is adenine; and at least one X moiety is -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
91. The compound of claim 90, wherein at least two of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
92. The compound of claim 90, wherein at least three of the X moieties of the nucleic acid prodrug are independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
93. The compound of claim 90, wherein each X moiety of the nucleic acid prodrug is independently selected from -OCH2CH2S-S(O)2R10, -OCH2CH2S-SCH2CH2OH, -OCH2CH2CO2H, wherein R10 is an alkyl group having 1 to 4 carbon atoms; R11 is alkyl, aryl, heteroaryl, heterocyclyl, or cycloalkyl; and R12 is hydrogen or alkyl.
94. The compound of claim 90, wherein the compound has the following formula:
CA2767253A 2009-07-06 2010-07-06 Novel nucleic acid prodrugs and methods of use thereof Abandoned CA2767253A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US22336909P 2009-07-06 2009-07-06
US61/223,369 2009-07-06
US24272209P 2009-09-15 2009-09-15
US61/242,722 2009-09-15
PCT/US2010/041068 WO2011005761A1 (en) 2009-07-06 2010-07-06 Novel nucleic acid prodrugs and methods use thereof

Publications (1)

Publication Number Publication Date
CA2767253A1 true CA2767253A1 (en) 2011-01-13

Family

ID=43429503

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2767253A Abandoned CA2767253A1 (en) 2009-07-06 2010-07-06 Novel nucleic acid prodrugs and methods of use thereof

Country Status (15)

Country Link
US (2) US9744183B2 (en)
EP (1) EP2451461A4 (en)
JP (2) JP5998326B2 (en)
KR (1) KR101885383B1 (en)
CN (1) CN102596204B (en)
AU (1) AU2010270714B2 (en)
BR (1) BR112012000828A8 (en)
CA (1) CA2767253A1 (en)
CL (1) CL2012000021A1 (en)
IL (1) IL217370A (en)
IN (1) IN2012DN00720A (en)
MX (1) MX342945B (en)
RU (1) RU2612521C2 (en)
SG (2) SG177564A1 (en)
WO (1) WO2011005761A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11390642B2 (en) 2016-08-23 2022-07-19 Dicerna Pharmaceuticals, Inc. Compositions comprising reversibly modified oligonucleotides and uses thereof

Families Citing this family (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103816174B (en) * 2008-04-03 2017-12-05 春堤制药公司 For treating the Compounds and methods for of virus infection
EP2370451B1 (en) 2008-12-02 2016-11-16 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
SG177564A1 (en) 2009-07-06 2012-02-28 Ontorii Inc Novel nucleic acid prodrugs and methods of use thereof
JP5868324B2 (en) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan Asymmetric auxiliary group
RU2014105311A (en) * 2011-07-19 2015-08-27 Уэйв Лайф Сайенсес Пте. Лтд. METHODS FOR SYNTHESIS OF FUNCTIONALIZED NUCLEIC ACIDS
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
SG11201500239VA (en) 2012-07-13 2015-03-30 Wave Life Sciences Japan Asymmetric auxiliary group
SG11201500243WA (en) 2012-07-13 2015-04-29 Shin Nippon Biomedical Lab Ltd Chiral nucleic acid adjuvant
IN2015DN01765A (en) * 2012-08-20 2015-05-29 Univ California
LT2961388T (en) * 2013-03-01 2019-08-26 Astex Pharmaceuticals, Inc. Drug combinations
JPWO2015108046A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antiallergic agent having antiallergic action
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
JPWO2015108047A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant having immunity induction activity and immunity induction activator
DK3094728T3 (en) 2014-01-16 2022-05-16 Wave Life Sciences Ltd KIRALT DESIGN
RU2708237C2 (en) 2014-08-22 2019-12-05 Общество с ограниченной ответственностью "НооГен" Modified oligonucleotides and method for production thereof
WO2016096938A1 (en) 2014-12-16 2016-06-23 Roche Innovation Center Copenhagen A/S Chiral toxicity screening method
WO2016097212A1 (en) 2014-12-17 2016-06-23 Proqr Therapeutics Ii B.V. Targeted rna editing
EP3315507A4 (en) * 2015-06-26 2019-02-27 Kyowa Hakko Kirin Co., Ltd. Oligonucleotide derivative
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
IL258230B (en) 2015-10-09 2022-09-01 Wave Life Sciences Ltd Oligonucleotide compositions and methods thereof
EP3426671A4 (en) * 2016-03-11 2019-11-20 Spring Bank Pharmaceuticals, Inc. Compounds and compositions for the treatment of infections
MA43822A (en) 2016-03-13 2018-11-28 Wave Life Sciences Ltd COMPOSITIONS AND SYNTHESIS OF PHOSPHORAMIDITE AND OLIGONUCLEOTIDES
CN109414511B (en) 2016-04-18 2023-05-23 萨勒普塔医疗公司 Antisense oligomers for treating diseases associated with acid alpha-glucosidase genes and methods of use thereof
EP3449000A1 (en) 2016-04-29 2019-03-06 Sarepta Therapeutics, Inc. Oligonucleotide analogues targeting human lmna
MA45270A (en) 2016-05-04 2017-11-09 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
US11013757B2 (en) 2016-06-03 2021-05-25 Wave Life Sciences Ltd. Oligonucleotides, compositions and methods thereof
US10988763B2 (en) 2016-06-22 2021-04-27 Proqr Therapeutics Ii B.V. Single-stranded RNA-editing oligonucleotides
KR20190024977A (en) 2016-06-30 2019-03-08 사렙타 쎄러퓨틱스 인코퍼레이티드 Exon skipping oligomer for myopathies
CR20190071A (en) * 2016-07-15 2019-10-29 Sperovie Biosciences Inc COMPOUNDS, COMPOSITIONS AND METHODS FOR THE TREATMENT OF DISEASES
ES2837076T3 (en) 2016-09-01 2021-06-29 Proqr Therapeutics Ii Bv Chemically modified oligonucleotides for editing single-stranded RNA
WO2018098264A1 (en) 2016-11-23 2018-05-31 Wave Life Sciences Ltd. Compositions and methods for phosphoramidite and oligonucleotide synthesis
JOP20170192A1 (en) 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co Cyclic dinucleotide
BR112019012651A2 (en) 2016-12-19 2020-01-28 Sarepta Therapeutics Inc exon jump oligomer conjugates for muscular dystrophy
IL297528A (en) 2016-12-19 2022-12-01 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
WO2018118599A1 (en) 2016-12-19 2018-06-28 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
JP7190794B2 (en) 2017-03-29 2022-12-16 塩野義製薬株式会社 Complex of nucleic acid medicine and hyperbranched lipid
WO2018177825A1 (en) * 2017-03-29 2018-10-04 Roche Innovation Center Copenhagen A/S Orthogonal protecting groups for the preparation of stereodefined phosphorothioate oligonucleotides
EP3630789A4 (en) 2017-06-02 2021-06-16 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
US11603532B2 (en) 2017-06-02 2023-03-14 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
EP3642182A4 (en) * 2017-06-21 2020-12-09 Wave Life Sciences Ltd. Compounds, compositions and methods for synthesis
EP3645546A4 (en) 2017-06-30 2021-12-01 Solstice Biologics, Ltd. Chiral phosphoramidite auxiliaries and methods of their use
WO2019032612A1 (en) 2017-08-08 2019-02-14 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
CN111108096A (en) 2017-09-18 2020-05-05 波涛生命科学有限公司 Oligonucleotide preparation technology
EA201991450A1 (en) 2017-09-22 2019-12-30 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXONISM SKIP IN MUSCULAR DYSTROPHY
EP3687577A1 (en) 2017-09-28 2020-08-05 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
EP3687519A1 (en) 2017-09-28 2020-08-05 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
JP2020536060A (en) 2017-09-28 2020-12-10 サレプタ セラピューティクス, インコーポレイテッド Combination therapy to treat muscular dystrophy
CN107892710B (en) * 2017-10-10 2021-04-13 河北大学 Supported platinum complex oxidant easy to recycle and use and preparation method and application thereof
WO2019075357A1 (en) 2017-10-12 2019-04-18 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
JP7394753B2 (en) 2017-10-18 2023-12-08 サレプタ セラピューティクス, インコーポレイテッド antisense oligomer compounds
US10758629B2 (en) 2018-05-29 2020-09-01 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
EP3806868A4 (en) 2018-06-13 2022-06-22 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
TW202020153A (en) 2018-07-27 2020-06-01 美商薩羅塔治療公司 Exon skipping oligomers for muscular dystrophy
WO2020027225A1 (en) 2018-07-31 2020-02-06 ファイメクス株式会社 Heterocyclic compound
BR112021011018A2 (en) 2018-12-13 2021-08-31 Sarepta Therapeutics, Inc. EXON SKIPPING OLIGOMER CONJUGATES FOR MUSCULAR DYSTROPHY
EP3901161A4 (en) * 2019-01-10 2022-03-23 Nankai University Cyclic dinucleotide prodrug molecule, preparation method therefor and application thereof
JP2022528725A (en) 2019-04-18 2022-06-15 サレプタ セラピューティクス, インコーポレイテッド Composition for treating muscular dystrophy
WO2021020585A1 (en) 2019-07-31 2021-02-04 ファイメクス株式会社 Heterocyclic compound
WO2021216572A1 (en) 2020-04-20 2021-10-28 Massachusetts Institute Of Technology Lipid compositions for delivery of sting agonist compounds and uses thereof
BR112022023465A2 (en) 2020-05-22 2023-01-10 Wave Life Sciences Ltd DOUBLE-STRANDED OLIGONUCLEOTIDE (DSRNAI) AGENT, CHIRALLY CONTROLLED OLIGONUCLEOTIDE COMPOSITION, DOUBLE-STRANDED OLIGONUCLEOTIDE, METHOD FOR REDUCING THE LEVEL AND/OR ACTIVITY OF A TRANSCRIPT OR A PROTEIN ENCODED BY THE SAME, AND METHOD FOR ALLELE-SPECIFIC DELETION OF A TRANSCRIPTION OF A NUCLEIC ACID SEQUENCE
TW202241454A (en) * 2021-02-01 2022-11-01 日商第一三共股份有限公司 Novel method for producing antibody-immunostimulator conjugate
CA3233242A1 (en) 2021-09-30 2023-04-06 Sarepta Therapeutics, Inc. Antisense oligonucleotides having one or more abasic units
WO2023152371A1 (en) 2022-02-14 2023-08-17 Proqr Therapeutics Ii B.V. Guide oligonucleotides for nucleic acid editing in the treatment of hypercholesterolemia
WO2024013361A1 (en) 2022-07-15 2024-01-18 Proqr Therapeutics Ii B.V. Oligonucleotides for adar-mediated rna editing and use thereof
WO2024013360A1 (en) 2022-07-15 2024-01-18 Proqr Therapeutics Ii B.V. Chemically modified oligonucleotides for adar-mediated rna editing
WO2024064237A2 (en) 2022-09-21 2024-03-28 Sarepta Therapeutics, Inc. Dmd antisense oligonucleotide-mediated exon skipping efficiency

