Methods for the synthesis of functionalized nucleic acids

Information

  • Patent Grant
  • 10280192
  • Patent Number
    10,280,192
  • Date Filed
    Friday, March 30, 2018
    6 years ago
  • Date Issued
    Tuesday, May 7, 2019
    5 years ago
Abstract
The present application, among other things, provides technologies, e.g., reagents, methods, etc. for preparing oligonucleotides comprising phosphorothiotriesters linkages. In some embodiments, provided methods comprise reacting an H-phosphonate of structure Ia or Ib with a silylating reagent to provide a silyloxyphosphonate, and reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIa or IIb to provide an oligonucleotide of structure IIIa or IIIb. In some embodiments, provided methods comprise reacting an H-phosphonate of structure Ic with a silylating reagent to provide a silyloxyphosphonate, reacting the silyloxyphosphonate with a bis(thiosulfonate) reagent of structure IVc to provide a phosphorothiotriester comprising a thiosulfonate group of structure Vc, and then reacting the phosphorothiotriester comprising a thiosulfonate group of structure Vc with a nucleophile of structure VIc to provide an oligonucleotide of structure IIIc. In some embodiments, the present application provides a thiosulfonate reagent of structure IIa:
Description
BACKGROUND OF THE INVENTION

Oligonucleotides are useful in therapeutic, diagnostic, research and nanomaterials applications. The use of natural sequences of DNA or RNA for therapeutics is limited because of their instability against extra and intracellular nucleases, poor cell penetration and distribution. Additionally, in vitro studies have shown that the properties of antisense nucleotides such as binding affinity, sequence specific binding to the complementary RNA (Cosstick and Eckstein, 1985; LaPlanche et al., 1986; Latimer et al., 1989; Hacia et al., 1994; Mesmaeker et al., 1995), stability to nucleases are affected by the configurations of the phosphorous atoms Therefore, there is a need for modified oligonucleotides to impart stability towards ubiquitous nucleases, increase binding affinity towards complementary RNA and increase cell penetration and bio-distribution for a number of in-vitro and in-vivo applications.


SUMMARY OF THE INVENTION

Described herein are methods for the synthesis of novel functionalized nucleic acids and nucleic acid prodrugs. In some embodiments, the nucleic acids comprise chiral phosphorous moieties.


One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa comprising the steps of:

    • i) reacting an H-phosphonate of structure Ia with an silylating reagent to provide a silyloxyphosphonate; and
    • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIa to provide a phosphorothiotriester of structure IIIa;
    • wherein,
    • the H-phosphonate of structure Ia has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200; and

    • the thiosulfonate reagent of structure IIa has the following structure:







embedded image



wherein,

    • X is alkyl, cycloalkyl, or heteroaryl;
    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
    • and the phosphorothiotriester of structure IIIa has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;

    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;

    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200.





Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein W is O.


Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein R1 is selected from:




embedded image



and

    • R2 is selected from:




embedded image


Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from

  • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
  • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
  • 1-(trimethylsilyl)imidazole;
  • N-trimethylsilyl-N-methyl trifluoroacetamide;
  • bis(dimethylamino)dimethylsilane;
  • bromotrimethylsilane;
  • chlorodimethyl(pentafluorophenyl)silane;
  • chlorotriethylsilane;
  • chlorotriisopropylsilane;
  • chlorotrimethylsilane;
  • dichlorodimethylsilane;
  • hexamethyldisilazane;
  • N,N′-bis(trimethylsilyl)urea;
  • N,N-bis(trimethylsilyl)methylamine;
  • N,N-dimethyltrimethylsilylamine;
  • N,O-bis(trimethylsilyl)acetamide;
  • N,O-bis(trimethylsilyl)carbamate;
  • N,O-bis(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-trimethylsilylacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • trimethylsilyltriflate;
  • triethylsilyltriflate;
  • triisopropylsilyltriflate; or
  • tert-butyldimethyl silyltriflate.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethyl silyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.


Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.


One embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb comprising the steps of:

    • i) reacting a H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib with an silylating reagent to provide a silyloxyphosphonate; and
    • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIb to provide a phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb;
    • wherein,
    • the H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, NH, or CH2;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200; and

    • the thiosulfonate reagent of structure IIb has the following structure:







embedded image



wherein,

    • X is alkyl, cycloalkyl, aryl, or heteroaryl;
    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
    • and the chiral phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, NH, or CH2;

    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;

    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;

    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200.





Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein W is O.


Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein R1 is selected from:




embedded image



and

    • R2 is selected from:




embedded image


Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein the silylating reagent is selected from

  • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
  • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
  • 1-(trimethylsilyl)imidazole;
  • N-trimethylsilyl-N-methyl trifluoroacetamide;
  • bis(dimethylamino)dimethylsilane;
  • bromotrimethylsilane;
  • chlorodimethyl(pentafluorophenyl)silane;
  • chlorotriethylsilane;
  • chlorotriisopropylsilane;
  • chlorotrimethylsilane;
  • dichlorodimethylsilane;
  • hexamethyldisilazane;
  • N,N′-bis(trimethylsilyl)urea;
  • N,N-bis(trimethylsilyl)methylamine;
  • N,N-dimethyltrimethylsilylamine;
  • N,O-bis(trimethylsilyl)acetamide;
  • N,O-bis(trimethylsilyl)carbamate;
  • N,O-bis(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-trimethylsilylacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • trimethyl silyltriflate;
  • triethylsilyltriflate;
  • triisopropylsilyltriflate; or
  • tert-butyldimethyl silyltriflate.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethyl silyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.


Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.


One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIc comprising the steps of:

    • i) reacting a H-phosphonate of structure Ic with an silylating reagent to provide a silyloxyphosphonate;
    • ii) reacting the silyloxyphosphonate with a bis(thiosulfonate) reagent of structure IVc to provide a phosphorothiotriester comprising a thiosulfonate group of structure Vc;
    • iii) reacting the phosphorothiotriester comprising a thiosulfonate group of structure Vc with a nucleophile of structure VIc to provide the phosphorothiotriesters of structure IIIc;
    • wherein,
    • the H-phosphonate of structure Ic has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200; and

    • the bis(thiosulfonate) reagent of structure IVc has the following structure:







embedded image



wherein,

    • X is alkylene, alkenylene, arylene, or heteroarylene;
    • each R6 is independently alkyl, cycloalkyl, aryl, or heteroaryl;
    • the nucleophile of structure VIc has the following structure:
    • R7—SH, wherein R7 is selected from alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
    • and phosphorothiotriesters of structure IIIc has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R is R7—S—S—X—

    • R7 is alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;

    • X is alkylene, alkenylene, arylene, or heteroarylene;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;

    • n is between 1 and about 200; and

    • wherein the phosphorous linkages of the H-phosphonate of structure Ic, the phosphorothiotriester comprising a thiosulfonate group of structure Vc, and the phosphorothiotriesters of structure IIIc may optionally comprise non-stereorandom phosphorous linkages.





Another embodiment provides the process wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH2. Another embodiment provides the process wherein W is O.


Another embodiment provides the process wherein R6 is methyl.


Another embodiment provides the process wherein bis(thiosulfonate) reagent of structure IVc is selected from:




embedded image


Another embodiment provides the process wherein the nucleophile of structure VIc has the following structure:




embedded image


Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from

  • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
  • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
  • 1-(trimethylsilyl)imidazole;
  • N-trimethylsilyl-N-methyl trifluoroacetamide;
  • bis(dimethylamino)dimethylsilane;
  • bromotrimethylsilane;
  • chlorodimethyl(pentafluorophenyl)silane;
  • chlorotriethylsilane;
  • chlorotriisopropylsilane;
  • chlorotrimethylsilane;
  • dichlorodimethylsilane;
  • hexamethyldisilazane;
  • N,N′-bis(trimethylsilyl)urea;
  • N,N-bis(trimethylsilyl)methylamine;
  • N,N-dimethyltrimethylsilylamine;
  • N,O-bis(trimethylsilyl)acetamide;
  • N,O-bis(trimethylsilyl)carbamate;
  • N,O-bis(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-trimethylsilylacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • trimethyl silyltriflate;
  • triethylsilyltriflate;
  • triisopropylsilyltriflate; or
  • tert-butyldimethyl silyltriflate.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethyl silyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.


Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.


INCORPORATION BY REFERENCE

All publications and patent applications mentioned in this specification are herein incorporated by reference 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

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 of which:



FIG. 1 provides the 31P NMR spectrum of Compound 100S in CD3CN as described in Example 6;



FIG. 2 provides the 31P NMR spectrum of Compound 100S in CD3CN after adding BSTFA as described in Example 6;



FIG. 3 provides the 31P NMR spectrum of Compound 100S in CD3CN after adding BSTFA, TEA and MTS as described in Example 6;



FIG. 4 provides the 31P NMR spectrum of Compound 100R in CD3CN as described in Example 6;



FIG. 5 provides the 31P NMR spectrum of Compound 100R in CD3CN as described in Example 6; and



FIG. 6 provides the 31P NMR spectrum of Compound 100R in CD3CN after adding BSTFA, TEA and MTS as described in Example 6.





DETAILED DESCRIPTION OF THE INVENTION

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

Unless otherwise noted, the use of general chemical terms, such as though not limited to “alkyl,” “amine,” “aryl,” are unsubstituted.


As used herein, C1-Cx includes C1-C2, C1-C3 . . . C1-Cx. By way of example only, a group designated as “C1-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 C1-C2 and C1-C3. Thus, by way of example only, “C1-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 10 carbon atoms.


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.


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-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-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 “C1-6 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 unsubstituted.


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 unsubstituted.


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 unsubstituted.


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 —O—O—, —S—S—, —O—S—, —S—O—, N—N═, —N═N—, —N═N—NH—, —P(O)2—, —O—P(O)2—, —P(O)2—O—, —S(O)—, —S(O)2—, —SnH2— and the like.


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-1-fluoro-1-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.


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.


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:




embedded image



and the like.

    • In one embodiment, the “cycloalkyl” is substituted. Unless otherwise indicated, the “cycloalkyl” is unsubstituted.


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:




embedded image



and the like.


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 unsubstituted.


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 unsubstituted.


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 O 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:




embedded image



and the like.