Family Cites Families (687)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2878264A (en) 1959-03-17 Substituted amino alcohols
CH372667A (en) 1957-09-26 1963-10-31 Robins Co Inc A H Process for the preparation of 3-aryl-3-pyrrolidinols
US3135766A (en) 1961-10-03 1964-06-02 Mead Johnson & Co 3-substituted-3-pyrrolidinols
US3484473A (en) 1967-05-12 1969-12-16 Buckman Labor Inc Methylene bisesters of thiolsulfonic acids
DE1934150A1 (en) 1968-07-10 1970-01-15 Pennwalt Corp New 1-alkanoyloxy-1,2,4,5-tetrahydro-3H, 3-benzazepine
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US3745162A (en) 1970-08-31 1973-07-10 Robins Co Inc A H 1,2,3,4-tetrahydroisoquinoline-2-(thio)-carboxamides
GB1448437A (en) 1973-02-24 1976-09-08 Beecham Group Ltd Diphenylpropylamines
US4022791A (en) 1975-06-03 1977-05-10 Pfizer Inc. 2-Aminomethyl-3,4-dihydronaphthalenes
GB1504424A (en) 1975-08-09 1978-03-22 Beecham Group Ltd Isoquinoline-derived aminoethers
BR7807288A (en) 1977-11-08 1979-06-12 Genentech Inc POLYNUCLEOTIDE SYNTHESIS PROCESS
DD133885B1 (en) 1978-01-04 1981-02-25 Hans Lehmann AGENTS FOR THE CONTROL OF PHYTOPATHOGENIC BACTERIA AND MUSHROOMS
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4542142A (en) 1982-11-22 1985-09-17 Roussel Uclaf Insecticidal cyclopropane carboxylic acid derivatives with 3-unsaturated-side chain
DE3329892A1 (en) 1983-08-18 1985-03-07 Köster, Hubert, Prof. Dr., 2000 Hamburg METHOD FOR PRODUCING OLIGONUCLEOTIDES
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5643889A (en) * 1984-07-11 1997-07-01 Temple University-Of The Commonwealth System Of Pennsylvania Cholesterol conjugates of 2'5'-oligoadenylate derivatives and antiviral uses thereof
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
FR2576898B1 (en) 1985-02-01 1988-01-08 Lafon Labor 3-PHENYL-TETRAHYDROPYRIDINE DERIVATIVES, METHOD OF PREPARATION AND THERAPEUTIC USE
JPH0658512B2 (en) 1985-04-12 1994-08-03 富士写真フイルム株式会社 Silver halide photographic light-sensitive material
US4659774A (en) 1985-11-01 1987-04-21 American Hoechst Corporation Support for solid-phase oligonucleotide synthesis
US4735949A (en) 1986-02-18 1988-04-05 Warner-Lambert Company Disubstituted-7-pyrrolidinonaphthyridine antibacterial agents
US4840956A (en) 1986-02-18 1989-06-20 Warner-Lambert Company Novel disubstituted-7-pyrrolidinoquinoline antibacterial agents
IL83663A0 (en) 1986-10-27 1988-01-31 Robins Co Inc A H Preparation of 3-pyrrolidinols
DE3851889T2 (en) 1987-06-24 1995-04-13 Florey Howard Inst NUCLEOSIDE DERIVATIVES.
ES2045028T3 (en) 1987-07-30 1994-01-16 Univ Bar Ilan BIOLOGICALLY ACTIVE CARBOXYL ACID ESTERS.
US4923901A (en) 1987-09-04 1990-05-08 Millipore Corporation Membranes with bound oligonucleotides and peptides
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US4943629A (en) 1988-08-12 1990-07-24 American Cyanamid Company Antidiabetic alpha-substituted phosphonates
US4945158A (en) 1988-08-12 1990-07-31 American Cyanamid Company Antidiabetic phosphonates
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
JP2794461B2 (en) 1989-08-17 1998-09-03 有機合成薬品工業株式会社 Phosphoramidite compounds and solid-phase synthesis of oligoribonucleotides using the same
US5141813A (en) 1989-08-28 1992-08-25 Clontech Laboratories, Inc. Multifunctional controlled pore glass reagent for solid phase oligonucleotide synthesis
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
CA2029273A1 (en) 1989-12-04 1991-06-05 Christine L. Brakel Modified nucleotide compounds
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
EP0507863A4 (en) 1989-12-28 1993-07-07 Virginia Commonwealth University Sigma receptor ligands and the use thereof
US5506212A (en) 1990-01-11 1996-04-09 Isis Pharmaceuticals, Inc. Oligonucleotides with substantially chirally pure phosphorothioate linkages
US5212295A (en) 1990-01-11 1993-05-18 Isis Pharmaceuticals Monomers for preparation of oligonucleotides having chiral phosphorus linkages
US5620963A (en) 1991-10-15 1997-04-15 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating protein kinase C having phosphorothioate linkages of high chiral purity
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5852188A (en) 1990-01-11 1998-12-22 Isis Pharmaceuticals, Inc. Oligonucleotides having chiral phosphorus linkages
US7101993B1 (en) 1990-01-11 2006-09-05 Isis Pharmaceuticals, Inc. Oligonucleotides containing 2′-O-modified purines
US6339066B1 (en) 1990-01-11 2002-01-15 Isis Pharmaceuticals, Inc. Antisense oligonucleotides which have phosphorothioate linkages of high chiral purity and which modulate βI, βII, γ, δ, Ε, ζ and η isoforms of human protein kinase C
US5635488A (en) 1991-10-15 1997-06-03 Isis Pharmaceuticals, Inc. Compounds having phosphorodithioate linkages of high chiral purity
US5914396A (en) 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
DE69133405T2 (en) 1990-01-11 2005-07-07 Isis Pharmaceutical, Inc., Carlsbad Oligonucleotide derivatives for detecting and modulating RNA activity and gene expression
US5292875A (en) 1990-04-20 1994-03-08 Lynx Therapeutics, Inc. Method of synthesizing sulfurized oligonucleotide analogs
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
WO1991017755A1 (en) 1990-05-23 1991-11-28 Isis Pharmaceuticals, Inc. Compositions and methods for modulating rna activity through modification of the 5' cap structure of rna
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5792844A (en) 1990-07-27 1998-08-11 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent nitrogen atoms
US6087482A (en) 1990-07-27 2000-07-11 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5834607A (en) 1990-07-27 1998-11-10 Isis Pharmaceuticals, Inc. Amines and methods of making and using the same
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5998603A (en) 1994-09-29 1999-12-07 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analogs, and oligomers thereof
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US6121433A (en) 1990-07-27 2000-09-19 Isis Pharmaceuticals, Inc. Oligomeric compounds having nitrogen-containing linkages
DE69126530T2 (en) 1990-07-27 1998-02-05 Isis Pharmaceuticals Inc NUCLEASE RESISTANT, PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECT AND MODULE GENE EXPRESSION
US5783682A (en) 1990-07-27 1998-07-21 Isis Pharmaceuticals, Inc. Oligonucleotide mimics having nitrogen-containing linkages
WO1994022886A1 (en) 1993-03-30 1994-10-13 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
JPH04348044A (en) 1991-02-21 1992-12-03 Matsushita Electron Corp Resin sealing device for semiconductor
US5512668A (en) 1991-03-06 1996-04-30 Polish Academy Of Sciences Solid phase oligonucleotide synthesis using phospholane intermediates
US20020183502A1 (en) 1991-05-21 2002-12-05 Mesmaeker Alain De Backbone-modified oligonucleotide analogs and methods for using same
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
JPH04348077A (en) 1991-05-24 1992-12-03 Nec Corp Thin-film transistor
US6414112B1 (en) 1991-05-24 2002-07-02 Ole Buchardt Peptide nucleic acids having 2,6-diaminopurine nucleobases
BR9206131A (en) 1991-06-10 1995-05-02 Lucky Ltd Hepatitis C: diagnosis and vaccines
US5359052A (en) 1991-08-05 1994-10-25 Polish Academy Of Sciences Chalcophospholanes useful in the synthesis of oligonucleoside phosphorothioates, phosphorodithioates and related selenates
US5646267A (en) 1991-08-05 1997-07-08 Polish Academy Of Sciences Method of making oligonucleotides and oligonucleotide analogs using phospholanes and enantiomerically resolved phospholane analogues
US6369209B1 (en) 1999-05-03 2002-04-09 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US7119184B2 (en) 1991-08-12 2006-10-10 Isis Pharmaceuticals, Inc. Oligonucleotides having A-DNA form and B-DNA form conformational geometry
US5607923A (en) 1991-10-15 1997-03-04 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating cytomegalovirus having phosphorothioate linkages of high chiral purity
US5654284A (en) 1991-10-15 1997-08-05 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating RAF kinase having phosphorothioate linkages of high chiral purity
ATE221127T1 (en) 1991-10-15 2002-08-15 Isis Pharmaceuticals Inc OLIGONUCLEOTIDES BONDED VIA CHIRAL PHOSPHORATOMS
US5599797A (en) 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5576302A (en) 1991-10-15 1996-11-19 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating hepatitis C virus having phosphorothioate linkages of high chiral purity
US5661134A (en) 1991-10-15 1997-08-26 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating Ha-ras or Ki-ras having phosphorothioate linkages of high chiral purity
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US6235887B1 (en) 1991-11-26 2001-05-22 Isis Pharmaceuticals, Inc. Enhanced triple-helix and double-helix formation directed by oligonucleotides containing modified pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
DK0618925T4 (en) 1991-12-24 2012-07-09 Isis Pharmaceuticals Inc Antisense oligonucleotides
GB9213601D0 (en) 1992-06-26 1992-08-12 Mastico Robert A Protein based delivery system
US7067497B2 (en) 1992-09-29 2006-06-27 Isis Pharmaceuticals, Inc. Modulation of telomere length by oligonucleotides having a G-core sequence
US6444656B1 (en) 1992-12-23 2002-09-03 Biochem Pharma, Inc. Antiviral phosphonate nucleotides
US6005107A (en) 1992-12-23 1999-12-21 Biochem Pharma, Inc. Antiviral compounds
DE69400208T2 (en) 1993-01-25 1996-11-28 Hybridon Inc OLIONUCLEOTIDALKYLPHOSPHONATES AND PHOSPHONOTHIOATES
HU9501978D0 (en) 1993-03-31 1995-09-28 Sterling Winthorp Inc Bifunctional nucleosides, oligomers thereof, and methods of making and using the same
CA2159632A1 (en) 1993-03-31 1994-10-13 Ashis Kumar Saha Novel 5'-substituted nucleosides and oligomers produced therefrom
US5955591A (en) 1993-05-12 1999-09-21 Imbach; Jean-Louis Phosphotriester oligonucleotides, amidites and method of preparation
US6015886A (en) 1993-05-24 2000-01-18 Chemgenes Corporation Oligonucleotide phosphate esters
DE69412704T2 (en) 1993-06-10 1999-02-04 Idemitsu Petrochemical Co Injection mold
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5643989A (en) 1993-10-29 1997-07-01 Azdel, Inc. Fiber reinforced functionalized polyolefin composites
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
DE4435728A1 (en) 1994-01-19 1995-07-20 Boehringer Mannheim Gmbh Biotin silane compounds and binding matrix containing these compounds
US6117679A (en) 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
MX9603542A (en) 1994-02-22 1997-03-29 Novo Nordisk As A method of preparing a variant of a lipolytic enzyme.
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5871966A (en) 1994-05-11 1999-02-16 Novo Nordisk A/S Enzyme with endo-1,3(4)-β- Glucanase activity
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
DK0685475T3 (en) 1994-05-31 1999-08-30 Bayer Ag Amino-benzofuryl and thienyl derivatives
HRP950288A2 (en) 1994-05-31 1997-08-31 Bayer Ag Oxalylamino-benzofuran- and benzothienyl-derivatives
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
PT772619E (en) 1994-07-15 2006-10-31 Univ Iowa Res Found OLIGONUCLEOTIDOS IMUNOMODULADORES
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
DE69521517T2 (en) 1994-09-07 2002-04-18 Hybridon Inc Prodrug-OLIGONUCLEOTIDES
US5681940A (en) 1994-11-02 1997-10-28 Icn Pharmaceuticals Sugar modified nucleosides and oligonucleotides
CN1175281A (en) 1994-12-22 1998-03-04 海布里登公司 Synthesis of stereospecific oligonucleotide phosphorothioates
GB9501465D0 (en) 1995-01-25 1995-03-15 King S College London Nucleoside phosphorothioate derivatives,synthesis and use thereof
US6222025B1 (en) 1995-03-06 2001-04-24 Isis Pharmaceuticals, Inc. Process for the synthesis of 2′-O-substituted pyrimidines and oligomeric compounds therefrom
US6166197A (en) 1995-03-06 2000-12-26 Isis Pharmaceuticals, Inc. Oligomeric compounds having pyrimidine nucleotide (S) with 2'and 5 substitutions
EP0739983B1 (en) 1995-04-27 2009-12-30 Takara Bio Inc. Gene encoding lacto-n-biosidase
WO1996035782A1 (en) 1995-05-11 1996-11-14 Applied Research Systems Il-6 activity inhibitor
CA2221589A1 (en) 1995-05-19 1996-11-21 Glycomed Incorporated Collection of activated glycoside compounds and their biological use
AU5871196A (en) 1995-05-23 1996-12-24 Hybridon, Inc. Methods and compounds for the synthesis of oligonucleotides and the oligonucleotides thereby produced
WO1996037504A1 (en) 1995-05-23 1996-11-28 Hybridon, Inc. Novel synthons for stereoselective oligonucleotide synthesis
JPH10510433A (en) 1995-06-06 1998-10-13 アイシス・ファーマシューティカルス・インコーポレーテッド Oligonucleotides with high chiral purity phosphorothioate linkages
US5932450A (en) 1995-06-07 1999-08-03 Gen-Probe Incorporated Enzymatic synthesis of oligonucleotides using digestible templates
US5795765A (en) 1995-06-29 1998-08-18 Takara Shuzo Co., Ltd. Gene encoding endoglycoceramidase
EP0759470B1 (en) 1995-06-29 2006-10-25 Takara Bio Inc. Gene encoding endoglycoceramidase activator
US6017700A (en) 1995-08-04 2000-01-25 Bayer Corporation Cationic oligonucleotides, and related methods of synthesis and use
US5936080A (en) 1996-05-24 1999-08-10 Genta Incorporated Compositions and methods for the synthesis of organophosphorus derivatives
WO1997009443A1 (en) 1995-09-05 1997-03-13 Michigan State University PROCESS FOR THE ISOLATION AND PURIFICATION OF TAXOL AND TAXANES FROM TAXUS spp
US6160109A (en) 1995-10-20 2000-12-12 Isis Pharmaceuticals, Inc. Preparation of phosphorothioate and boranophosphate oligomers
US5734041A (en) 1995-10-20 1998-03-31 Mcgill University Preparation of chiral phosphorothioate oligomers
US6476216B1 (en) 1995-10-20 2002-11-05 Mcgill University Preparation of phosphorothioate oligomers
US7018793B1 (en) 1995-12-07 2006-03-28 Diversa Corporation Combinatorial screening of mixed populations of organisms
AU708535B2 (en) 1996-02-15 1999-08-05 Cleveland Clinic Foundation, The RNase L activators and antisense oligonucleotides effective to treat RSV infections
US6214805B1 (en) 1996-02-15 2001-04-10 The United States Of America As Represented By The Department Of Health And Human Services RNase L activators and antisense oligonucleotides effective to treat RSV infections
GB9604669D0 (en) 1996-03-05 1996-05-01 Ciba Geigy Ag Chemical compounds
US5824669A (en) 1996-03-22 1998-10-20 Nitromed, Inc. Nitrosated and nitrosylated compounds and compositions and their use for treating respiratory disorders
DE69738254T2 (en) 1996-05-10 2008-08-14 Novozymes A/S METHOD FOR PROVISION OF DNA SEQUENCES
US5856465A (en) 1996-05-24 1999-01-05 Polska Akademia Nauk Centrum Badan Molekularnych I Makromolekularnych Compositions and methods for the synthesis of chirally pure organophosphorus nucleoside derivatives
WO2005121371A2 (en) 2004-06-03 2005-12-22 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
DE19622783A1 (en) 1996-06-07 1997-12-11 Hoechst Ag Isolation of the biosynthetic genes for pseudo-oligosaccharides from Streptomyces glaucescens GLA.O and their use
DK0912767T3 (en) 1996-07-16 2006-10-30 Gen Probe Inc Method for Tracking and Amplifying Nucleic Acid Sequences Using Modified Oligonucleotides with Enhanced Target Specific TM
CA2261566A1 (en) 1996-07-24 1998-01-29 Buchardt, Dorte Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
WO1998007734A1 (en) * 1996-08-21 1998-02-26 Hybridon, Inc. Oligonucleotide prodrugs
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
GB9621522D0 (en) 1996-10-16 1996-12-04 Biocompatibles Ltd Synthesis of phosphorus compounds
US6639062B2 (en) 1997-02-14 2003-10-28 Isis Pharmaceuticals, Inc. Aminooxy-modified nucleosidic compounds and oligomeric compounds prepared therefrom
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6369237B1 (en) 1997-03-07 2002-04-09 President And Fellows Of Harvard College DNA glycosylase inhibitors, and uses related thereto
US6015887A (en) 1997-04-11 2000-01-18 Isis Pharmaceuticals, Inc. Chiral peptide nucleic acids and methods for preparing same
US6468983B2 (en) 1997-04-21 2002-10-22 The Cleveland Clinic Foundation RNase L activators and antisense oligonucleotides effective to treat telomerase-expressing malignancies
PL184612B1 (en) 1997-04-25 2002-11-29 Pan Method of obtaining p-chiral analoques of nucleotides
AU745309B2 (en) 1997-05-28 2002-03-21 Peter E. Nielsen Conjugated peptide nucleic acids having enhanced cellular uptake
AR013142A1 (en) 1997-06-27 2000-12-13 Procter & Gamble PRO-FRAGRANCE COMPOUND WITH CYCLIC ACETALS DETERGENT COMPOSITION FOR CLOTHES WASHING, SOFTENING COMPOSITION OF FABRICS AND MANUFACTURING ARTICLE TO PROVIDE APPEARANCE BENEFITS TO FABRICS, WHICH INCLUDE IT
AU8512598A (en) 1997-07-25 1999-02-16 Hybridon, Inc. Oligonuclotides having 3' terminal stereospecific phosphorothioates
GB9717158D0 (en) 1997-08-13 1997-10-22 King S College London Solution synthesis of oligonucleotides and their phosphorothioate analogues
US6767739B2 (en) 2001-07-30 2004-07-27 Isis Pharmaceuticals Inc. Antisense modulation of microsomal triglyceride transfer protein expression
US6383808B1 (en) 2000-09-11 2002-05-07 Isis Pharmaceuticals, Inc. Antisense inhibition of clusterin expression
US6750344B1 (en) 1997-09-05 2004-06-15 Isis Pharmaceuticals, Inc. Amine compounds and combinatorial libraries comprising same
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
DE19741715A1 (en) 1997-09-22 1999-03-25 Hoechst Ag New pentopyranosyl nucleoside compounds
US6232463B1 (en) 1997-10-09 2001-05-15 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US6528640B1 (en) 1997-11-05 2003-03-04 Ribozyme Pharmaceuticals, Incorporated Synthetic ribonucleic acids with RNAse activity
US6617438B1 (en) 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US6080543A (en) 1997-12-08 2000-06-27 E. & J. Gallo Winery Detection of fungal pathogens
US6582936B1 (en) 1997-12-12 2003-06-24 The Regents Of The University Of California Methods for making nucleic acids
US6248519B1 (en) 1998-03-11 2001-06-19 E & J Gallo Winery Detection of fermentation-related microorganisms
US7045610B2 (en) 1998-04-03 2006-05-16 Epoch Biosciences, Inc. Modified oligonucleotides for mismatch discrimination
JP2002513763A (en) 1998-05-06 2002-05-14 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Methods for preventing and treating parasitic infections and related diseases using CPG oligonucleotides
WO1999058118A2 (en) 1998-05-14 1999-11-18 Cpg Immunopharmaceuticals Gmbh METHODS FOR REGULATING HEMATOPOIESIS USING CpG-OLIGONUCLEOTIDES
US6867294B1 (en) 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
EP1100807A1 (en) 1998-07-27 2001-05-23 University Of Iowa Research Foundation STEREOISOMERS OF CpG OLIGONUCLEOTIDES AND RELATED METHODS
DK1104306T3 (en) 1998-08-10 2006-05-22 Antigenics Inc Preparations of CpG and Saponin Adjuvants and Methods for Using Them
WO2000023444A1 (en) 1998-10-21 2000-04-27 Abbott Laboratories 5,7-disubstituted-4-aminopyrido[2,3-d]pyrimidine compounds
US6995259B1 (en) 1998-10-23 2006-02-07 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
AU1742600A (en) 1998-11-25 2000-06-13 Isis Pharmaceuticals, Inc. Identification of disease predictive nucleic acids
US6451524B1 (en) 1998-11-25 2002-09-17 Isis Pharmaceuticals, Inc. Identification of disease predictive nucleic acids
DK1141335T3 (en) 1998-12-21 2009-11-09 Genencor Int Chemically modified enzymes with multiple charged variants
CA2702148C (en) 1999-01-06 2014-03-04 Genenews Inc. Method of profiling gene expression in a human subject having an infectious disease
US6265172B1 (en) 1999-02-08 2001-07-24 University Of Kentucky Diagnostic test and therapy for manganese superoxide dismutate (mNsod) associated diseases
US6121437A (en) 1999-03-16 2000-09-19 Isis Pharmaceuticals, Inc. Phosphate and thiophosphate protecting groups
US6506594B1 (en) 1999-03-19 2003-01-14 Cornell Res Foundation Inc Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
GB9907245D0 (en) 1999-03-29 1999-05-26 Goldsborough Andrew Cleavage of nucleic acids from solid supports
JP3072345B1 (en) 1999-03-31 2000-07-31 農林水産省家畜衛生試験場長 Swine erysipelas recombinant subunit vaccine
US5998148A (en) 1999-04-08 1999-12-07 Isis Pharmaceuticals Inc. Antisense modulation of microtubule-associated protein 4 expression
US6977245B2 (en) 1999-04-12 2005-12-20 The United States Of America As Represented By The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US6300069B1 (en) 1999-05-03 2001-10-09 Qiagen Gmbh Generation and amplification of nucleic acids from ribonucleic acids
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6066500A (en) 1999-06-25 2000-05-23 Isis Pharmaceuticals Inc. Antisense modulation of Beta catenin expression
US6271004B1 (en) 1999-06-25 2001-08-07 Display Systems Biotech A/S Method for improved reverse transcription at high temperatures
US6414135B1 (en) 1999-07-07 2002-07-02 Isis Pharmaceuticals, Inc. C3′-methylene hydrogen phosphonate monomers and related compounds
US20030092647A1 (en) 2001-08-08 2003-05-15 Crooke Rosanne M. Antisense modulation of cholesteryl ester transfer protein expression
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US7264932B2 (en) 1999-09-24 2007-09-04 Applera Corporation Nuclease inhibitor cocktail
SK287400B6 (en) 1999-09-25 2010-08-09 University Of Iowa Research Foundation Immunostimulatory nucleic acid composition and the use thereof for stimulating an immune response
US6949520B1 (en) 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
ES2265980T5 (en) 1999-09-27 2010-12-28 Coley Pharmaceutical Group, Inc. METHODS RELATED TO INTERFERON INDUITED BY IMMUNE STIMULATING NUCLEIC ACIDS.
US20020082227A1 (en) 1999-09-30 2002-06-27 Scott Henry Use of oligonucleotides for inhibition of complement activation
AU7863200A (en) 1999-10-06 2001-05-10 Quark Biotech, Inc. Method for enrichment of natural antisense messenger rna
GB9924285D0 (en) 1999-10-14 1999-12-15 Avecia Ltd Process
US20010055761A1 (en) 1999-10-29 2001-12-27 Agilent Technologies Small scale dna synthesis using polymeric solid support with functionalized regions
FR2800750B1 (en) 1999-11-05 2003-01-31 Centre Nat Rech Scient MEMBRANE PROTEINS CTL (CHOLINE TRANSPORTER LIKE) INVOLVED IN THE TRANSPORT OF CHOLINE
WO2001040515A1 (en) 1999-11-12 2001-06-07 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6322985B1 (en) 1999-12-27 2001-11-27 Technion Research And Development Foundation Ltd. Abundant, well distributed and hyperpolymorphic simple sequence repeats in prokaryote genomes and use of same for prokaryote classification and typing
US7501091B2 (en) 1999-12-30 2009-03-10 Smiths Detection Inc. Sensors with improved properties
US7055094B2 (en) 1999-12-30 2006-05-30 Rutgers, The State University Of New Jersey Virtual tags and the process of virtual tagging utilizing user feedback in transformation rules
US6649750B1 (en) 2000-01-05 2003-11-18 Isis Pharmaceuticals, Inc. Process for the preparation of oligonucleotide compounds
US6159697A (en) 2000-01-19 2000-12-12 Isis Pharmaceuticals, Inc. Antisense modulation of Smad7 expression
US7585847B2 (en) 2000-02-03 2009-09-08 Coley Pharmaceutical Group, Inc. Immunostimulatory nucleic acids for the treatment of asthma and allergy
US6495677B1 (en) 2000-02-15 2002-12-17 Kanda S. Ramasamy Nucleoside compounds
GB0004889D0 (en) 2000-03-01 2000-04-19 Avecia Ltd Synthesis of oligonucleotides
EP1130091A3 (en) 2000-03-01 2001-11-14 Message Pharmaceuticals, Inc. Bacterial RNaseP Proteins and their use in identifying antibacterial compounds
WO2001070663A2 (en) 2000-03-17 2001-09-27 Corixa Corporation Novel amphipathic aldehydes and their use as adjuvants and immunoeffectors
DE10019756A1 (en) 2000-04-20 2001-10-25 Bayer Ag Process for the production of superabsorbent polymers from polyacrylonitriles
EP1278728B1 (en) 2000-04-20 2004-08-25 F. Hoffmann-La Roche Ag Pyrrolidine and piperidine derivatives and their use for the treatment of neurodegenerative disorders
WO2001085751A1 (en) 2000-05-09 2001-11-15 Reliable Biopharmaceutical, Inc. Polymeric compounds useful as prodrugs
US6492171B2 (en) 2000-05-16 2002-12-10 Isis Pharmaceuticals, Inc. Antisense modulation of TERT expression
US6815542B2 (en) 2000-06-16 2004-11-09 Ribapharm, Inc. Nucleoside compounds and uses thereof
JP4331476B2 (en) 2000-08-03 2009-09-16 エフ.ホフマン−ラ ロシュ アーゲー Nucleic acid binding compounds containing pyrazolo [3,4-d] pyrimidine analogs of purine-2,6-diamine and uses thereof
US6725412B1 (en) 2000-08-15 2004-04-20 Dolby Laboratories Licensing Corporation Low latency data encoder
US6809195B1 (en) 2000-08-16 2004-10-26 Isis Pharmaceuticals, Inc. Process for the preparation of oligonucleotides
US6559279B1 (en) 2000-09-08 2003-05-06 Isis Pharmaceuticals, Inc. Process for preparing peptide derivatized oligomeric compounds
WO2002022809A2 (en) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
EP1191097A1 (en) 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
GB0024752D0 (en) 2000-10-10 2000-11-22 Univ Belfast Oxidative halogenation of aromatic compounds