    • In one embodiment, the “heteroaryl” is substituted. Unless otherwise indicated, the “heteroaryl” is unsubstituted.


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., C1-C6 heterocycle), at least one non-carbon atom (the heteroatom) must be present in the ring. Designations such as “C1-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 unsubstituted.


The terms “halogen”, “halo” or “halide” as used herein, alone or in combination refer to fluoro, chloro, bromo and/or iodo.


The compounds, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, such as (R)- or (S)-. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both Z and E geometric isomers (e.g., cis or trans). Likewise, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included.


A “stereoisomer” refers to the relationship between two or more compounds made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not superimposable. The term “enantiomer” refers to two stereoisomers that are nonsuperimposeable mirror images of one another. It is contemplated that the various stereoisomers of the compounds disclosed herein, and mixtures thereof, are within the scope of the present disclosure and specifically includes enantiomers.


A “tautomer” refers to a compound wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible. The compounds presented herein may exist as tautomers. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. Some examples of tautomeric equilibrium are shown below.




embedded image


The term “non-stereorandom phosphorous linkage(s)” as used herein refers to a chiral phosphorous atom in the phosphodiester, or other isosteric linkage type, internucleotide linkage. For embodiments comprising more than one phosphorous internucleotide linkage, the handedness of chirality at phosphorous is independently selected at each phosphorous atom. In one embodiment, the oligonucleotide described herein is a pure diastereomer. In another embodiment, the oligonucleotide is greater that 95% diastereomeric purity. In another embodiment, the oligonucleotide is greater that 90% diastereomeric purity.


“Optional” or “optionally” means that a subsequently described event or circumstance may or may not occur and that the description includes instances when the event or circumstance occurs and instances in which it does not. For example, “optionally substituted alkyl” means that the alkyl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.


Certain Nucleic Acid Terminology

Natural nucleic acids have a phosphate backbone; artificial nucleic acids may contain other types of backbones, but contain the same bases.


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.


The term “nucleoside” refers to a moiety wherein a nucleobase or a modified nucleobase is covalently bound to a sugar or modified sugar.


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.


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.


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.


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).


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.


The term “chiral reagent” refers to a compound that is chiral or enantiopure and can be used for asymmetric induction in nucleic acid synthesis.


The term “chiral ligand” or “chiral auxiliary” refers to a moiety that is chiral or enantiopure and controls the stereochemical outcome of a reaction.


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.


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.


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.


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.


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.


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).


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.


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.


Synthetic Methods for the Preparation Novel Functionalized Nucleic Acids and Nucleic Acid Prodrugs


Described herein are methods for the synthesis of novel functionalized nucleic acids and nucleic acid prodrugs. In some embodiments, the nucleic acids comprise chiral phosphorous moieties.


One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa comprising the steps of:

    • i) reacting an H-phosphonate of structure Ia with an silylating reagent to provide a silyloxyphosphonate; and
    • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIa to provide a phosphorothiotriester of structure IIIa;
    • wherein,
    • the H-phosphonate of structure Ia has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200; and

    • the thiosulfonate reagent of structure IIa has the following structure:







embedded image



wherein,

    • X is alkyl, cycloalkyl, or heteroaryl;
    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
    • and the phosphorothiotriester of structure IIIa has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;

    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;

    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200.





Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein W is O.


Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein R1 is selected from:




embedded image



and

    • R2 is selected from:




embedded image


Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from

  • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
  • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
  • 1-(trimethylsilyl)imidazole;
  • N-trimethylsilyl-N-methyl trifluoroacetamide;
  • bis(dimethylamino)dimethylsilane;
  • bromotrimethylsilane;
  • chlorodimethyl(pentafluorophenyl)silane;
  • chlorotriethylsilane;
  • chlorotriisopropylsilane;
  • chlorotrimethylsilane;
  • dichlorodimethylsilane;
  • hexamethyldisilazane;
  • N,N′-bis(trimethylsilyl)urea;
  • N,N-bis(trimethylsilyl)methylamine;
  • N,N-dimethyltrimethylsilylamine;
  • N,O-bis(trimethylsilyl)acetamide;
  • N,O-bis(trimethylsilyl)carbamate;
  • N,O-bis(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-trimethylsilylacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • trimethyl silyltriflate;
  • triethylsilyltriflate;
  • triisopropylsilyltriflate; or
  • tert-butyldimethyl silyltriflate.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethyl silyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.


Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.


One embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb comprising the steps of:

    • i) reacting a H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib with an silylating reagent to provide a silyloxyphosphonate; and
    • ii) reacting the silyloxyphosphonate with a thiosulfonate reagent of structure IIb to provide a phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb;
    • wherein,
    • the H-phosphonate comprising non-stereorandom phosphorous linkages of structure Ib has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, NH, or CH2;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200; and

    • the thiosulfonate reagent of structure IIb has the following structure:







embedded image



wherein,

    • X is alkyl, cycloalkyl, aryl, or heteroaryl;
    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;
    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;
    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;
    • and the chiral phosphorothiotriester comprising non-stereorandom phosphorous linkages of structure IIIb has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, NH, or CH2;

    • R is alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or R1-R2;

    • R1 is selected from —S-alkenylene-, —S-alkylene-, —S-alkylene-aryl-alkylene-, —S—CO-aryl-alkylene-, or —S—CO-alkylene-aryl-alkylene-;

    • R2 is selected from heterocyclo-alkylene-S—, heterocyclo-alkenylene-S—, aminoalkyl-S—, or (alkyl)4N-alkylene-S—;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200.





Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein W is O.


Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein R1 is selected from:




embedded image



and

    • R2 is selected from:




embedded image


Another embodiment provides a process for the preparation of phosphorothiotriesters comprising non-stereorandom phosphorous linkages of structure IIIb, wherein the silylating reagent is selected from

  • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
  • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
  • 1-(trimethylsilyl)imidazole;
  • N-trimethylsilyl-N-methyl trifluoroacetamide;
  • bis(dimethylamino)dimethylsilane;
  • bromotrimethylsilane;
  • chlorodimethyl(pentafluorophenyl)silane;
  • chlorotriethylsilane;
  • chlorotriisopropylsilane;
  • chlorotrimethylsilane;
  • dichlorodimethylsilane;
  • hexamethyldisilazane;
  • N,N′-bis(trimethylsilyl)urea;
  • N,N-bis(trimethylsilyl)methylamine;
  • N,N-dimethyltrimethylsilylamine;
  • N,O-bis(trimethylsilyl)acetamide;
  • N,O-bis(trimethylsilyl)carbamate;
  • N,O-bis(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-trimethylsilylacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • trimethyl silyltriflate;
  • triethylsilyltriflate;
  • triisopropylsilyltriflate; or
  • tert-butyldimethyl silyltriflate.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethyl silyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.


Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.


One embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIc comprising the steps of:

    • i) reacting a H-phosphonate of structure Ic with an silylating reagent to provide a silyloxyphosphonate;
    • ii) reacting the silyloxyphosphonate with a bis(thiosulfonate) reagent of structure IVc to provide a phosphorothiotriester comprising a thiosulfonate group of structure Vc;
    • iii) reacting the phosphorothiotriester comprising a thiosulfonate group of structure Vc with a nucleophile of structure VIc to provide the phosphorothiotriesters of structure IIIc;
    • wherein,
    • the H-phosphonate of structure Ic has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 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 between 1 and about 200; and

    • the bis(thiosulfonate) reagent of structure IVc has the following structure:







embedded image



wherein,

    • X is alkylene, alkenylene, arylene, or heteroarylene;
    • each R6 is independently alkyl, cycloalkyl, aryl, or heteroaryl;
    • the nucleophile of structure VIc has the following structure:
    • R7—SH, wherein R7 is selected from alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;
    • and phosphorothiotriesters of structure IIIc has the following structure:




embedded image




    • wherein,

    • W is independently selected from O, S, NH, or CH2;

    • R is R7—S—S—X—

    • R7 is alkyl, alkenyl, aryl, heterocyclo, aminoalkyl, or (heterocyclo)alkyl;

    • X is alkylene, alkenylene, arylene, or heteroarylene;

    • R3 is —OH, —SH, —NRdRd, —N3, halogen, hydrogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —P(O)(Re)2, —HP(O)(Re), —ORa or —SRc;

    • Y1 is O, NRd, S, or Se;

    • Ra is a blocking group;

    • Rc 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 O, NRd, or S;

    • each instance of R4 is independently hydrogen, —OH, —SH, —NRdRd, —N3, halogen, alkyl, alkenyl, alkynyl, alkyl-Y1—, alkenyl-Y1—, alkynyl-Y1—, aryl-Y1—, heteroaryl-Y1—, —ORb, or —SRc, and Rb is a blocking group;

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

    • R5 is hydrogen, a blocking group, a linking moiety connected to a solid support or a linking moiety connected to a nucleic acid;

    • n is between 1 and about 200; and

    • wherein the phosphorous linkages of the H-phosphonate of structure Ic, the phosphorothiotriester comprising a thiosulfonate group of structure Vc, and the phosphorothiotriesters of structure IIIc may optionally comprise non-stereorandom phosphorous linkages.





Another embodiment provides the process wherein the phosphorothiotriesters of structure IIIb comprise non-stereorandom phosphorous linkages and the H-phosphonate of structure Ic comprise non-stereorandom phosphorous linkages; and W is independently selected from O, NH, or CH2. Another embodiment provides the process wherein W is O.


Another embodiment provides the process wherein R6 is methyl.