BR0114786A (en) 2000-10-18 2003-08-12 Glaxosmithkline Biolog Sa Immunogenic composition, method of treatment of a patient suffering from, or susceptible to, a cancer, and, use of a combination of a saponin, an immunostimulating oligotide and a cancer antigen, and, method for the manufacture of a composition.
US6372492B1 (en) 2000-10-30 2002-04-16 Isis Pharmaceuticals, Inc. Antisense modulation of talin expression
US6682889B1 (en) 2000-11-08 2004-01-27 Becton, Dickinson And Company Amplification and detection of organisms of the Chlamydiaceae family
NL1016978C2 (en) 2000-12-22 2002-06-25 Robert Jan Colenbrander Device and method for packaging and preparing food and method for manufacturing such a device.
AU2002243791B2 (en) 2001-01-22 2006-06-29 Isis Pharmaceuticals, Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
US8008459B2 (en) 2001-01-25 2011-08-30 Evolva Sa Concatemers of differentially expressed multiple genes
ATE501251T1 (en) 2001-01-25 2011-03-15 Evolva Ltd CELL LIBRARY
AU2002224668B2 (en) 2001-01-26 2007-09-20 Commonwealth Scientific And Industrial Research Organisation Methods and means for producing efficient silencing construct using recombinational cloning
US20050277133A1 (en) 2001-05-18 2005-12-15 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20030207804A1 (en) 2001-05-25 2003-11-06 Muthiah Manoharan Modified peptide nucleic acids
GB0113523D0 (en) 2001-06-04 2001-07-25 Torotrak Dev Ltd An Hydraulic control circuit for a continuosly variable transmission
US20030069410A1 (en) 2001-06-14 2003-04-10 Isis Pharmaceuticals, Inc. Methods for preparing oligonucleotides having chiral phosphorothioate linkages
US20050019915A1 (en) 2001-06-21 2005-01-27 Bennett C. Frank Antisense modulation of superoxide dismutase 1, soluble expression
CZ20033515A3 (en) 2001-06-29 2005-01-12 Chiron Corporation HCV E1E2 vaccine composition
JP2005504020A (en) 2001-07-03 2005-02-10 アイシス・ファーマシューティカルス・インコーポレーテッド Nuclease resistant chimeric oligonucleotide
US7205399B1 (en) 2001-07-06 2007-04-17 Sirna Therapeutics, Inc. Methods and reagents for oligonucleotide synthesis
US6440739B1 (en) 2001-07-17 2002-08-27 Isis Pharmaceuticals, Inc. Antisense modulation of glioma-associated oncogene-2 expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US6455308B1 (en) 2001-08-01 2002-09-24 Isis Pharmaceuticals, Inc. Antisense modulation of serum amyloid A4 expression
US7888324B2 (en) 2001-08-01 2011-02-15 Genzyme Corporation Antisense modulation of apolipoprotein B expression
US7259150B2 (en) 2001-08-07 2007-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein (a) expression
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US7354909B2 (en) 2001-08-14 2008-04-08 The United States Of America As Represented By Secretary Of The Department Of Health And Human Services Method for rapid generation of mature dendritic cells
US20030232978A1 (en) 2001-08-24 2003-12-18 Seeberger Peter H. Reagents that facilitate the purification of compounds synthesized on a solid support
US7049122B2 (en) 2001-09-21 2006-05-23 Academia Sinica Mutant-type lipases and applications thereof
US6933288B2 (en) 2002-02-04 2005-08-23 Isis Pharmaceuticals, Inc. Pyranosyl cytosines: pharmaceutical formulations and methods
JP4348044B2 (en) 2002-02-12 2009-10-21 株式会社キラルジェン Method for producing highly stereoregular dinucleoside phosphorothioates
US20040149587A1 (en) 2002-02-15 2004-08-05 George Hradil Electroplating solution containing organic acid complexing agent
US20030159938A1 (en) 2002-02-15 2003-08-28 George Hradil Electroplating solution containing organic acid complexing agent
US20050096284A1 (en) 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US8232383B2 (en) 2002-02-20 2012-07-31 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
WO2003073989A2 (en) 2002-02-28 2003-09-12 Biota, Inc. Nucleoside 5'-monophosphate mimics and their prodrugs
EP1485395A4 (en) 2002-02-28 2011-04-13 Biota Scient Management Nucleotide mimics and their prodrugs
US7288376B2 (en) 2002-03-22 2007-10-30 Council Of Scientific And Industrial Research Method of detection of SP-A2 gene variants useful for prediction of predisposition to aspergillosis
US20040102394A1 (en) 2002-11-23 2004-05-27 Isis Pharmaceuticals Inc. Modulation of huntingtin interacting protein 2 expression
US7247621B2 (en) 2002-04-30 2007-07-24 Valeant Research & Development Antiviral phosphonate compounds and methods therefor
WO2003097662A1 (en) 2002-05-15 2003-11-27 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein b expression
AU2003241621A1 (en) 2002-05-24 2003-12-12 Isis Pharmaceuticals, Inc. Oligonucleotides having modified nucleoside units
AU2003237249A1 (en) 2002-05-24 2003-12-12 Isis Pharmaceuticals, Inc. Oligonucleotides having modified nucleoside units
US7507808B2 (en) 2002-12-12 2009-03-24 Isis Pharmaceuticals, Inc. Modulation of endothelial lipase expression
AU2003248708A1 (en) 2002-06-17 2003-12-31 Isis Pharmaceuticals, Inc. Oligomeric compounds that include carbocyclic nucleosides and their use in gene modulation
CN1662253A (en) 2002-06-20 2005-08-31 赛托斯生物技术公司 Packaged virus-like particles for use as adjuvants: method of preparation and use
WO2004003228A1 (en) 2002-07-01 2004-01-08 Unisearch Limited Genotyping method
EP1520022B1 (en) 2002-07-10 2015-07-22 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Rna-interference by single-stranded rna molecules
US20040023905A1 (en) 2002-07-31 2004-02-05 Isis Pharmaceuticals Inc. Antisense modulation of LAR expression
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20080274989A1 (en) 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
US20050255086A1 (en) 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
US8729036B2 (en) 2002-08-07 2014-05-20 University Of Massachusetts Compositions for RNA interference and methods of use thereof
AU2003259735A1 (en) 2002-08-08 2004-02-25 Sirna Therapeutics, Inc. Small-mer compositions and methods of use
AR040996A1 (en) 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
US7414116B2 (en) 2002-08-23 2008-08-19 Illumina Cambridge Limited Labelled nucleotides
CN1694959B (en) 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
US7030230B2 (en) 2002-10-25 2006-04-18 Isis Pharmaceuticals, Inc. Process of purifying phosphoramidites
BR0315810A (en) 2002-10-29 2005-09-13 Coley Pharmaceutical Group Ltd Use of cpg oligonucleotides in the treatment of hepatitis c virus infection
WO2004042018A2 (en) 2002-11-01 2004-05-21 The Regents Of The University Of Colorado Dopamine neurons from human embryonic stem cells
CA2505090A1 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Conjugated oligomeric compounds and their use in gene modulation
US7696345B2 (en) 2002-11-05 2010-04-13 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
AU2003290598A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
WO2004044134A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Phosphorous-linked oligomeric compounds and their use in gene modulation
US7381527B2 (en) 2002-11-06 2008-06-03 Council Of Scientific And Industrial Research Method of detection of SP-A2 gene variants
US7511131B2 (en) 2002-11-13 2009-03-31 Genzyme Corporation Antisense modulation of apolipoprotein B expression
CA2505801A1 (en) 2002-11-13 2004-05-27 Rosanne Crooke Antisense modulation of apolipoprotein b expression
EP2284266B1 (en) 2002-11-14 2013-11-06 Thermo Fisher Scientific Biosciences Inc. siRNA targeting tp53
AU2003225705A1 (en) 2003-03-07 2004-09-30 Ribapharm Inc. Cytidine analogs and methods of use
ATE479752T1 (en) 2003-03-07 2010-09-15 Alnylam Pharmaceuticals Inc THERAPEUTIC COMPOSITIONS
AU2003225410A1 (en) 2003-03-21 2004-10-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
GB0306657D0 (en) 2003-03-24 2003-04-30 Avecia Ltd Process and compounds
US7537767B2 (en) 2003-03-26 2009-05-26 Cytis Biotechnology Ag Melan-A- carrier conjugates
AU2004224762B2 (en) 2003-03-26 2009-12-24 Kuros Us Llc Packaging of immunostimulatory oligonucleotides into virus-like particles: method of preparation and use
ITRM20030149A1 (en) 2003-04-02 2004-10-03 Giuliani Spa ANTISENSE OLIGONUCLEOTIDES (ODN) FOR SMAD7 AND THEIR USE IN THE MEDICAL FIELD
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
DK1628685T3 (en) 2003-04-25 2011-03-21 Gilead Sciences Inc Antiviral phosphonate analogues
CN101410120A (en) 2003-04-25 2009-04-15 吉里德科学公司 Anti-inflammatory phosphonate compounds
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
US7470724B2 (en) 2003-04-25 2008-12-30 Gilead Sciences, Inc. Phosphonate compounds having immuno-modulatory activity
US20090247488A1 (en) 2003-04-25 2009-10-01 Carina Cannizzaro Anti-inflammatory phosphonate compounds
WO2004096233A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Nucleoside phosphonate conjugates
EP1617848A2 (en) 2003-04-25 2006-01-25 Gilead Sciences, Inc. Anti-cancer phosphonate conjugates
US7407965B2 (en) 2003-04-25 2008-08-05 Gilead Sciences, Inc. Phosphonate analogs for treating metabolic diseases
WO2005002626A2 (en) 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds
US20050261237A1 (en) 2003-04-25 2005-11-24 Boojamra Constantine G Nucleoside phosphonate analogs
US7432261B2 (en) 2003-04-25 2008-10-07 Gilead Sciences, Inc. Anti-inflammatory phosphonate compounds
US7045306B2 (en) 2003-04-28 2006-05-16 The General Hospital Corporation Method for identifying compounds in vitro that modulate the dysregulation of transcription of transcription mediated by mutant huntingtin protein
US7214491B2 (en) 2003-05-07 2007-05-08 E. I. Du Pont De Nemours And Company Δ-12 desaturase gene suitable for altering levels of polyunsaturated fatty acids in oleaginous yeasts
WO2004101787A1 (en) 2003-05-14 2004-11-25 Japan Science And Technology Agency Inhibition of the expression of huntington gene
WO2005002507A2 (en) * 2003-06-03 2005-01-13 Isis Pharmaceuticals, Inc. Modulation of survivin expression
JP2007524615A (en) 2003-06-20 2007-08-30 コーリー ファーマシューティカル ゲーエムベーハー Low molecular weight Toll-like receptor (TLR) antagonist
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
JP2005089441A (en) 2003-08-08 2005-04-07 Toudai Tlo Ltd Manufacturing method of highly stereoregular phosphorus atom-modified nucleotide analogue
JP2011088935A (en) 2003-08-08 2011-05-06 Chiralgen Ltd Optically-active nucleoside 3'-phosphoroamidite for production of phosphorus atom modified nucleotide analog
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
BRPI0413684A (en) 2003-08-21 2006-10-24 Univ Griffith compounds, pharmaceutical composition and methods of preparation, treatment of microbial infection and extermination of microorganism and uses
EP1664071A1 (en) 2003-08-21 2006-06-07 Griffith University Novel sulfenamide oxides
RU2006109491A (en) 2003-08-27 2006-08-10 Байота, Инк. (Au) NEW TRICYCLIC NUCLEOSIDES OR NUCLEOTIDES AS A THERAPEUTIC MEDICINES
ES2382807T3 (en) 2003-08-28 2012-06-13 Takeshi Imanishi New artificial nucleic acids of the N-O link type with cross-linking
JP4616175B2 (en) 2003-09-02 2011-01-19 株式会社キラルジェン Method for producing 5'-phosphitylated monomer and H-phosphonate oligonucleotide derivative
JP4580870B2 (en) 2003-09-02 2010-11-17 株式会社キラルジェン Method for producing ribonucleotide or ribonucleotide derivative
PL1667522T3 (en) 2003-09-09 2018-06-29 Geron Corporation Modified oligonucleotides for telomerase inhibition
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US20050074801A1 (en) 2003-09-09 2005-04-07 Monia Brett P. Chimeric oligomeric compounds comprising alternating regions of northern and southern conformational geometry
EP2821085B1 (en) 2003-09-12 2020-04-29 University of Massachusetts Rna interference for the treatment of gain-of-function disorders
US8680063B2 (en) 2003-09-12 2014-03-25 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
GB0323968D0 (en) 2003-10-13 2003-11-19 Glaxosmithkline Biolog Sa Immunogenic compositions
UA88457C2 (en) 2003-10-30 2009-10-26 Коли Фармасьютикал Гмбх Immunostimulatory nucleic acid with enhanced immunostimulatory potency
WO2005042716A2 (en) 2003-10-31 2005-05-12 President And Fellows Of Harvard College Nucleic acid binding oligonucleotides
US7846436B2 (en) * 2003-11-28 2010-12-07 Chemgenes Corporation Oligonucleotides and related compounds
WO2005063983A1 (en) 2003-12-29 2005-07-14 Galapagos Genomics N.V. Modulators of bone homeostasis identified in a high-throughput screen
WO2005070859A1 (en) 2004-01-27 2005-08-04 Takeshi Wada Fluorous supports and processes for production of oligonucleotide derivatives with the same
US20050176045A1 (en) 2004-02-06 2005-08-11 Dharmacon, Inc. SNP discriminatory siRNA