Another embodiment provides the process wherein bis(thiosulfonate) reagent of structure IVc is selected from:




embedded image


Another embodiment provides the process wherein the nucleophile of structure VIc has the following structure:




embedded image


Another embodiment provides a process for the preparation of phosphorothiotriesters of structure IIIa, wherein the silylating reagent is selected from

  • 1,1,3,3-tetramethyl-1,3-diphenyldisilazane;
  • 1,3-dimethyl-1,1,3,3-tetraphenyldisilazane;
  • 1-(trimethylsilyl)imidazole;
  • N-trimethylsilyl-N-methyl trifluoroacetamide;
  • bis(dimethylamino)dimethylsilane;
  • bromotrimethylsilane;
  • chlorodimethyl(pentafluorophenyl)silane;
  • chlorotriethylsilane;
  • chlorotriisopropylsilane;
  • chlorotrimethylsilane;
  • dichlorodimethylsilane;
  • hexamethyldisilazane;
  • N,N′-bis(trimethylsilyl)urea;
  • N,N-bis(trimethylsilyl)methylamine;
  • N,N-dimethyltrimethylsilylamine;
  • N,O-bis(trimethylsilyl)acetamide;
  • N,O-bis(trimethylsilyl)carbamate;
  • N,O-bis(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-(trimethylsilyl)trifluoroacetamide;
  • N-methyl-N-trimethylsilylacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide;
  • N-methyl-N-trimethylsilylheptafluorobutyramide;
  • trimethyl silyltriflate;
  • triethylsilyltriflate;
  • triisopropylsilyltriflate; or
  • tert-butyldimethyl silyltriflate.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide, trimethyl silyltriflate, chlorotrimethylsilane, or 1-(trimethyl silyl)imidazole.


Another embodiment provides the process, wherein the silylating reagent is selected from N,O-bis(trimethylsilyl)trifluoroacetamide.


Another embodiment provides the process, wherein the H-phosphonate is covalently linked to a solid phase.


Modified Oligonucleotides


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. Described herein are methods for the synthesis of modified oligonucleotides or pronucleotides.


Reaction Conditions and Reagents Used in the Methods of the Invention.


Conditions


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


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.


Solid supports for solid-phase nucleic acid synthesis include the supports described in, e.g., U.S. Pat. Nos. 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 al. U.S. Pat. Nos. 5,047,524, 5,262,530; and Koster U.S. Pat. No. 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 poreglass (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.


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 Primer™ 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.


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


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.


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


Synthesis of the nucleic acids is performed in an aprotic organic solvent. In some embodiments, the solvent is acetonitrile, pyridine, or NMP. In some embodiments, the solvent is acetone, acetontrile, NMP, ethyl acetate, THF, dioxane, DMF, DMSO, DCM, chloroform, pyridine, 2,6-lutidine, HMPA, HMPT, DMA, glyme, diglyme, sulfone, methyl tert-butyl ether, or combinations thereof. In some embodiments, the solvent is a polar, aprotic organic solvent. In some embodiments, the solvent is anhydrous.


Acidification Conditions to Remove Blocking Groups.


Acidification to remove blocking groups is accomplished by a Brønsted acid or Lewis acid. In some embodiments, acidification is used to remove R1 blocking groups. Useful Brønsted 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.




embedded image


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.


In some embodiments, the acidifying comprises adding an amount of a Brønsted or Lewis acid effective to convert the condensed intermediate into the compound of Formula 4 without removing purine moieties from the condensed intermediate.


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 Brønsted 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.


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, R1 is deblocked prior to the step of acidifying the condensed intermediate. In some embodiments, R1 is deblocked by acidification, which comprises adding 1% trifluoroacetic acid in an organic solvent. In some embodiments, R1 is deblocked by acidification, which comprises adding 3% dichloroacetic acid in an organic solvent. In some embodiments, R1 is deblocked by acidification, which comprises adding 3% trichloroacetic acid in an organic solvent.


Removal of Blocking Moieties or Groups


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-fluorenylmethoxycarbonyl (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 al., 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).


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 R1. The blocking groups at R1 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 R1 position is commonly referred to as front end deprotection.


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.


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, R1 can denote ORa, 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.


Stereochemistry of Oligonucleoside Phosphorothioate Linkages


Oligonucleoside phosphorothioates have shown therapeutic potential (Stein et al., Science (1993), 261:1004-12; Agrawal et al., Antisence Res. and Dev. (1992), 2:261-66; Bayever et al., 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 al (Nucleic Acids Symposium Series No. 51 p. 119-120; doi:10.1093/nass/nrm060) 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


The nucleobase Ba utilized in the compounds and methods described herein 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, R N A, 2003, 9, 1034-1048, Limbach et al. 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 the compounds and methods described herein.


Compounds represented by the following general formulae are also contemplated as modified nucleobases:




embedded image


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.


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 A T et al, Acc. Chem. Res., 2007, 40, 141-150; Kool, E T, 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:




embedded image


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-Roj as, H and Kool, E T, Org. Lett., 2002, 4, 4377-4380. Shown below is an example of a porphyrin-derived ring which can be used as a base replacement:




embedded image


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




embedded image


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:




embedded image


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.


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.


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 U.S. Pat. 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.


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.


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.


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, O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, 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 C1-C10 alkyl or C2-C10 alkenyl and alkynyl. Examples of substituents include, and are not limited to, O(CH2)nOCH3, and O(CH2)NH2, 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., Helv. 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.


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, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; and 5,359,044. Some modified sugars that are contemplated include:




embedded image


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 al., J. Am. Chem. Soc., 2005, 127, 4174-4175 and Tsai C H et al., PNAS, 2007, 14598-14603:




embedded image




    • 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 G F et al., PNAS, 1987, 84, 4398-4402 and Heuberger B D and Switzer C, J. Am. Chem. Soc., 2008, 130, 412-413, and is shown below:







embedded image


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:




embedded image


Pentopyranosyl (4′ to 2′) sugars contemplated include:




embedded image


Pentopyranosyl (4′ to 3′) sugars contemplated include:




embedded image


Tetrofuranosyl (3′ to 2′) sugars contemplated include:




embedded image


Other modified sugars contemplated include:




embedded image


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




embedded image


embedded image


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 al, 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 al, Science (2000), 290:1347-1351; A. Eschenmoser et al, Helv. Chim. Acta (1992), 75:218; J. Hunziker et al, Helv. Chim. Acta (1993), 76:259; G. Otting et al, Helv. Chim. Acta (1993), 76:2701; K. Groebke et al, Helv. Chim. Acta (1998), 81:375; and A. Eschenmoser, Science (1999), 284:2118.


Blocking Groups


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.


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.


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(0)-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.


Typically blocking/protecting groups useful in the synthesis of the compounds described herein are, by way of example only:




embedded image


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.


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).


In some embodiments, thiol moieties are incorporated in the compounds described herein and are protected. In some embodiments, the protecting groups include, but are not limited to, pixyl, trityl, benzyl, p-methoxybenzyl (PMB), or tert-butyl (t-Bu).


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, N.Y., 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, N.Y., 1994, which are incorporated herein by reference for such disclosure.


The examples 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—Synthesis of Methanethiosulfonate Reagents



embedded image


embedded image


embedded image


Compound 2

A solution of (Z)-but-2-ene-1,4-diol (0.93 ml, 11.3 mmol) and triethylamine (3.3 ml, 24 mmol) in DCM (50 mL) was added in a dropwise fashion to a stirring ice cold solution of methanesulfonyl chloride (1.9 ml, 24 mmol) in DCM (50 mL). After stirring for 0.5h at r.t. the mixture was poured onto ice and extracted. The organic layer was collected, dried (MgSO4), filtered and reduced to 2.66 g, 96% of compound 2, which was judged by NMR to be sufficiently pure for direct use in the next step of the reaction.



1H NMR (399 MHz, CDCl3) δ 5.94 (ddd, J=5.4, 4.1, 1.3 Hz, 2H), 4.83 (dd, J=4.1, 1.3 Hz, 4H), 3.04 (s, 6H); 13C NMR 128.34, 64.38, 38.27; MS (ESI+ve): calc (M+NH4): 262.04, found: 262.05. Rf=0.3 (1:1 EtOAc/hexane).


Compound 3

A solution of sodium methanesulfonothioate (1.51 g, 11.3 mmol) in MeOH (20 ml) was treated with neat (Z)-but-2-ene-1,4-diyl dimethanesulfonate (1.25 g, 5.12 mmol) at r.t. After 5 min, precipitation was observed to occur. After 36 h, the mixture was partitioned between water and DCM. The organic layer was separated, dried (MgSO4), filtered and reduced to afford a colorless oil. Column chromatography (ISCO) gave the pure product as a pale colorless oil. Column chromatography gave pure compound 3 (0.89 g, 63%) as a colorless oil.



1H NMR (399 MHz, CDCl3) δ 5.84 (ddd, J=6.6, 5.1, 1.5 Hz, 2H), 3.92 (dd, J=5.1, 1.5 HZ, 4H), 3.33 (s, 6H); 13C NMR 128.1, 51.47, 33.13; MS (ESI+ve): calc (M+NH4): 294.00, found: 294.04. Rf=0.4 (1:1 EtOAc/hexane).


Compound 4

Under argon atmosphere, morpholine (10 g, 115 mmol) was added to ethylene sulfide (15 g, 250 mmol) in a round bottom flask. The reaction was stirred for 7 hrs and was directly loaded on to a silica gel column. The column was washed with DCM first and then 2% MeOH/DCM was used to obtain compound 4 (15.3 g, 91%) as colorless oil.



1H NMR (399 MHz, CDCl3) δ 3.67-3.59 (m, 4H), 2.63-2.52 (m, 2H), 2.51-2.45 (m, 2H), 2.44-2.34 (m, 4H); MS (ESI+ve): calc (M+H)+=148.07, found: 148.1.


Compound 5

A DCM solution (1 mL) of 2-morpholinoethanethiol (0.21 g, 1.44 mmol) was added dropwise via syringe to a stirring solution compound 3 (0.40 g, 1.44 mmol) in DCM (10 mL) at r.t. Immediately after addition, the TLC was checked, to reveal rapid formation of product and some quantity of dimer. After 0.5 h, the mixture was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 5 (0.29 g, 58%) as colorless oil.



1H NMR (399 MHz, CDCl3) δ 5.78 (m, 2H), 3.92 (d, J=7.3 Hz, 2H), 3.70 (t, J=4.7 Hz, 4H), 3.46 (d, J=5.5 Hz, 2H), 3.31 (s, 3H), 2.84 (dd, J=7.8, 6.7 Hz, 2H), 2.66 (dd, J=7.8, 6.7, 2H), 2.48 (t, J=4.6 Hz, 4H); 13C NMR 130.35, 126.27, 66.97, 58.20, 53.67, 51.52, 36.22, 35.16, 33.67; MS (ESI+ve): calc (M+H): 344.05, found: 344.06. Rf=0.3 (EtOAc).


Compound 5b

A DCM solution (1 mL) of compound 4b (395 mg, 1.085 mmol) was added dropwise via syringe to a stirring DCM (15 mL) solution compound 3 (300 mg, 1.085 mmol) at r.t. After 1h, the resulting solution was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 5b as a colorless oil (0.35 g, 58%). 1H NMR (399 MHz, CDCl3) δ 5.83-5.70 (m, 2H), 5.35-5.21 (dt, J=26.0, 9.3 Hz, 2H), 5.16-5.07 (m, 1H), 4.59-4.54 (d, J=9.5 Hz, 1H), 4.29-4.23 (m, 1H), 4.23-4.18 (m, 1H), 3.99-3.88 (dd, J=6.7, 1.2 Hz, 2H), 3.80-3.72 (ddd, J=10.1, 4.6, 2.6 Hz, 1H), 3.64-3.56 (m, 1H), 3.50-3.43 (m, 1H), 3.31 (s, 3H), 2.09 (s, 3H), 2.03 (s, 6H), 2.00 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 170.68, 170.30, 169.51, 169.30, 129.43, 127.14, 87.73, 76.49, 73.89, 69.16, 67.99, 61.99, 51.64, 35.89, 33.58, 20.95, 20.80, 20.74, 20.71; MS (ESI+ve): calc (M+NH4+): 578.07, found: 577.96. Rf=0.5 (1:1 EtOAc/hexane).


Compound 6

An ice cold solution of (Z)-but-2-ene-1,4-diol (0.93 ml, 11.3 mmol) and triethylamine (1.6 mL, 11.5 mmol) in DCM (50 ml) was treated dropwise via syringe with pivaloyl chloride (1.4 ml, 11.4 mmol) over 2 min. After 1 h, TLC showed good reaction.


The resulting mixture was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. This crude compound was found: by TLC (Rf=0.6, 1:1 EtOAc/hexane) to contain no starting diol and was used crude to prepare the mesylate. The crude material was taken up in DCM (50 ml) containing triethylamine (1.7 mL, 12 mmol) and cooled on an ice bath. Methanesulfonyl chloride (0.98 ml, 12.66 mmol) was added dropwise via syringe over 2 min. TLC immediately after addition indicated complete consumption of starting material. The resulting mixture was partioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave pure compound 6, 1.48 g, 52%, as a colorless oil.


1H NMR (399 MHz, CDCl3) δ 5.89-5.75 (m, 2H), 4.89-4.84 (d, J=5.7 Hz, 2H), 4.68-4.63 (d, J=5.9 Hz, 2H), 3.03 (s, 3H), 1.19 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 178.28, 130.61, 126.11, 65.08, 59.65, 38.84, 38.21, 27.25; MS (ESI+ve): calc (M+NH4): 268.12, found: 268.20; Rf=0.3 (20% EtOAc/hexane).


Compound 7

A MeOH (10 ml) solution of sodium methanesulfonothioate (0.63 g, 4.70 mmol) and (Z)-4-(methylsulfonyloxy)but-2-enyl pivalate (1.00 g, 4.00 mmol) was stirred at r.t. for 18 h with formation of a white precipitate (after 10 min). The resulting mixture was partitioned by addition of water and DCM. Upon extraction into DCM, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 7, 0.83 g, 78% as a colorless oil.



1H NMR (399 MHz, CDCl3) δ 5.82-5.73 (m, 2H), 4.73-4.66 (m, 2H), 3.95-3.87 (m, 2H), 3.32 (s, 3H), 1.19 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 178.35, 129.37, 127.32, 59.50, 51.44, 38.84, 33.61, 27.28; MS (ESI+ve): calc (M+NH4): 284.10, found: 284.19; Rf=0.4 (20% EtOAc/hexane).


Compound 9

Pivaloyl chloride (0.60 g, 5.0 mmol) was added in a dropwise fashion to a stirring solution of S-2-hydroxyethyl methanesulfonothioate (0.65 g, 4.16 mmol) in DCM (20 ml). After 2 h at r.t. the resulting mixture with white precipitate was partitioned with water. The organic layer was separated, dried (Ns2SO4), filtered and reduced to an oil. Column gave compound 9 as a colorless oil (0.45 g, 45%). 1H NMR (399 MHz, CDCl3) δ 4.39-4.34 (t, J=6.3 Hz, 2H), 3.44-3.39 (t, J=6.3 Hz, 2H), 3.36 (s, 3H), 1.20 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 62.10, 51.11, 38.96, 35.19, 27.24; MS (ESI+ve): calc (M+NH4): 158.08, found: 158.04. Rf=0.3 (20% EtOAc/hexane).


Compound 11

Pivaloyl chloride (4.96 ml, 40.3 mmol) was added dropwise via syringe to an ice cold DCM solution (50 mL) of 2-(hydroxymethyl)phenol (5 g, 40.3 mmol) and triethylamine (5.61 ml, 40.3 mmol). An ice-cold solution of the crude pivalate ester was treated with triethylamine (6.74 ml, 48.4 mmol) and 50 mL DCM. Methanesulfonyl chloride (3.43 ml, 44.3 mmol) was then added slowly (5 min) via syringe and the resulting mixture was warmed to r.t. The mixture was poured onto ice and the organic layer was separated then washed with sat NaHCO3 (aq), dried (MgSO4), filtered and reduced to 10.5 g crude pale yellow oil.


Column (ISCO) gave pure 11 5.45 g, 47%.



1H NMR (399 MHz, CDCl3) δ 7.53-7.46 (dd, 7.7, 1.8 Hz, 1H), 7.46-7.40 (dt, 7.7, 1.8 Hz, 1H), 7.32-7.24 (t, 7.7 Hz, 1H), 7.13-7.06 (d, 7.7 Hz, 1H), 5.21 (s, 2H), 2.79 (s, 3H), 1.40 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 177.05, 150.06, 131.18, 131.07, 126.35, 125.94, 123.21, 66.88, 39.48, 38.82, 27.30, 27.26. MS (ESI+ve): calc (M+NH4): 304.12, found: 303.99. Rf=0.4 (20% EtOAc/hexane).


Compound 12

A MeOH (20 mL) solution of sodium methanesulfonothioate (0.825 g, 6.15 mmol) was treated with 2-((methylsulfonyloxy)methyl)phenyl pivalate (1.76 g, 6.15 mmol) at r.t. and left to stir for 18 h. The mixture was partitioned between water and DCM. The organic layer was separated, dried (MgSO4), filtered and reduced to afford a colorless oil. Column chromatography gave pure compound 12 as a pale colorless oil, 0.754 g, 41%.



1H NMR (399 MHz, CDCl3) δ 7.48-7.44 (dd, J 7.7, 1.7 Hz, 1H), 7.39-7.34 (td, J 7.8, 1.7 Hz, 1H), 7.25-7.20 (td, J 7.6, 1.2 Hz, 1H), 7.10-7.06 (dd, J 8.2, 1.2 Hz, 1H), 4.29 (s, 2H), 2.90 (s, 3H), 1.39 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 176.69, 149.59, 131.17, 129.85, 127.41, 126.18, 123.40, 51.43, 39.47, 36.01, 27.30; MS (ESI+ve): calc (M+NH4): 320.10, found: 320.09. Rf=0.4 (20% EtOAc/hexane).


Compound 14

Chloromethyl pivalate (0.478 ml, 3.32 mmol) was added to a stirring mixture of sodium iodide (0.050 g, 0.33 mmol) and sodium methanesulfonothioate (0.445 g, 3.32 mmol) in acetone (7 ml) at r.t. After 24 h, TLC showed good conversion to product. The solvent was removed, and the residue was partitioned between water and DCM. The organic layer was separated and dried (MgSO4), filtered and reduced to afford a colorless oil. Column chromatography gave pure 14 as a slightly pink solid, 0.41 g, 55%.



1H NMR (399 MHz, CDCl3) δ 5.67 (s, 2H), 3.39 (s, 3H), 1.24 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 177.35, 67.84, 52.20, 38.93, 27.05. Rf=0.5 (20% EtOAc/hexane).


Compound 16

Prepared from 15 and NaMTS as described previously: U.S. Pat. No. 3,484,473 1H NMR (399 MHz, CDCl3) δ 4.86 (s, 2H), 3.45 (s, 6H); 13C NMR (100 MHz, CDCl3) δ 52.15, 41.50.


Compound 18

Prepared from 17 and NaMTS as described previously: Chem. Pharm. Bull. Vol. 12(11) p. 1271, 1964.



1H NMR (399 MHz, CDCl3) δ 3.55 (s, 4H), 3.40 (s, 6H); 13C NMR (100 MHz, CDCl3) δ 50.67, 35.96.


Compound 19

A DCM solution (1 mL) of 2-morpholinoethanethiol (0.17 g, 1.2 mmol) was added dropwise via syringe to a stirring solution of compound 18 (300 mg, 1.2 mmol) in DCM (10 mL) at r.t. Immediately after addition, the TLC was checked, to reveal rapid formation of product and some dimer. After 0.5 h, the mixture was partitioned by addition of NaHCO3. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave pure 19 (0.20 g, 53%) as a colorless oil. 1H NMR (399 MHz, CDCl3) δ 3.73-3.67 (t, J=4.7 Hz, 4H), 3.51-3.46 (m, 2H), 3.35 (s, 3H), 3.07-3.01 (m, 2H), 2.88-2.83 (m, 2H), 2.69-2.63 (m, 2H), 2.52-2.43 (t, J=4.6 Hz, 4H); 13C NMR (100 MHz, CDCl3) δ 66.96, 57.91, 53.58, 50.79, 37.66, 36.10, 35.52; MS (ESI+ve): calc (M+H): 318.03, found: 318.04. Rf=0.3 (EtOAc).


Compound 21

Compound 20 is converted to compound 21 by a procedure analogous to that described for compound 11.


Compound 22

Compound 21 is converted to compound 22 by a procedure analogous to that described for compound 12.


Compound 23

Compound 23 is prepared according to a literature method (Journal of Medicinal Chemistry, 50(23), 5568-5570; 2007.)