US20080221303A1 (en) 2004-02-18 2008-09-11 Jehoshua Katzhendler Method for the Preparation of Peptide-Oligonucleotide Conjugates
JP3976742B2 (en) 2004-02-27 2007-09-19 江守商事株式会社 Immunostimulatory oligonucleotides that induce interferon alpha
WO2005085272A1 (en) 2004-03-05 2005-09-15 Takeshi Wada Boranophosphate monomer and process for producing oligonucleotide derivative therefrom
WO2005092909A1 (en) 2004-03-25 2005-10-06 Toudai Tlo, Ltd. Processes for producing ribonucleotide analogue with high stereoregularity and deoxyribonucleotide analogue
EP2540734B1 (en) 2004-04-05 2016-03-30 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
TWI350168B (en) 2004-05-07 2011-10-11 Incyte Corp Amido compounds and their use as pharmaceuticals
AU2005248410B2 (en) 2004-05-27 2010-04-22 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Differential expression of molecules associated with acute stroke
US7759318B1 (en) 2004-05-28 2010-07-20 Isis Pharmaceuticals, Inc. Identification of novel pathways, genes and promoter motifs regulating adipogenesis
SI1766010T1 (en) 2004-06-28 2011-06-30 Univ Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
PL2409713T3 (en) 2004-08-10 2016-02-29 Kastle Therapeutics Llc Oligonucleotides for use in modulating lipoprotein and cholesterol levels in humans
CN101048423B (en) 2004-08-26 2011-06-08 日本新药株式会社 Phosphoramidite compound and method for producing oligo-rna
AU2005282380A1 (en) 2004-09-07 2006-03-16 Archemix Corp. Aptamer medicinal chemistry
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
WO2006031461A2 (en) 2004-09-09 2006-03-23 Isis Pharmaceuticals, Inc. Pyrrolidinyl groups for attaching conjugates to oligomeric compounds
EP1799859B1 (en) 2004-09-17 2014-07-02 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
US20090036355A1 (en) 2004-10-13 2009-02-05 Sanjay Bhanot Antisense Modulation of PTP1B Expression
US7608594B2 (en) 2004-11-03 2009-10-27 University Of Kansas Novobiocin analogues as anticancer agents
US8212012B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues having modified sugar moieties
US9120774B2 (en) 2004-11-03 2015-09-01 University Of Kansas Novobiocin analogues having modified sugar moieties
WO2010096650A1 (en) 2009-02-20 2010-08-26 University Of Kansas Novobiocin analogues having modified sugar moieties
US7622451B2 (en) 2004-11-03 2009-11-24 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
US8212011B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues
KR100721928B1 (en) 2004-11-05 2007-05-28 주식회사 바이오씨에스 Pharmaceutical composition for treating or preventing dermatitis comprising CpG oligodeoxynucleotide
EP1657307A1 (en) 2004-11-16 2006-05-17 Immunotech S.A. Oligonucleotides that induce the secretion of GM-CSF
US8003619B2 (en) 2004-12-09 2011-08-23 Alnylam Pharmaceuticals, Inc. Method of stimulating an immune response and inhibiting expression of a gene using an oligonucleotide
US9809824B2 (en) 2004-12-13 2017-11-07 The United States Of America, Represented By The Secretary, Department Of Health And Human Services CpG oligonucleotide prodrugs, compositions thereof and associated therapeutic methods
WO2006066260A2 (en) 2004-12-17 2006-06-22 Thiosense, Inc. Compositions of and methods for producing phosphorus-chiral monomers and oligomers
US20070099851A1 (en) 2004-12-30 2007-05-03 Linn Gregory S Stable analogues of ribose-1-phosphate and methods for treating diabetes and other metabolic disorders
US20060183763A1 (en) 2004-12-31 2006-08-17 Pfizer Inc Novel pyrrolidyl derivatives of heteroaromatic compounds
WO2006080596A1 (en) 2005-01-28 2006-08-03 Hyung-Joo Kwon Oligonucleotides derived from mycobacterium for stimulating immune function, treating immune-related diseases, atopic dermatitis and/or protecting normal immune cell
CA2598992A1 (en) 2005-02-24 2006-08-31 Coley Pharmaceutical Group, Inc. Immunostimulatory oligonucleotides
ATE534652T1 (en) 2005-04-01 2011-12-15 Univ California PHOSPHONO-PENT-2-EN-1-YL NUCLEOSIDES AND ANALOGS
US8097597B2 (en) 2005-05-05 2012-01-17 Antisense Pharma Gmbh Use of low doses of oligonucleotides antisense to TGF-β genes in the treatment of brain tumors
US7902352B2 (en) 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
US20060257912A1 (en) 2005-05-06 2006-11-16 Medtronic, Inc. Methods and sequences to suppress primate huntington gene expression
DK2548560T3 (en) 2005-06-23 2015-07-13 Cold Spring Harbor Lab Compositions and Methods for Modulating SMN2 Splicing
EP2322657B1 (en) 2005-06-28 2014-11-05 Medtronic, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin gene.
US9133517B2 (en) 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
US20090162316A1 (en) 2005-07-05 2009-06-25 Harvard University Liver targeted conjugates
JP4984634B2 (en) 2005-07-21 2012-07-25 ソニー株式会社 Physical information acquisition method and physical information acquisition device
PL1924252T3 (en) 2005-07-28 2012-02-29 Id Fish Tech Inc Method for improving cell permeability to foreign particles
WO2007027775A2 (en) 2005-08-29 2007-03-08 Isis Pharmaceuticals, Inc. Methods for use in modulating mir-122a
US8501703B2 (en) 2005-08-30 2013-08-06 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds for modulation of splicing
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
US20070077993A1 (en) 2005-09-30 2007-04-05 Midgley Timothy M Method and apparatus for collecting user game play data and crediting users in a gaming environment
EP1934239B8 (en) 2005-10-12 2015-11-04 Idera Pharmaceuticals, Inc. Immune regulatory oligonucleotide (iro) compounds to modulate toll-like receptor based immune response
US9308252B2 (en) 2005-10-27 2016-04-12 Cook Biotech, Inc. Extracellular matrix materials as vaccine adjuvants for diseases associated with infectious pathogens or toxins
EP2325315B1 (en) 2005-10-28 2014-05-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of huntingtin gene
CN101466834B (en) 2005-10-28 2013-09-25 东曹株式会社 Method for production of carotenoid-synthesizing microorganism and method for production of carotenoid
AU2006315632A1 (en) * 2005-11-11 2007-05-24 Pfizer, Inc. Combinations and methods of using an immunomodulatory oligodeoxynucleotide
WO2007064291A1 (en) 2005-11-30 2007-06-07 Jyoti Chattopadhyaya Method and compounds for rna synthesis
EP1957507B1 (en) 2005-12-02 2018-10-24 Ionis Pharmaceuticals, Inc. Antibacterial 4,5-substituted aminoglycoside analogs having multiple substituents
US8076303B2 (en) * 2005-12-13 2011-12-13 Spring Bank Pharmaceuticals, Inc. Nucleotide and oligonucleotide prodrugs
US7884117B2 (en) 2005-12-21 2011-02-08 Pfizer Inc. Carbonylamino pyrrolopyrazoles, potent kinase inhibitors
US7951934B2 (en) 2006-01-26 2011-05-31 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
JP5342881B2 (en) 2006-01-27 2013-11-13 アイシス ファーマシューティカルズ, インコーポレーテッド 6-modified bicyclic nucleic acid analogues
ES2526879T3 (en) 2006-02-15 2015-01-16 Adiutide Pharmaceuticals Gmbh Compositions and procedures for oligonucleotide formulations
US7759470B2 (en) 2006-02-20 2010-07-20 Roche Diagnostics Operations, Inc. Labeling reagent
US8383660B2 (en) 2006-03-10 2013-02-26 Pfizer Inc. Dibenzyl amine compounds and derivatives
AU2007234451A1 (en) 2006-03-31 2007-10-11 Atom Acquisition, Llc Reagents useful for synthesizing rhodamine-labeled oligonucleotides
WO2007118222A2 (en) 2006-04-06 2007-10-18 Ibis Biosciences, INC Compositions for the use in identification of fungi
RU2439065C2 (en) 2006-04-20 2012-01-10 Ф.Хоффманн-Ля Рош Аг Diazepane derivatives as modulators of chemokine receptors
CN101432440B (en) 2006-04-24 2013-08-21 西格马食品可变资本有限公司 Method for detection and multiple, simultaneous quantification of pathogens by means of real-time polymerase chain reaction
US20130129752A1 (en) 2006-04-25 2013-05-23 Immune Disease Institute, Inc. Targeted delivery to leukocytes using protein carriers
GB0608838D0 (en) 2006-05-04 2006-06-14 Novartis Ag Organic compounds
WO2007134014A2 (en) 2006-05-05 2007-11-22 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of gcgr
EP2023936A4 (en) 2006-05-05 2010-11-24 Isis Pharmaceuticals Inc Compositions and their uses directed to ptpr alpha
US20090012120A1 (en) 2006-05-10 2009-01-08 Board Of Trustees Of Michigan State University Synthesis of N-heterocycles, beta-amino acids, and allyl amines via aza-payne mediated reaction of ylides and hydroxy aziridines
ES2389737T3 (en) 2006-05-11 2012-10-31 Isis Pharmaceuticals, Inc. 5 'modified bicyclic nucleic acid analogs
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
KR101065760B1 (en) 2006-05-31 2011-09-19 오사카 유니버시티 Immunostimulatory oligonucleotide and pharmaceutical application thereof
US8097596B2 (en) 2006-06-30 2012-01-17 Lakewood-Amedex, Inc. Compositions and methods for the treatment of muscle wasting
EP2046993A4 (en) 2006-07-07 2010-11-17 Univ Massachusetts Rna silencing compositions and methods for the treatment of huntington's disease
AU2007272906B2 (en) 2006-07-12 2013-01-31 The Regents Of The University Of California Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups
AU2007275301A1 (en) 2006-07-20 2008-01-24 Amgen Inc. Substituted azole aromatic heterocycles as inhibitors of 11-beta-HSD-1
GB0614947D0 (en) 2006-07-27 2006-09-06 Isis Innovation Epitope reduction therapy
AU2007281082A1 (en) 2006-08-04 2008-02-07 Isis Pharmaceuticals, Inc. Compositions and methods for the modulation of JNK proteins
AT504194B1 (en) 2006-09-07 2008-07-15 Oesterr Rotes Kreuz BACTERIA DETECTION
US8138330B2 (en) 2006-09-11 2012-03-20 Sigma-Aldrich Co. Llc Process for the synthesis of oligonucleotides
KR101251707B1 (en) 2006-09-27 2013-04-11 콜리 파마슈티칼 게엠베하 CpG oligonucleotide analogs containing hydrophobic T analogs with enhanced immunostimulatory activity
ES2526295T5 (en) 2006-10-18 2021-05-04 Ionis Pharmaceuticals Inc Antisense compounds
AU2007309195A1 (en) 2006-10-23 2008-05-02 Irm Llc Cathepsin proteases inhibitors
EP2104738A2 (en) * 2006-10-26 2009-09-30 Coley Pharmaceuticals GmbH Oligoribonucleotides and uses thereof
FR2908414B1 (en) 2006-11-13 2012-01-20 Centre Nat Rech Scient IMMOBILIZATION OF MEMBRANE PROTEINS ON A SUPPORT THROUGH AN AMPHIPHILE MOLECULE
MX2009005252A (en) 2006-11-17 2009-05-28 Abbott Lab Aminopyrrolidines as chemokine receptor antagonists.
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
JP2010510807A (en) 2006-11-27 2010-04-08 アイシス ファーマシューティカルズ, インコーポレーテッド Methods for treating hypercholesterolemia
JP2010512421A (en) 2006-12-12 2010-04-22 イデラ ファーマシューティカルズ インコーポレイテッド Synthetic agonist of TLR9
UY30892A1 (en) 2007-02-07 2008-09-02 Smithkline Beckman Corp AKT ACTIVITY INHIBITORS
EP2125852B1 (en) 2007-02-15 2016-04-06 Ionis Pharmaceuticals, Inc. 5'-substituted-2'-f modified nucleosides and oligomeric compounds prepared therefrom
RU2015127794A (en) 2007-03-24 2018-12-21 Касл Терапьютикс, ЭлЭлСи Introduction of antisense oligonucleotides complementary to human apolipoprotein B
US7960353B2 (en) 2007-05-10 2011-06-14 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
PT2158316E (en) 2007-05-11 2015-07-20 Adynxx Inc Gene expression and pain
US8222407B2 (en) 2007-05-24 2012-07-17 Kyorin Pharmaceutical Co., Ltd. Mutilin derivative having heterocyclic aromatic ring carboxylic acid structure in substituent at 14-position
GB0710186D0 (en) 2007-05-29 2007-07-04 Texas Instr Denmark PWM loop with minimum allasing error property
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
UA98783C2 (en) 2007-06-05 2012-06-25 Нсаб, Филиал Аф Ньюросерч Свиден Аб, Свериге Disubstituted phenylpyrrolidines as modulators of cortical catecholaminergic neurotransmission
WO2008154401A2 (en) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
WO2008151833A2 (en) 2007-06-13 2008-12-18 Hochschule Mannheim Compounds for the modulation of huntingtin aggregation, methods and means for identifying such compounds
DE602007005629D1 (en) 2007-06-18 2010-05-12 Commissariat Energie Atomique Reversible siRNA silencing of a mutated and endogenous HD wild-type gene and its use in the treatment of Huntington's disease
GB0712494D0 (en) 2007-06-27 2007-08-08 Isis Innovation Substrate reduction therapy
US8278283B2 (en) 2007-07-05 2012-10-02 Isis Pharmaceuticals, Inc. 6-disubstituted or unsaturated bicyclic nucleic acid analogs
CN101795715A (en) 2007-07-09 2010-08-04 艾德拉药物股份有限公司 Stabilized immune modulatory RNA (SIMRA) compounds
TWI413530B (en) 2007-07-20 2013-11-01 Kao Corp Organopolysiloxane
AU2008281281A1 (en) 2007-07-31 2009-02-05 University Of Saskatchewan Genetic variation in Pro-Melanin-Concentrating Hormone gene affects carcass traits in cattle