Compound 24

An ice-cold pyridine solution (10 mL) of compound 23 (1 mmol) is treated successively, in a dropwise fashion with acetyl chloride (1 mmol), then after 5 min with MsCl (1.1 mmol). The solution is warmed to room temperature then the solvent is removed. The residue is dissolved in EtOAc, washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 24.


Compound 25

Compound 24 is converted to compound 25 by a procedure analogous to that described for compound 12.


Compound 27

Compound 26 is converted to compound 27 by a procedure analogous to that described for compound 14.


Compound 29

Compound 28 is converted to compound 29 by a procedure analogous to that described for compound 14.


Compound 30

Compound 30 is prepared according to a literature method (Tetrahedron, 42(2), 601-7; 1986.)


Compound 31

Compound 31 is prepared from compound 30 according to a patent procedure (US 20090181444)


Compound 33

Compound 33 is prepared from compound 32 according to a patent procedure (US 20090181444)


Compound 36

An ice-cold DCM (20 mL) solution of compound 34 (1 mmol) is treated with NEt3 (1 mmol) followed by the dropwise addition of TMS-Cl (1.1 mmol). After 1 h, the solution is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude TMS protected material is redissolved in THF (10 mL), whereon PPh3 (1.2 mmol), compound 35 (1.2 mmol), then DEAD (1.2 mmol, dropwise) are added in succession. After stirring at r.t. for 18 h, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 36.


Compound 37

A THF (10 mL) solution of compound 36 (0.5 mmol) is treated with TBAF (1 mmol of a 1M solution in THF), with monitoring by TLC. On completion of TMS cleavage, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude alcohol is redissolved in pyridine (5 mL), and TsCl (0.55 mmol) is added. After 18 h at r.t., the solvent is removed, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 37.


Compound 38

Compound 37 is converted to compound 38 by a procedure analogous to that described for compound 12.


Compound 40

An ice-cold DCM (20 mL) solution of compound 39 (1 mmol) is treated with NEt3 (1 mmol) followed by the dropwise addition of TMS-Cl (1.1 mmol). After 1 h, the solution is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude TMS protected material is redissolved in THF (10 mL), whereon PPh3 (1.2 mmol), potassium p-toluenethiosulfonate (KTTS, 1.2 mmol), anhydrous ZnCl2 (1 mmol) then DEAD (1.2 mmol, dropwise) are added in succession. After stirring at r.t. for 18 h, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 40.


Compound 41

A THF (10 mL) solution of compound 40 (0.5 mmol) is treated with TBAF (1 mmol of a 1M solution in THF), with monitoring by TLC. On completion of TMS cleavage, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. The crude alcohol is redissolved in THF (10 mL), whereon PPh3 (1.2 mmol), compound 35 (1.2 mmol), then DEAD (1.2 mmol, dropwise) are added in succession. After stirring at r.t. for 18 h, the solvent is removed under vacuum, the residue is redissolved in DCM, the solution of which is washed with water, dried (MgSO4), filtered and reduced in vacuo. Purification by column chromatography affords pure compound 40.


Compound 42

Compound 41 is converted to compound 42 by a procedure analogous to that described for compound 14.


Example 2—Thioalkylation of H-Phosphonates to Provide Phosphorothiotriesters in Solution Phase



embedded image


embedded image


Compound 100

The synthetic procedure for Di-DMTr H-phosphonate TT dimer (100) has been previously described described (Froehler, Brian C.; Ng, Peter G.; Matteucci, Mark D., Nucleic Acids Research (1986), 14(13), 5399-5407; Garegg, Per J.; Lindh, Ingvar; Regberg, Tor; Stawinski, Jacek; Stroemberg, Roger; Henrichson, Christina Tetrahedron Letters (1986), 27(34), 4051-4054).


Compound 101

Compound 100, mixture of diastereomers (200 mg, 0.176 mmol) was dissolved in ACN (6 mL) then trimethylsilyl 2,2,2-trifluoro-N-(trimethylsilyl)acetimidate (227 mg, 0.882 mmol) was added. A solution of (Z)—S-4-((2-morpholinoethyl)disulfanyl)but-2-enyl methanesulfonothioate (121 mg, 0.353 mmol) in ACN (2 mL) was then added, over the course of 1 h in 3 approximately equal portions, with monitoring by TLC and HPLC/MS. After 3 h, the resulting solution was partitioned by addition of water. Upon extraction, the organic layer was separated then dried (MgSO4), filtered and reduced in vacuo. Column chromatography gave compound 101 as a white foam, 225 mg, 91%.



1H NMR (399 MHz, CDCl3) δ 9.72 (d, br, 1H), 9.27, (d, br, 1H), 7.53 (dd, J 25.0, 1 Hz, 1H), 7.42, (t, J 7.0 Hz, 2H), 7.37-7.16 (m, 17H), 6.83 (m, 8H), 6.43-6.28 (m, 2H), 5.63-5.42 (m, 2H), 5.21 (q, J 7.1 Hz, 1H), 4.27 (m, br, 1H), 3.94 (m, br, 2H), 3.77 (m, 12H), 3.74-3.60 (m, 6H), 3.51-3.22 (m, 5H), 2.82-2.76 (m, 2H), 2.68-2.60 (m, 2H), 2.59-2.46 (m, 5H), 2.44-2.33 (m, 2H), 2.03-1.88 (m, 1H), 1.84 (m, 3H), 1.75-1.66 (m, 1H), 1.48-1.32 (dd, J 11.8, 1.2 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 164.10, 164.07, 164.00, 163.94, 159.14, 159.10, 150.80, 150.78, 150.75, 150.63, 145.09, 144.30, 144.27, 136.31, 136.27, 136.22, 136.18, 135.95, 135.82, 135.43, 135.35, 135.33, 135.24, 135.22, 130.52, 130.43, 130.40, 129.49, 129.30, 128.54, 128.43, 128.39, 127.64, 127.57, 113.78, 113.76, 113.73, 113.67, 112.05, 111.56, 87.77, 87.66, 87.58, 85.77, 85.59, 84.63, 84.51, 74.42, 74.33, 67.02, 66.95, 63.63, 63.49, 58.27, 58.23, 55.60, 55.58, 53.69, 53.62, 39.48, 39.26, 39.18, 35.88, 35.61, 35.43, 35.36, 28.18, 12.83, 12.79, 12.02, 11.95.; 31P NMR (162 MHz, CDCl3) δ 29.25, 29.12; MS (ESI+ve): calc (M+H): 1398.46, found: 1398.64. Rf=0.4 (5% MeOH/DCM).


Compound 201

Compound 101 (0.150 g, 0.107 mmol) was stirred with 3% TCA/DCM (10 mL) over 10 min. TLC and HPLC/MS showed that the reaction was complete. 10 mL of MeOH was added and stirring continued for 2 min. Solvents were evaporated and the residue was purified by column chromatography to give compound 201 (85 mg, 100%) as a white solid.



1H NMR (399 MHz, CD3OD) δ 7.78 (dd, J=7.2, 1.3 Hz, 1H), 7.53 (d, J=1.3 Hz, 1H), 6.33-6.27 (m, 2H), 5.83-5.70 (m, 2H), 5.25-5.19 (m, 1H), 4.47-4.30 (m, 3H), 4.27-4.22 (m, 1H), 4.11-4.05 (m, 1H), 3.89-3.82 (t, J=4.8 Hz, 4H), 3.85 (m, 2H), 3.76-3.70 (ddd, J=15.5, 7.2, 1.7 Hz, 2H), 3.52 (dd, J=7.3, 3.7 Hz, 2H), 3.28-3.19 (br, 2H), 3.16-3.05 (br, 4H), 3.05-2.98 (ddd, J=9.8, 5.5, 2.0 Hz, 2H), 2.62-2.52 (tdd, J=11.5, 5.7, 1.9 Hz, 1H), 2.47-2.36 (m, 1H), 2.33-2.28 (m, 2H), 1.92-1.87 (m, 6H); 31P NMR (162 MHz, CD3OD) δ 30.22, 30.19; MS (ESI+ve): calc (M+H): 794.20, found: 794.18. Rf=0.3 (10% MeOH/DCM).


Compound 102

Compound 100 (400 mg, 0.352 mmol) was converted to compound 102 by a procedure analogous to that described for compound 101 (417 mg, 90%).



1H NMR (399 MHz, CDCl3) δ 9.17 (d, J=6.0 Hz, 1H), 9.13-9.00 (d, J=25.7 Hz, 1H), 7.58-7.49 (dd, J=26.3, 1.5 Hz, 1H), 7.45-7.40 (ddd, J=8.0, 5.2, 1.3 Hz, 2H), 7.40-7.18 (m, 17H), 6.87-6.81 (m, 8H), 6.44-6.30 (m, 2H), 5.65-5.53 (m, 1H), 5.53-5.44 (m, 1H), 5.26-5.16 (quintet, J=6.4 Hz, 1H), 4.61-4.54 (m, 2H), 4.30-4.24 (m, 1H), 4.19-4.13 (m, 1H), 3.97-3.88 (m, 2H), 3.80-3.72 (m, 12H), 3.69-3.57 (m, 1H), 3.54-3.30 (m, 5H), 2.61-2.49 (dt, J=14.4, 5.4 Hz, 1H), 2.44-2.32 (m, 1H), 2.02-1.91 (dt, J=12.5, 5.4 Hz, 1H), 1.85-1.80 (dd, J=5.0, 1.3 Hz, 3H), 1.76-1.63 (m, 1H), 1.43-1.36 (dd, J=10.2, 1.2 Hz, 3H), 1.19-1.14 (d, J=2.0 Hz, 8H); 13C NMR (100 MHz, CDCl3) δ 178.22, 178.17, 163.82, 163.80, 163.75, 158.92, 158.88, 150.52, 150.43, 144.90, 144.88, 144.10, 144.05, 136.11, 136.08, 136.05, 136.01, 135.59, 135.28, 135.16, 135.03, 135.01, 130.30, 130.23, 130.19, 130.16, 128.69, 128.64, 128.59, 128.39, 128.34, 128.23, 128.21, 128.17, 127.42, 127.34, 113.54, 113.45, 111.85, 111.82, 111.41, 111.36, 87.59, 87.43, 87.37, 85.47, 85.33, 84.43, 84.29, 84.08, 84.00, 83.92, 74.24, 67.36, 63.38, 63.26, 59.42, 55.37, 39.22, 38.77, 27.94, 27.24, 12.57, 11.80, 11.74; 31P NMR (162 MHz, CDCl3) δ 29.23, 28.97; MS (ESI+ve): calc (M+H): 1338.51, found: 1338.84. Rf=0.5 (5% MeOH/DCM).


Compound 202

Compound 102 (200 mg, 0.151 mmol) was converted to compound 202 by a procedure analogous to that described for compound 101 (105 mg, 97%).



1H NMR (399 MHz, CD3OD) δ 7.81-7.75 (dd, J=8.2, 1.3 Hz, 1H), 7.57-7.51 (dd, J=8.2, 1.3 Hz, 1H), 6.33-6.23 (m, 2H), 5.85-5.75 (m, 1H), 5.75-5.66 (m, 1H), 5.26-5.19 (m, 1H), 4.72-4.66 (m, 2H), 4.47-4.30 (m, 3H), 4.27-4.20 (m, 1H), 4.11-4.04 (m, 1H), 3.83-3.76 (m, 2H), 3.74-3.64 (m, 2H), 2.62-2.51 (m, 1H), 2.45-2.35 (td, J=8.7, 6.5 Hz, 1H), 2.32-2.24 (m, 2H), 1.93-1.82 (m, 6H), 1.20-1.15 (d, J=2.1 Hz, 9H); 13C NMR (126 MHz, CD3OD) δ 179.65, 166.28, 152.30, 152.28, 152.22, 137.90, 137.81, 137.79, 130.07, 130.04, 129.26, 129.24, 111.93, 111.88, 111.87, 87.26, 87.22, 86.96, 86.90, 86.76, 86.54, 86.12, 86.07, 85.98, 85.92, 85.88, 85.82, 80.54, 80.49, 80.46, 80.41, 71.84, 71.67, 68.71, 68.66, 68.45, 68.40, 62.58, 62.50, 60.72, 40.51, 40.44, 39.70, 39.52, 39.48, 28.67, 28.64, 28.61, 27.53, 12.64, 12.48; 31P NMR (162 MHz, CDCl3) δ 29.23, 28.97; MS (ESI+ve): calc (M+H): 717.22, found: 717.23. Rf=0.5 (10% MeOH/DCM).


Compound 103

Compound 100 (400 mg, 0.352 mmol) was converted to compound 103 by a procedure analogous to that described for compound 101 (379 mg, 83%).



1H NMR (399 MHz, CDCl3) δ 9.48 (s, 1H), 9.41-9.29 (m, 1H), 7.60-7.48 (dd, J=9.0, 1.0 Hz, 1H), 7.46-7.40 (dt, J=6.9, 1.2 Hz, 2H), 7.39-7.17 (m, 17H), 6.89-6.79 (m, 8H), 6.44-6.31 (m, 2H), 5.27-5.20 (t, J=6.5 Hz, 1H), 4.30-4.24 (t, J=6.1 Hz, 1H), 4.19-4.15 (m, 2H), 4.13-4.07 (t, J=7.1 Hz, 1H), 3.99-3.90 (m, 2H), 3.79-3.74 (m, 12H), 3.70-3.58 (m, 1H), 3.51-3.43 (td, J=8.8, 7.2, 2.3 Hz, 1H), 3.40-3.32 (m, 1H), 3.02-2.85 (m, 2H), 2.61-2.49 (dt, J=18.5, 7.0 Hz, 1H), 2.47-2.33 (m, 1H), 1.98-1.90 (dt, J=10.2, 5.0 Hz, 1H), 1.85-1.81 (m, 3H), 1.74-1.62 (td, J=14.2, 7.1 Hz, 1H), 1.42-1.36 (m, 3H), 1.19-1.13 (d, J=4.9 Hz, 9H); 31P NMR (162 MHz, CDCl3) δ 29.36, 29.18; 13C NMR (126 MHz, CDCl3) δ 177.97, 177.89, 163.94, 163.91, 163.90, 163.86, 158.91, 158.87, 150.63, 150.54, 150.53, 150.50, 144.88, 144.85, 144.10, 144.04, 136.09, 135.99, 135.52, 135.50, 135.24, 135.16, 135.12, 135.04, 135.00, 130.31, 130.29, 130.20, 130.16, 130.13, 128.34, 128.20, 128.18, 128.14, 127.39, 127.31, 124.89, 113.55, 113.52, 113.43, 111.84, 111.38, 87.58, 87.42, 87.36, 85.30, 84.98, 84.95, 84.40, 84.33, 84.27, 83.98, 83.91, 83.84, 79.31, 79.27, 78.88, 78.84, 74.16, 74.08, 67.56, 67.50, 67.46, 67.41, 63.33, 63.24, 62.79, 62.75, 55.34, 39.21, 39.16, 39.04, 39.00, 38.85, 38.82, 29.95, 29.92, 29.66, 29.63, 27.17, 12.53, 11.80, 11.72; MS (ESI+ve): calc (M+H): 1312.69, found: 1312.49. Rf=0.4 (5% MeOH/DCM).


Compound 203

Compound 103 (200 mg, 0.154 mmol) was converted to compound 203 by a procedure analogous to that described for compound 201 (103 mg, 98%).



1H NMR (399 MHz, CD3OD) δ 7.80-7.76 (dd, J=8.2, 1.2 Hz, 1H), 7.55-7.51 (dd, 7.1, 1.2 Hz, 1H), 6.32-6.24 (m, 2H), 5.26-5.19 (m, 1H), 4.46-4.20 (m, 6H), 4.10-4.05 (m, 1H), 3.82-3.78 (dd, J=6.5, 3.2 Hz, 2H), 3.22-3.14 (ddd, J=16.6, 7.0, 5.8 Hz, 2H), 2.61-2.51 (tdd, J=13.0, 5.9, 2.1 Hz, 1H), 2.46-2.37 (ddd, J=14.3, 8.3, 6.0 Hz, 1H), 2.31-2.26 (t, J=5.8 Hz, 2H), 1.91-1.86 (dt, J=11.0, 1.2 Hz, 6H), 1.21-1.17 (m, 9H); 31P NMR (162 MHz, CD3OD) δ 30.15; 13C NMR (100 MHz, CD3OD) δ 179.45, 179.42, 166.29, 152.31, 152.29, 152.23, 137.82, 137.80, 137.78, 111.91, 111.88, 87.21, 87.17, 86.94, 86.87, 86.63, 86.52, 86.11, 86.06, 85.92, 85.84, 85.77, 80.67, 80.60, 80.49, 80.43, 71.79, 71.64, 68.80, 68.74, 68.58, 68.52, 64.11, 64.07, 64.02, 62.54, 62.44, 40.48, 40.43, 39.81, 39.71, 39.68, 39.52, 39.47, 30.74, 30.72, 30.68, 27.52, 12.65, 12.50; MS (ESI+ve): calc (M+H): 691.21, found: 691.09. Rf=0.5 (10% MeOH/DCM).


Compound 104

Compound 100 (400 mg, 0.352 mmol) was converted to compound 104 by a procedure analogous to that described for compound 101 (451 mg, 94%).



1H NMR (399 MHz, CDCl3) δ 9.17-9.01 (m, 2H), 7.51-7.46 (dd, J 7.8, 1.5 Hz, 1H), 7.45-7.38 (m, 2H), 7.37-7.09 (m, 19H), 7.01-6.90 (m, 2H), 6.87-6.78 (m, 8H), 6.39-6.27 (m, 2H), 5.15-5.01 (m, 1H), 4.20-4.13 (m, 1H), 3.96-3.90 (m, 1H), 3.90-3.83 (m, 2H), 3.80-3.68 (m, 14H), 3.52-3.20 (m, 3H), 2.45-2.16 (m, 2H), 2.01-1.88 (ddd, J 23.3, 13.6, 5.6 Hz, 1H), 1.85-1.79 (dd, J 9.3, 1.2 Hz, 3H), 1.69-1.53 (m, 1H), 1.40-1.31 (m, 12H); 13C NMR (100 MHz, CDCl3) δ 176.46, 176.37, 163.84, 163.78, 158.90, 158.87, 150.52, 150.50, 150.43, 149.38, 149.28, 144.95, 144.88, 144.16, 144.10, 136.13, 136.11, 136.09, 136.03, 135.57, 135.49, 135.37, 135.26, 135.21, 135.08, 135.04, 130.83, 130.74, 130.29, 130.21, 130.16, 129.51, 129.49, 129.40, 129.36, 129.35, 129.31, 128.38, 128.35, 128.27, 128.23, 128.19, 128.14, 127.39, 127.33, 126.05, 125.94, 122.94, 122.86, 113.53, 113.42, 111.77, 111.73, 111.39, 111.28, 87.55, 87.52, 87.37, 87.32, 85.33, 84.95, 84.90, 84.29, 84.20, 84.00, 83.92, 83.87, 83.79, 79.05, 79.00, 74.29, 74.24, 67.31, 67.24, 67.17, 67.11, 63.37, 55.37, 55.35, 39.37, 39.32, 39.15, 39.10, 38.64, 30.51, 30.41, 30.36, 27.28, 27.24, 12.59, 12.51, 11.75, 11.67; 31P NMR (162 MHz, CDCl3) δ 29.12, 28.49; MS (ESI+ve): calc (M+NH4): 1374.51, found: 1374.74. Rf=0.4 (5% MeOH/DCM).


Compound 204

Compound 104 (200 mg, 0.147 mmol) was converted to compound 204 by a procedure analogous to that described for compound 201 (98 mg, 88%).