EP2185700A4 (en) 2007-08-02 2010-11-24 Texas A & M Univ Sys Antisense microrna and uses therefor
CA2694973A1 (en) 2007-08-15 2009-02-19 Idera Pharmaceuticals, Inc. Toll like receptor modulators
AU2008308691B2 (en) 2007-10-01 2013-11-07 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
KR100886139B1 (en) 2007-11-13 2009-02-27 주식회사 삼천리제약 Method for preparing oligonucleotide
TW200930375A (en) 2007-12-21 2009-07-16 Exelixis Inc Benzofuropyrimidinones
TWI340765B (en) 2007-12-26 2011-04-21 Ind Tech Res Inst Oligonucleotide sequences and dna chip for identifying filamentous microorganisms and the identification method thereof
WO2009089659A1 (en) 2008-01-18 2009-07-23 Shanghai Targetdrug Co., Ltd. Pyrollidine-based compounds
CA2711587A1 (en) 2008-02-04 2009-08-13 Galapagos Nv Target sequences and methods to identify the same, useful in treatment of neurodegenerative diseases
JP2009190983A (en) 2008-02-12 2009-08-27 Tokyo Institute Of Technology Oligonucleotide derivative
WO2009117589A1 (en) 2008-03-21 2009-09-24 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucleosides and methods for their use
WO2009142822A2 (en) 2008-03-26 2009-11-26 Alnylam Pharmaceuticals, Inc. 2-f modified rna interference agents
CN103816174B (en) 2008-04-03 2017-12-05 春堤制药公司 For treating the Compounds and methods for of virus infection
DE112009001327A5 (en) 2008-04-04 2011-03-31 Universität Hamburg Process for the stereoselective synthesis of phosphorus compounds
EP2285819B1 (en) 2008-04-04 2013-10-16 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
US8679750B2 (en) 2008-05-09 2014-03-25 The University Of British Columbia Methods and compositions for the treatment of Huntington'S disease
EP2297341A4 (en) 2008-05-09 2013-01-09 Univ British Columbia Methods and compositions for the treatment of huntington's disease
US8815817B2 (en) 2008-05-15 2014-08-26 Dynavax Technologies Corporation Long term disease modification using immunostimulatory oligonucleotides
EP2291200A4 (en) 2008-05-22 2012-05-30 Isis Pharmaceuticals Inc Methods for modulating expression of rbp4
WO2009143387A2 (en) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc. Modulation of smrt expression
WO2009143391A2 (en) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc Methods for modulation expression of creb
WO2009148605A2 (en) 2008-06-04 2009-12-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
CA2728559A1 (en) 2008-07-03 2010-01-07 Exelixis Inc. Cdk modulators
WO2010030849A1 (en) 2008-09-12 2010-03-18 University Of Louisville Research Foundation, Inc. Compositions and methods for treating cancer,inhibiting proliferation, and inducing cell death
WO2010030858A1 (en) 2008-09-15 2010-03-18 Enanta Pharmaceuticals, Inc. 4'-allene-substituted nucleoside derivatives
US8691971B2 (en) 2008-09-23 2014-04-08 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating RNA interference
US8604192B2 (en) 2008-09-24 2013-12-10 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acids analogs
US8501805B2 (en) 2008-09-24 2013-08-06 Isis Pharmaceuticals, Inc. Substituted alpha-L-bicyclic nucleosides
WO2010042636A2 (en) 2008-10-07 2010-04-15 President And Fellows Of Harvard College Telomerase inhibitors and methods of use thereof
CA2738957A1 (en) 2008-10-22 2010-04-29 Quark Pharmaceuticals, Inc. Methods for treating eye disorders
WO2010048585A2 (en) 2008-10-24 2010-04-29 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
AU2009308217B2 (en) 2008-10-24 2016-01-21 Ionis Pharmaceuticals, Inc. 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
EP2370451B1 (en) 2008-12-02 2016-11-16 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
KR20110099333A (en) 2008-12-23 2011-09-07 기린두스 아메리카 인코포레이티드 Sulfurizing reagents and their use for oligonucleotides synthesis
WO2010080953A1 (en) 2009-01-08 2010-07-15 Isis Pharmaceuticals, Inc. Transgenic murine model of human lipoprotein metabolism, hypercholesterolemia and cardiovascular disease
KR20100087540A (en) 2009-01-28 2010-08-05 삼성전자주식회사 Ink composition for inkjet recording
US20120264806A1 (en) 2009-02-06 2012-10-18 Bennett C Frank Oligomeric compounds and excipients
AU2010213795A1 (en) 2009-02-10 2011-08-04 Idera Pharmaceuticals, Inc. Synthetic RNA-based agonists of TLR7
EP3424939A1 (en) 2009-03-02 2019-01-09 Alnylam Pharmaceuticals Inc. Nucleic acid chemical modifications
US9107933B2 (en) 2009-03-16 2015-08-18 Isis Pharmaceuticals, Inc. Compositions and methods of targeting apolipoprotein B for the reduction of apolipoprotein C-III
WO2010113937A1 (en) 2009-03-31 2010-10-07 武田薬品工業株式会社 Process for producing nucleoside
US8987222B2 (en) 2009-04-08 2015-03-24 University Of Massachusetts Single nucleotide polymorphism (SNP) targeting therapies for the treatment of huntington'S disease
WO2010120262A1 (en) 2009-04-14 2010-10-21 Smith Holdings, Llc Methods and compositions for the treatment of medical conditions involving cellular programming
WO2010129853A2 (en) 2009-05-07 2010-11-11 The Regents Of The University Of California TRANSDUCIBLE DELIVERY OF NUCLEIC ACIDS USING MODIFIED dsRNA BINDING DOMAINS
AU2010256836A1 (en) 2009-06-01 2012-01-19 The Regents Of The University Of California Nucleic acid delivery compositions and methods of use thereof
HUE031051T2 (en) 2009-06-05 2017-06-28 Infectious Disease Res Inst Synthetic glucopyranosyl lipid adjuvants and vaccine compositions containing them
RU2566724C9 (en) 2009-06-17 2019-03-12 Байоджен Ma Инк. Compositions and methods of modulating smn2 splicing in subject
JP5670097B2 (en) 2009-06-19 2015-02-18 花王株式会社 Two-layer separated hair cosmetic
US20120108800A1 (en) 2009-06-23 2012-05-03 Shumpei Murata Method for synthesizing nucleic acid
SG177564A1 (en) 2009-07-06 2012-02-28 Ontorii Inc Novel nucleic acid prodrugs and methods of use thereof
US8329024B2 (en) 2009-07-06 2012-12-11 Ada Technologies, Inc. Electrochemical device and method for long-term measurement of hypohalites
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
EP2451974A2 (en) 2009-07-08 2012-05-16 Idera Pharmaceuticals, Inc. Oligonucleotide-based compounds as inhibitors of toll-like receptors
WO2011010706A1 (en) 2009-07-23 2011-01-27 武田薬品工業株式会社 Fgf21 cis-element binding substance
WO2011015573A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
WO2011015572A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
UA107360C2 (en) 2009-08-05 2014-12-25 Biogen Idec Inc Bicyclic aryl sphingosine 1-phosphate analogs
WO2011017521A2 (en) 2009-08-06 2011-02-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
US8927553B2 (en) 2009-08-10 2015-01-06 Daljit Singh Dhanoa Deuterium-enriched alkyl sulfonamides and uses thereof
KR102175230B1 (en) 2009-09-11 2020-11-06 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of huntingtin expression
CN102574888A (en) 2009-09-16 2012-07-11 株式会社启拉坚 Novel protecting group for synthesizing RNA and derivative thereof
WO2011038288A1 (en) 2009-09-25 2011-03-31 Isis Pharmaceuticals, Inc. Modulation of ttc39 expression to increase hdl
JP5629324B2 (en) 2009-10-15 2014-11-19 ファイザー・インク Pyrrolo [2,3-D] pyrimidine compounds
TWI475051B (en) 2009-11-18 2015-03-01 Kao Corp Organic polysiloxane
JP5809408B2 (en) 2009-11-25 2015-11-10 花王株式会社 Hair cosmetics
US8735417B2 (en) 2009-12-17 2014-05-27 Merck Sharp & Dohme Corp. Aminopyrimidines as Syk inhibitors
CN102812361A (en) 2009-12-28 2012-12-05 阿茨拉实验室有限公司 Diagnostic Gel Composition, Method For Making A Diagnostic Gel Composition
US8653047B2 (en) 2010-01-08 2014-02-18 Isis Pharmaceuticals, Inc. Modulation of angiopoietin-like 3 expression
US8750507B2 (en) 2010-01-25 2014-06-10 Cisco Technology, Inc. Dynamic group creation for managed key servers
EP3321361B1 (en) 2010-02-08 2019-03-27 Ionis Pharmaceuticals, Inc. Selective reduction of allelic variants
AU2011213563B2 (en) 2010-02-08 2015-12-24 Ionis Pharmaceuticals, Inc. Selective reduction of allelic variants
CN102753553B (en) 2010-02-10 2016-03-30 葛兰素史密丝克莱恩有限责任公司 6-amino-2-{ [(1S)-1-methyl butyl] oxygen base }-9-[5-(piperidino)-7,9-dihydro-8H-purine-8-ketone maleate
WO2011108682A1 (en) 2010-03-05 2011-09-09 国立大学法人 東京大学 Ribonucleoside phosphorothioate manufacturing method
WO2011127175A1 (en) 2010-04-06 2011-10-13 Isis Pharmaceuticals, Inc. Modulation of cd130 (gp130) expression
AU2011237426A1 (en) 2010-04-07 2012-11-22 Isis Pharmaceuticals, Inc. Modulation of CETP expression
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
KR101869570B1 (en) 2010-04-28 2018-06-20 아이오니스 파마수티컬즈, 인코포레이티드 Modified nucleosides and oligomeric compounds prepared therefrom
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
WO2011139911A2 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Lipid formulated single stranded rna
US8906607B2 (en) 2010-04-30 2014-12-09 Cellectis Method for modulating double-strand break-induced homologous recombination
GB201008902D0 (en) 2010-05-27 2010-07-14 Imp Innovations Ltd Membrane enhanced polymer sythesis
WO2012030683A2 (en) 2010-08-31 2012-03-08 Merck Sharp & Dohme Corp. Novel single chemical entities and methods for delivery of oligonucleotides
JP5868324B2 (en) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan Asymmetric auxiliary group
US9238812B2 (en) 2010-09-30 2016-01-19 Lsip, Llc Agent for suppressing expression of dominant mutant gene
KR101381048B1 (en) 2010-10-20 2014-04-14 씨제이제일제당 (주) A microorganism producing O-phosphoserine and the method of producing L-cysteine or derivatives thereof from O-phosphoserine using the same
ES2663009T3 (en) 2010-10-29 2018-04-10 Sirna Therapeutics, Inc. Inhibition of RNA-mediated gene expression using short interference nucleic acids (ANic)
EP3260540A1 (en) 2010-11-12 2017-12-27 The General Hospital Corporation Polycomb-associated non-coding rnas
US8822671B2 (en) 2010-11-30 2014-09-02 The University Of Tokyo 2'-O-modified RNA
US20140050778A1 (en) 2010-12-28 2014-02-20 University Of Rochester Nucleic acid binding compounds, methods of making, and use thereof
EP3467109A1 (en) 2011-02-08 2019-04-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2012109667A1 (en) 2011-02-12 2012-08-16 University Of Iowa Research Foundation Therapeutic compounds
WO2012151324A1 (en) 2011-05-02 2012-11-08 Isis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with usher syndrome
FR2975600B1 (en) 2011-05-24 2013-07-05 Assist Publ Hopitaux De Paris AGENTS FOR THE TREATMENT OF TUMORS
AU2012283994A1 (en) 2011-07-19 2014-03-06 University Of Idaho Embodiments of a probe and method for targeting nucleic acids
RU2014105311A (en) 2011-07-19 2015-08-27 Уэйв Лайф Сайенсес Пте. Лтд. METHODS FOR SYNTHESIS OF FUNCTIONALIZED NUCLEIC ACIDS
EP2742136B1 (en) 2011-08-11 2017-09-27 Ionis Pharmaceuticals, Inc. Gapped oligomeric compounds comprising 5'-modified deoxyribonucleosides in the gap and uses thereof
EP2751269B1 (en) 2011-08-29 2016-03-23 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
US20140080896A1 (en) 2011-08-30 2014-03-20 The Regents Of The University Of California Identification of small molecules that facilitate therapeutic exon skipping
EP2751284B1 (en) 2011-08-31 2017-01-11 The University Of Manchester Method for diagnosing a neurodegenerative disease.
US20140255936A1 (en) 2011-09-09 2014-09-11 Mayo Foundation For Medical Education And Research Detecting frontotemporal dementia and amyotrophic lateral sclerosis
EP2785729B1 (en) 2011-11-30 2020-11-04 Sarepta Therapeutics, Inc. Oligonucleotides for treating expanded repeat diseases
KR200460641Y1 (en) 2011-12-07 2012-06-04 이형호 Easy to carry wine glass
EP2790736B1 (en) 2011-12-12 2018-01-31 Oncoimmunin, Inc. In vivo delivery of oligonucleotides
AU2012353330B2 (en) 2011-12-16 2018-04-19 National University Corporation Tokyo Medical And Dental University Chimeric double-stranded nucleic acid
CN102675386B (en) 2011-12-24 2014-07-02 河南科技大学 Method for separating and purifying gentiamarin
US8957042B2 (en) 2012-03-07 2015-02-17 The Texas A&M University System Cancer treatment targeting non-coding RNA overexpression
ES2621217T3 (en) 2012-03-13 2017-07-03 Gilead Sciences, Inc. 2'-substituted carba-nucleoside analogues for antiviral treatment
KR20130114435A (en) 2012-04-09 2013-10-17 삼성전자주식회사 Biomolecule detection apparatus including a plurality of electrode
HUE033431T2 (en) 2012-04-23 2017-11-28 Biomarin Tech Bv RNA modulating oligonucleotides with improved characteristics for the treatment of neuromuscular disorders
WO2013177219A1 (en) 2012-05-22 2013-11-28 Idenix Pharmaceuticals, Inc. D-amino acid compounds for liver disease