1H NMR (399 MHz, CD3OD) δ 7.77-7.73 (m, 1H), 7.51-7.43 (m, 2H), 7.38-7.31 (m, 1H), 7.25-7.19 (ddd, J=9.2, 5.4, 1.6 Hz, 1H), 7.08-7.02 (ddd, J=8.0, 3.8, 1.3 Hz, 1H), 6.28-6.17 (m, 2H), 5.10-5.01 (m, 1H), 4.30-4.16 (m, 3H), 4.11-4.03 (m, 3H), 4.03-3.97 (d, J=5.3 Hz, 2H), 3.74-3.63 (m, 2H), 2.48-2.11 (m, 5H), 1.90-1.82 (m, 6H), 1.43-1.36 (d, J=3.4 Hz, 9H); 13C NMR (100 MHz, CD3OD) δ 178.05, 166.26, 152.25, 152.19, 150.78, 137.80, 137.76, 132.13, 132.09, 130.61, 130.56, 127.24, 124.10, 111.92, 111.84, 111.79, 87.14, 87.09, 86.80, 86.71, 86.50, 85.98, 85.95, 85.92, 85.87, 85.83, 85.75, 80.55, 80.48, 80.32, 80.27, 71.97, 71.73, 68.67, 68.61, 68.35, 68.29, 62.51, 62.42, 40.41, 40.36, 40.32, 39.66, 39.64, 39.35, 39.29, 31.08, 31.04, 27.61, 12.68, 12.65, 12.49; 31P NMR (162 MHz, CD3OD) δ 29.54, 29.29; MS (ESI+ve): calc (M+H): 753.22, found: 753.12. Rf=0.5 (10% MeOH/DCM).


Compound 105

Compound 100 (200 mg, 0.176 mmol) was converted to compound 105 by using compound 14 in a procedure analogous to that described for compound 101 (158 mg, 70%).



1H NMR (400 MHz, CDCl3) δ 7.46-7.39 (m, 2H) 7.38-7.16 (m, 18H), 6.90-6.77 (m, 8H), 6.43-6.27 (m, 1H), 5.39-5.18 (m, 2H), 4.31-4.23 (dd, J=12.0, 6.2 Hz, 1H), 4.20-4.12 (m, 1H), 3.98-3.86 (m, 1H), 3.82-3.70 (m, 12H), 3.69-3.52 (m, 1H), 3.50-3.43 (td, J=9.9, 8.9, 2.7 Hz, 1H), 3.41-3.29 (ddd, J=17.2, 10.8, 2.5 Hz, 1H), 2.59-2.49 (m, 1H), 2.44-2.30 (m, 1H), 2.03-1.93 (m, 1H), 1.86-1.79 (d, J=2.9 Hz, 3H), 1.75-1.67 (m, 4H), 1.43-1.36 (d, 3H), 1.16-1.08 (d, J=9.3 Hz, 9H); 31P NMR (162 MHz, CDCl3) δ 28.14, 27.81 (two diastereomers). MS (ESI+ve): calc (M+H): 1281.4, found: 1281.1 (M+H)+ and 1298.6 (M+NH4)+


Compound 205

Compound 105 (137 mg, 0.107 mmol) was converted to compound 205 by a procedure analogous to that described for compound 201 (66 mg, 91%). 1H NMR (399 MHz, CD3OD) δ 7.83-7.76 (m, 1H), 7.56-7.50 (m, 1H), 6.34-6.22 (m, 2H), 5.51-5.43 (m, H), 5.28-5.20 (qt, J=7.8, 1.8 Hz, 1H), 4.47-4.31 (m, 3H), 4.29-4.21 (m, 1H), 4.10-4.05 (m, 1H), 3.87-3.73 (dd, J=7.6, 3.1 Hz, 2H), 2.62-2.50 (tdd, J=16.9, 5.7, 1.9 Hz, 1H), 2.45-2.36 (m, 1H), 2.32-2.25 (ddd, J=6.9, 5.4, 1.5 Hz, 3H), 1.92-1.84 (m, 6H), 1.22-1.18 (d, J=5.3 Hz, 9H); 31P NMR (162 MHz, CD3OD) δ 28.71, 28.42 (two diastereomers). MS (ESI+ve): calc (M+H): 677.2, found: 677.2 (M+H)+, 694.2 (M+NH4)+


Compound 106

Compound 100 (405 mg, 0.357 mmol) was converted to compound 106 by using compound 19 and following a procedure analogous to that described for compound 101 (0.35 g, 71%). 1H NMR (399 MHz, CDCl3) δ 9.97-9.42 (m, 2H), 7.58-7.47 (m, 1H), 7.46-7.39 (m, 2H), 7.39-7.13 (m, 17H), 6.87-6.78 (m, 8H), 6.44-6.29 (dtd, J=20.4, 9.2, 4.7 Hz, 2H), 5.27-5.16 (dt, J=14.7, 7.3 Hz, 1H), 4.30-4.22 (m, 1H), 4.22-4.12 (m, 1H), 4.02-3.90 (q, J=3.8, 3.4 Hz, 2H), 3.80-3.73 (m, 12H), 3.72-3.65 (m, 5H), 3.51-3.43 (m, 1H), 3.40-3.31 (m, 1H), 3.14-2.93 (m, 2H), 2.85-2.72 (m, 4H), 2.67-2.59 (m, 2H), 2.57-2.34 (m, 6H), 1.97-1.87 (td, J=13.7, 13.1, 5.7 Hz, 1H), 1.84 (s, 3H), 1.73-1.61 (td, J=14.1, 6.8 Hz, 1H), 1.42-1.37 (d, J=6.7 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 163.97, 163.94, 163.91, 158.88, 158.84, 150.64, 150.60, 150.52, 144.86, 144.83, 144.09, 144.04, 136.06, 136.04, 135.95, 135.93, 135.54, 135.19, 135.09, 135.03, 134.99, 130.28, 130.17, 130.13, 128.29, 128.17, 128.14, 127.38, 127.31, 113.51, 113.42, 111.82, 111.79, 111.44, 111.38, 87.53, 87.38, 87.33, 85.29, 85.26, 84.89, 84.85, 84.41, 84.36, 84.29, 84.25, 83.88, 83.85, 83.80, 83.76, 79.28, 79.23, 78.72, 78.67, 74.04, 67.53, 67.46, 67.37, 67.29, 66.77, 63.33, 63.21, 57.84, 55.34, 53.41, 53.34, 39.23, 39.09, 39.01, 38.92, 38.55, 38.51, 38.46, 38.42, 35.64, 35.59, 30.35, 30.30, 30.26, 12.60, 11.79, 11.74; 31P NMR (162 MHz, CDCl3) δ 29.30, 29.14; MS (ESI+ve): calc (M+H): 1372.44, found: 1372.79. Rf=0.4 (5% MeOH/DCM).


Compound 206

Compound 106 (200 mg, 0.146 mmol) was converted to compound 206 by a procedure analogous to that described for compound 201 (110 mg, 98%). 1H NMR (399 MHz, CD3OD) δ 7.83-7.75 (dd, J=7.6, 1.4 Hz, 1H), 7.56-7.48 (d, J=1.6 Hz, 1H), 6.35-6.23 (m, 2H), 5.27-5.20 (m, 1H), 4.48-4.31 (m, 3H), 4.28-4.21 (dd, J=9.7, 2.1 Hz, 1H), 4.11-4.04 (t, J=4.0 Hz, 1H), 3.97-3.84 (br, 4H), 3.83-3.77 (dd, J=6.0, 3.2 Hz, 2H), 3.43-3.36 (m, 2H), 3.29-3.18 (m, 6H), 3.11-3.00 (m, 4H), 2.62-2.51 (tdd, J=11.7, 5.7, 1.7 Hz, 1H), 2.47-2.38 (ddd, J=14.3, 8.4, 6.0 Hz, 1H), 2.38-2.25 (q, J=5.3, 4.8 Hz, 2H), 1.91 (s, 3H), 1.88 (s, 3H); 31P NMR (162 MHz, CD3OD) δ 30.19, 30.12; 13C NMR (100 MHz, CD3OD) δ 166.28, 166.24, 166.23, 152.32, 152.27, 152.24, 138.05, 138.00, 137.77, 137.75, 112.08, 112.03, 111.97, 111.94, 87.28, 87.24, 87.01, 86.96, 86.62, 86.51, 86.10, 86.06, 85.76, 85.68, 71.73, 71.51, 68.91, 68.58, 68.51, 65.44, 62.60, 62.50, 57.50, 53.50, 40.25, 40.16, 39.64, 39.57, 39.20, 39.16, 39.06, 32.56, 32.55, 31.04, 31.00, 12.73, 12.69, 12.52; MS (ESI+ve): calc (M+H): 768.18, found: 768.14. Rf=0.3 (10% MeOH/DCM).


Compound 107

Using compound 22 in place of compound 5, compound 100 is converted to compound 107 by a procedure analogous to that described for compound 101.


Compound 207

Compound 107 is converted to compound 207 by a procedure analogous to that described for compound 201.


Compound 108

Using compound 25 in place of compound 5, compound 100 is converted to compound 108 by a procedure analogous to that described for compound 101.


Compound 208

Compound 108 is converted to compound 208 by a procedure analogous to that described for compound 201.


Compound 109

Using compound 27 in place of compound 5, compound 100 is converted to compound 109 by a procedure analogous to that described for compound 101.


Compound 209

Compound 109 is converted to compound 209 by a procedure analogous to that described for compound 201.


Compound 110

Using compound 29 in place of compound 5, compound 100 is converted to compound 110 by a procedure analogous to that described for compound 101.


Compound 210

Compound 110 is converted to compound 210 by a procedure analogous to that described for compound 201.


Compound 111

Using compound 31 in place of compound 5, compound 100 is converted to compound 111 by a procedure analogous to that described for compound 101.


Compound 211

Compound 111 is converted to compound 211 by a procedure analogous to that described for compound 201.


Compound 112

Using compound 33 in place of compound 5, compound 100 is converted to compound 112 by a procedure analogous to that described for compound 101.


Compound 212

Compound 112 is converted to compound 212 by a procedure analogous to that described for compound 201.


Compound 113

Using compound 38 in place of compound 5, compound 100 is converted to compound 113 by a procedure analogous to that described for compound 101.


Compound 213

Compound 113 is converted to compound 213 by a procedure analogous to that described for compound 201.


Compound 114

Using compound 41 in place of compound 5, compound 100 is converted to compound 114 by a procedure analogous to that described for compound 101.


Compound 214

Compound 114 is converted to compound 214 by a procedure analogous to that described for compound 201.


Compound 115

Using compound 43 in place of compound 5, compound 100 is converted to compound 115 by a procedure analogous to that described for compound 101.


Compound 215

Compound 115 is converted to compound 215 by a procedure analogous to that described for compound 201.