IN2014DN10144A (en) 2012-05-30 2015-08-21 Hokkaido System Science Co Ltd
SG11201500243WA (en) 2012-07-13 2015-04-29 Shin Nippon Biomedical Lab Ltd Chiral nucleic acid adjuvant
SG11201500239VA (en) 2012-07-13 2015-03-30 Wave Life Sciences Japan Asymmetric auxiliary group
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
EP2879718B1 (en) 2012-08-06 2023-06-07 Alnylam Pharmaceuticals, Inc. Processes for the preparation of carbohydrate conjugated rna agents
CN104736551B (en) 2012-08-15 2017-07-28 Ionis制药公司 The method for preparing oligomeric compounds using improved end-blocking scheme
DK2906256T3 (en) 2012-10-12 2018-11-19 Ionis Pharmaceuticals Inc SELECTIVE ANTISENSE COMPOUNDS AND APPLICATIONS THEREOF
ES2762326T5 (en) 2012-10-15 2023-04-27 Ionis Pharmaceuticals Inc Methods to modulate the expression of C9ORF72
WO2014062691A2 (en) 2012-10-15 2014-04-24 Isis Pharmaceuticals, Inc. Compositions for modulating c9orf72 expression
EP2906697A4 (en) 2012-10-15 2016-06-22 Ionis Pharmaceuticals Inc Methods for monitoring c9orf72 expression
EP2725029A1 (en) 2012-10-29 2014-04-30 Laboratoire Biodim New antibacterial compounds and biological applications thereof
JP2015535261A (en) 2012-10-29 2015-12-10 コクリスタル ファーマ,インコーポレイテッド Pyrimidine nucleotides and their monophosphate prodrugs for the treatment of viral infections and cancer
JP6358955B2 (en) 2012-10-31 2018-07-18 武田薬品工業株式会社 Novel modified nucleic acid
US20150291958A1 (en) 2012-11-15 2015-10-15 Roche Innovation Center Copenhagen A/S Anti apob antisense conjugate compounds
WO2014080004A1 (en) 2012-11-26 2014-05-30 Santaris Pharma A/S Compositions and methods for modulation of fgfr3 expression
WO2014099941A1 (en) 2012-12-19 2014-06-26 Idenix Pharmaceuticals, Inc. 4'-fluoro nucleosides for the treatment of hcv
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
DK2951305T3 (en) 2013-01-30 2018-10-29 Hoffmann La Roche LNA oligonucleotide KULHYDRATKONJUGATER
WO2014121287A2 (en) 2013-02-04 2014-08-07 Isis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
CA2900238A1 (en) 2013-02-22 2014-08-28 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) molecules containing a 2' internucleoside linkage
AU2014222150A1 (en) 2013-03-01 2015-09-10 National University Corporation Tokyo Medical And Dental University Chimeric single-stranded antisense polynucleotides and double-stranded antisense agent
BR112015024526A2 (en) 2013-03-28 2017-07-18 Syngenta Ltd neonicotinoid resistant pest control methods
JP2016520536A (en) 2013-03-28 2016-07-14 シンジェンタ パーティシペーションズ アーゲー Neonicotinoid-resistant pest control method
SG10201906382QA (en) 2013-05-01 2019-08-27 Ionis Pharmaceuticals Inc Compositions and methods for modulating hbv and ttr expression
JP6477464B2 (en) 2013-05-24 2019-03-06 味の素株式会社 Method for producing morpholino oligonucleotide
DK3004352T3 (en) 2013-05-24 2017-11-27 Roche Innovation Ct Copenhagen As B-cell Cl1 / Lymphoma 11a (bcl11a) oligonucleotide modulators and uses thereof
CN105264073A (en) 2013-05-30 2016-01-20 国立大学法人东京医科齿科大学 Double-stranded agents for delivering therapeutic oligonucleotides
US10190117B2 (en) 2013-06-16 2019-01-29 National University Corporation Tokyo Medical And Dental University Double-stranded antisense nucleic acid with exon-skipping effect
CA2916252A1 (en) 2013-06-21 2014-12-24 Isis Pharmaceuticals, Inc. Compositions and methods for modulation of target nucleic acids
BR112015032432B1 (en) 2013-06-27 2023-02-07 Roche Innovation Center Copenhagen A/S ANTI-SENSE OLIGOMER, ANTI-SENSE OLIGONUCLEOTIDE CONJUGATES, PHARMACEUTICAL COMPOSITION, USE THEREOF FOR THE TREATMENT OF HYPERCHOLESTEROLEMIA OR RELATED DISORDERS, AND IN VITRO METHOD FOR REDUCING THE EXPRESSION LEVELS AND/OR THE ACTIVITY OF PCSK9 IN A CELL
TWI657819B (en) 2013-07-19 2019-05-01 美商Ionis製藥公司 Compositions for modulating tau expression
US10894963B2 (en) 2013-07-25 2021-01-19 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
US10435430B2 (en) 2013-07-31 2019-10-08 Ionis Pharmaceuticals, Inc. Methods and compounds useful in conditions related to repeat expansion
BR112016004352A2 (en) 2013-09-06 2017-09-12 Syngenta Participations Ag insecticide compounds
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US9988627B2 (en) 2013-10-04 2018-06-05 Novartis Ag Formats for organic compounds for use in RNA interference
CN105637090B (en) 2013-10-11 2020-10-16 Ionis制药公司 Compositions for modulating expression of C9ORF72
US20160237432A1 (en) 2013-10-14 2016-08-18 Ionis Pharmaceuticals, Inc. Methods for modulating expression of c9orf72 antisense transcript
US20160230172A1 (en) 2013-10-14 2016-08-11 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
JP2016537341A (en) 2013-11-11 2016-12-01 サンガモ バイオサイエンシーズ, インコーポレイテッド Methods and compositions for treating Huntington's disease
RU2016122168A (en) 2013-11-14 2017-12-19 Рош Инновейшен Сентер Копенгаген А/С ANTI-SENSE CONJUGATES AIMED AT APOLIPOPROTEIN B
EP2918275B1 (en) 2013-12-13 2016-05-18 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
JPWO2015108047A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant having immunity induction activity and immunity induction activator
JPWO2015108046A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antiallergic agent having antiallergic action
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
DK3094728T3 (en) 2014-01-16 2022-05-16 Wave Life Sciences Ltd KIRALT DESIGN
JP6756700B2 (en) 2014-03-18 2020-09-16 ユニバーシティ オブ マサチューセッツ RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
DK3137476T3 (en) 2014-04-28 2019-11-18 Ionis Pharmaceuticals Inc LINKER-MODIFIED OLIGOMER COMPOUNDS
SI3137605T1 (en) 2014-05-01 2021-02-26 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
AU2015255877B2 (en) 2014-05-08 2020-03-26 Chdi Foundation, Inc. Methods and compositions for treating huntington's disease
US9523093B2 (en) 2014-05-20 2016-12-20 University Of Iowa Research Foundation Huntington's disease therapeutic compounds
US20160017327A1 (en) 2014-07-11 2016-01-21 The Johns Hopkins University Phosphorodiamidate morpholino oligomers (pmos) and their use in suppression of mutant huntingtin expression and attenuation of neurotoxicity
CA2955250A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
EP2982758A1 (en) 2014-08-04 2016-02-10 Centre Hospitalier Universitaire Vaudois (CHUV) Genome editing for the treatment of huntington's disease
AU2015300046B2 (en) 2014-08-07 2019-05-09 Takeda Pharmaceutical Company Limited Cationic lipid
WO2016024205A1 (en) 2014-08-15 2016-02-18 Pfizer Inc. Oligomers targeting hexanucleotide repeat expansion in human c9orf72 gene
WO2016037191A1 (en) 2014-09-05 2016-03-10 Health Research, Inc. Use of huntingtin-derived plasmids and peptides for active immunization as a huntington's disease (hd) therapeutic
CN107250362B (en) 2014-11-17 2021-10-22 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
JP2017536119A (en) 2014-11-19 2017-12-07 ロシュ イノベーション センター コペンハーゲン エーエス LNA gapmer oligonucleotides containing chiral phosphorothioate linkages
WO2016096938A1 (en) 2014-12-16 2016-06-23 Roche Innovation Center Copenhagen A/S Chiral toxicity screening method
PT3237618T (en) 2014-12-24 2019-07-04 Uniqure Ip Bv Rnai induced huntingtin gene suppression
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
DK3253875T3 (en) 2015-02-04 2020-04-14 H Hoffmann La Roche Ag Tau antisense oligomers and uses thereof
EP3253871A1 (en) 2015-02-04 2017-12-13 Bristol-Myers Squibb Company Lna oligonucleotides with alternating flanks
US11761951B2 (en) 2015-02-04 2023-09-19 Bristol-Myers Squibb Company Methods of selecting therapeutic molecules
SG11201706444TA (en) 2015-02-10 2017-09-28 Genzyme Corp VARIANT RNAi
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
EP3262174A4 (en) 2015-02-23 2018-10-17 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing antisense activity
WO2016141236A1 (en) 2015-03-03 2016-09-09 Ionis Pharmaceuticals, Inc. Compositions for modulating mecp2 expression
WO2016145142A1 (en) 2015-03-10 2016-09-15 Emory University Nucleotide and nucleoside therapeutics compositions and uses related thereto
WO2016154096A1 (en) 2015-03-20 2016-09-29 Ionis Pharmaceuticals, Inc. Modulation of smggds expression
CN116004624A (en) 2015-04-03 2023-04-25 马萨诸塞大学 Oligonucleotide compounds for targeting huntingtin mRNA
WO2016164896A2 (en) 2015-04-10 2016-10-13 Ionis Pharmaceuticals, Inc. Modulation of smn expression
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
RS60230B1 (en) 2015-04-16 2020-06-30 Ionis Pharmaceuticals Inc Compositions for modulating c9orf72 expression
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
EP3313989A4 (en) 2015-06-29 2018-12-05 Ionis Pharmaceuticals, Inc. Modified crispr rna and modified single crispr rna and uses thereof
MY192997A (en) 2015-07-10 2022-09-20 Ionis Pharmaceuticals Inc Modulators of diacyglycerol acyltransferase 2 (dgat2)
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
EP3919619A1 (en) 2015-07-17 2021-12-08 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
TW201718620A (en) 2015-07-27 2017-06-01 阿尼拉製藥公司 Xanthine dehydrogenase (XDH) iRNA compositions and methods of use thereof
SG10201912341SA (en) 2015-07-31 2020-02-27 Alnylam Pharmaceuticals Inc TRANSTHYRETIN (TTR) iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES
WO2017032726A1 (en) 2015-08-24 2017-03-02 Roche Innovation Center Copenhagen A/S Lna-g process
EP3340994A1 (en) 2015-08-25 2018-07-04 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a proprotein convertase subtilisin kexin (pcsk9) gene-associated disorder
CN114525280A (en) 2015-09-02 2022-05-24 阿尔尼拉姆医药品有限公司 iRNA compositions of programmed cell death 1 ligand 1(PD-L1) and methods of use thereof
WO2017048620A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
US10874686B2 (en) 2015-10-01 2020-12-29 Memorial Sloan-Kettering Cancer Center Anthranilyl-adenosinemonosulfamate analogs and uses thereof
US20180273573A1 (en) 2015-10-01 2018-09-27 Memorial Sloan-Kettering Cancer Center Inhibitors of menaquinone biosynthesis
JP6893505B2 (en) 2015-10-02 2021-06-23 ロシュ イノベーション センター コペンハーゲン エーエス Oligonucleotide Conjugation Method
IL258230B (en) 2015-10-09 2022-09-01 Wave Life Sciences Ltd Oligonucleotide compositions and methods thereof
EP3183347A4 (en) 2015-10-17 2018-04-18 Lifesplice Pharma LLC Splice modulating oligonucleotides and methods of use thereof
US10955407B2 (en) 2015-10-22 2021-03-23 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
WO2017068087A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S Oligonucleotide detection method
US11260073B2 (en) 2015-11-02 2022-03-01 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating C90RF72
CA3004799A1 (en) 2015-11-12 2017-05-18 F. Hoffmann-La Roche Ag Oligonucleotides for inducing paternal ube3a expression
MA43822A (en) 2016-03-13 2018-11-28 Wave Life Sciences Ltd COMPOSITIONS AND SYNTHESIS OF PHOSPHORAMIDITE AND OLIGONUCLEOTIDES
MY194912A (en) 2016-03-14 2022-12-22 Hoffmann La Roche Oligonucleotides for Reduction of PD-L1 Expression
KR102372122B1 (en) 2016-03-18 2022-03-07 로슈 이노베이션 센터 코펜하겐 에이/에스 Acyl-protected L-LNA-guanosine monomer
US20230000893A1 (en) 2016-03-23 2023-01-05 Emory University Antiviral Agents and Nucleoside Analogs for Treatment of Zika Virus
ES2933435T3 (en) 2016-04-13 2023-02-08 Ionis Pharmaceuticals Inc Methods to reduce the expression of C9ORF72
US11248019B2 (en) 2016-04-14 2022-02-15 Hoffmann-La Roche Inc. Trityl-mono-GalNAc compounds and their use
MA45270A (en) 2016-05-04 2017-11-09 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
MA45290A (en) 2016-05-04 2019-03-13 Wave Life Sciences Ltd PROCESSES AND COMPOSITIONS OF BIOLOGICALLY ACTIVE AGENTS
CN109311925B (en) 2016-05-12 2022-06-03 罗氏创新中心哥本哈根有限公司 Enhanced coupling of sterically defined oxaazaphospholane phosphoramidite monomers to nucleosides or oligonucleotides
WO2017194664A1 (en) 2016-05-13 2017-11-16 Roche Diagnostics Gmbh Protein-based sample collection matrices and devices
US10882884B2 (en) 2016-05-18 2021-01-05 Eth Zurich Stereoselective synthesis of phosphorothioate oligoribonucleotides
US11013757B2 (en) 2016-06-03 2021-05-25 Wave Life Sciences Ltd. Oligonucleotides, compositions and methods thereof
CN109310765A (en) 2016-06-20 2019-02-05 领先基因生物技术股份有限公司 Antibody-drug conjugates
US20190264267A1 (en) 2016-07-25 2019-08-29 Wave Life Sciences Ltd. Phasing
WO2018098264A1 (en) 2016-11-23 2018-05-31 Wave Life Sciences Ltd. Compositions and methods for phosphoramidite and oligonucleotide synthesis

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11390642B2 (en) 2016-08-23 2022-07-19 Dicerna Pharmaceuticals, Inc. Compositions comprising reversibly modified oligonucleotides and uses thereof

Also Published As

Publication number Publication date
AU2010270714B2 (en) 2015-08-13
JP2012532199A (en) 2012-12-13
CL2012000021A1 (en) 2012-07-20
AU2010270714A1 (en) 2012-02-16
SG10201403841QA (en) 2014-09-26
US9744183B2 (en) 2017-08-29
US10307434B2 (en) 2019-06-04
KR101885383B1 (en) 2018-08-03
IN2012DN00720A (en) 2015-06-19
MX342945B (en) 2016-10-18
EP2451461A4 (en) 2013-05-29
CN102596204A (en) 2012-07-18
MX2012000380A (en) 2012-05-23
RU2012102480A (en) 2013-08-20
IL217370A0 (en) 2012-02-29
CN102596204B (en) 2016-11-23
US20160347784A1 (en) 2016-12-01
WO2011005761A8 (en) 2012-02-02
IL217370A (en) 2017-09-28
RU2612521C2 (en) 2017-03-09
BR112012000828A2 (en) 2016-02-23
EP2451461A1 (en) 2012-05-16
US20120316224A1 (en) 2012-12-13
JP5998326B2 (en) 2016-09-28
SG177564A1 (en) 2012-02-28
BR112012000828A8 (en) 2017-10-10
WO2011005761A1 (en) 2011-01-13
JP2015205910A (en) 2015-11-19
KR20120046238A (en) 2012-05-09

Similar Documents

Publication Publication Date Title
US10307434B2 (en) Nucleic acid prodrugs and methods of use thereof
US10329318B2 (en) Method for the synthesis of phosphorus atom modified nucleic acids
US20110294869A1 (en) Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
EP2845607A1 (en) Antisense oligonucleotides with improved pharmacokinetic properties
US20230123981A1 (en) 4&#39;-o-methylene phosphonate nucleic acids and analogues thereof
JP2023537499A (en) Systemic delivery of oligonucleotides
JP7263236B2 (en) Novel bicyclic nucleosides and oligomers prepared therefrom
US9463200B2 (en) Cell-penetrating oligonucleotides
AU2015255202B2 (en) Novel nucleic acid prodrugs and methods of use thereof
RU2797833C1 (en) Oligonucleotide compositions and methods related to them

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20150703

FZDE Discontinued

Effective date: 20200220