Example 3—Alternative Synthesis of Phosphorothiotriesters Using Bis(Methanethiosulfonate) Reagents



embedded image


embedded image


embedded image


Compound 150

Compound 100 (300 mg, 0.264 mmol) was converted to compound 150 by a procedure analogous to that described for compound 101 (170 mg, 50%).



1H NMR (399 MHz, CDCl3) δ 9.34-9.30 (s, 1H), 9.28-9.17 (d, J 30.6 Hz, 1H), 7.57-7.47 (m, 1H), 7.47-7.40 (m, 2H), 7.38-7.18 (m, 17H), 7.18-7.07 (d, J 1.4 Hz, 1H), 6.88-6.77 (dd, J 9.0, 1.5 Hz, 8H), 6.44-6.34 (ddd, J 15.6, 8.9, 5.4 Hz, 1H), 6.32-6.21 (ddd, J 18.9, 8.5, 5.9 Hz, 1H), 5.27-5.19 (q, J 5.9 Hz, 1H), 4.46-4.33 (m, 2H), 4.31-4.16 (m, 2H), 4.03-3.91 (m, 2H), 3.81-3.67 (m, 12H), 3.54-3.46 (m, 1H), 3.42-3.34 (m, 1H), 3.34-3.25 (d, J 20.2 Hz, 3H), 2.64-2.53 (td, J 13.4, 5.4 Hz, 1H), 2.47-2.34 (dq, J 19.9, 6.5, 5.9 Hz, 1H), 1.99-1.91 (m, 1H), 1.85-1.80 (t, J 1.5 Hz, 3H), 1.78-1.65 (tt, J 14.1, 7.5 Hz, 1H), 1.44-1.37 (dd, J 7.3, 1.2 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 171.27, 163.83, 163.80, 158.95, 158.93, 158.90, 150.64, 150.53, 150.46, 150.38, 144.91, 144.88, 144.09, 144.02, 136.00, 135.98, 135.94, 135.81, 135.11, 135.04, 134.98, 134.97, 130.34, 130.27, 130.20, 128.30, 128.23, 128.20, 127.46, 127.36, 113.59, 113.56, 113.48, 111.95, 111.38, 87.60, 87.47, 87.43, 86.03, 85.83, 84.44, 84.34, 83.81, 79.82, 79.58, 73.99, 73.91, 67.85, 67.78, 63.31, 63.20, 55.39, 51.77, 51.70, 39.16, 38.99, 38.90, 37.21, 37.16, 37.12, 37.05, 12.63, 12.57, 11.85, 11.80; 31P NMR (162 MHz, CDCl3) δ 26.15, 25.60; MS (ESI+ve): calc (M+H): 1308.37, found: 1308.70. Rf=0.5 (5% MeOH/DCM).


Compound 151

A DCM (5 mL) solution of compound 150 (150 mg, 0.116 mmol) was treated with 2-morpholinoethanethiol (17 mg, 0.116 mmol) at r.t. with monitoring by TLC. After 0.5 h, the mixture was washed with NaHCO3, extracting 5× into DCM. The organic extracts were dried (MgSO4), filtered and reduced. Column chromatography gave compound 151 as a colorless solid foam (81 mg, 51%).



1H NMR (399 MHz, CDCl3) δ 9.68-9.54 (m, 1H), 9.44 (s, 1H), 7.59-7.48 (m, 1H), 7.47-7.40 (m, 2H), 7.40-7.13 (m, 17H), 6.90-6.76 (ddd, J=9.3, 4.4, 2.7 Hz, 8H), 6.45-6.27 (m, 2H), 5.32-5.22 (dd, J=8.5, 5.7 Hz, 1H), 4.34-4.25 (m, 1H), 4.23-4.14 (m, 1H), 4.07-3.89 (m, 2H), 3.79-3.74 (m, 12H), 3.74-3.65 (m, 6H), 3.51-3.33 (m, 2H), 2.90-2.79 (dd, J=14.2, 7.6 Hz, 2H), 2.73-2.55 (m, 3H), 2.55-2.34 (m, 6H), 2.02-1.91 (m, 1H), 1.87-1.81 (dd, J=4.9, 1.2 Hz, 3H), 1.77-1.66 (ddd, J=14.2, 8.7, 6.4 Hz, 1H), 1.41-1.35 (dd, J=6.6, 1.2 Hz, 3H); 13C NMR (100 MHz, CDCl3) δ 163.97, 163.93, 163.88, 158.90, 158.86, 158.71, 150.64, 150.59, 150.53, 150.50, 144.92, 144.88, 144.13, 144.08, 136.11, 136.07, 136.03, 136.00, 135.73, 135.60, 135.22, 135.14, 135.08, 135.04, 135.02, 130.32, 130.30, 130.23, 130.18, 128.33, 128.19, 128.17, 127.39, 127.33, 113.56, 113.52, 113.45, 111.85, 111.82, 111.38, 111.29, 87.56, 87.41, 87.38, 85.71, 85.35, 84.91, 84.38, 84.27, 84.22, 84.05, 83.97, 83.85, 83.78, 79.36, 79.11, 79.05, 74.25, 74.07, 67.39, 66.88, 66.79, 63.27, 57.80, 55.36, 53.55, 53.51, 53.40, 43.06, 40.72, 40.54, 39.25, 39.16, 39.01, 35.91, 12.64, 12.60, 11.78, 11.74; 31P NMR (162 MHz, CDCl3) δ 27.76, 27.46; MS (ESI+ve): calc (M+H): 1358.43, found: 1358.74. Rf=0.4 (5% MeOH/DCM).


Compound 251

Compound 151 (75 mg, 0.055 mmol) was converted to compound 251 by a procedure analogous to that described for compound 201 (10 mg, 24%). MS (ESI+ve): calc (M+H): 754.17, found: 754.19. Rf=0.3 (10% MeOH/DCM).


Compound 152

Compound 100 is converted to compound 152 by a procedure analogous to that described for compound 101.


Compound 153

Using 1-Thio-β-D-glucose tetraacetate in place of compound 4, compound 152 is converted to compound 153 by a procedure analogous to that described for compound 151.


Compound 253

Compound 153 is converted to compound 253 by a procedure analogous to that described for compound 201.




embedded image


Compound 154

Compound 100 is converted to compound 154 by a procedure analogous to that described for compound 101.


Compound 155

Compound 154 is converted to compound 155 by a procedure analogous to that described for compound 151.


Compound 255

Compound 155 is converted to compound 255 by a procedure analogous to that described for compound 201.


Example 4—Thioalkylation of H-Phosphonates to Provide Phosphorothiotriesters in Solid Phase
Compound 300

Synthesis of (Rp)-CAGT-H-phosphonate-oxalyl linker-CPG was carried out on an Applied Biosystems 394 DNA/RNA synthesizer according to the reported methods (Journal of American Chemical Society 2008, 130, 16031-16037; Angewandte Chemie International Edition 2009, 48, 496-499).


Compound 301: (Sp)-CAGT-phosphorothioate (R═H)

(Rp)-CAGT-H-phosphonate-oxalyl linker-CPG was treated by 0.2 M Beaucage Reagent/CH3CN-BSA (9:1, v/v), stirred for 1 h at rt, then washed successively with CS2 and acetonitrile and dried under reduced pressure. The resultant CPG was treated with 2 mL of 28% aqueous NH3 and stirred for 18 h at rt. After removal of NH3 under reduced pressure, the resulting product was analyzed by LC/MS and HPLC.


Compound 302: (Sp)-CAGT-S-methyl phosphorothiotriester (R=Me)

BSTFA (50 μL, 188 μmol) and acetonitrile (500 μL) were added to (Rp)-CAGT-H-phosphonate-oxalyl linker-CPG (14.7 mg, 1 μmol) then the mixture was shaken for 20 min at rt. S-methyl methane sulfonothioate (20 μL, 212 μmol) and NEt3 (50 μL) were added and shaking was continued for 1 h at rt. The CPG was washed with CH3CN then dried in vacuo. 20% PrNH2 in dry CH3CN (2 mL) was added to the CPG and the mixture was stirred for 16 h at rt. Solvents were removed under reduced pressure and CH3CN was added to the mixture. The CPG was removed by filtration and the filtrate was concentrated under reduced pressure. CH3CN/DMSO/0.5 M AA buffer (1:1:1, v/v/v) was added, the mixture was stirred for 16 h at rt, then analyzed by LC/MS and HPLC.


Compound 303

Compound 303 is prepared by sulfurization of compound 300 on support followed by cleavage. ACN (450 μL), BSTFA (50 μL) and compound 12 (20 mg) are added to compound 300 (1 μmol) which is shaken for 18 h. The CPG is collected by filtration resuspended in 20% PrNH2 in dry CH3CN (2 mL) and shaken for 16 h at rt. Solvents were removed under reduced pressure and the residue is purified by RPHPLC to provide pure compound 303.


Example 5—Thioalkylation of H-Phosphonates to Provide Phosphorothiotriesters in Solution Phase



embedded image


Compound 305

Compound 300 (0.5 μmol) was taken up in ACN (125 μL) then BSTFA (62 μL) was added and the mixture was shaken for 20 min. PrNH2 (125 μL) was added and the vial was rotated for 18 h. After filtration and washing with 1 mL ACN, the solvent was removed in vacuo and the residue was co-evaporated 3× with toluene to provide crude compound 304. The residue was redissolved in pyridine (375 μL) and treated with BSTFA for (16 Cl, 60.0 μmol) followed by compound 9 (7.2 mg, 30.0 μmol) with stirring under Ar. After 2 h at r.t. the solvent was removed and the residue was treated with MeOH (0.125 mL) for 1 h, then AA (0.5 M, 0.125 mL) was added and the mixture was stirred at r.t. for 2 h. The product was purified by RPHPLC to provide compound 305.


Compound 303

Substituting compound 12 for compound 9, compound 303 was prepared by a procedure analogous to that described for compound 305.


Compound 306

Substituting compound 12 for compound 14, compound 306 was prepared by a procedure analogous to that described for compound 305.


Compound 307

Substituting compound 12 for compound 29, compound 307 is prepared by a procedure analogous to that described for compound 305.


Compound 308

Substituting compound 12 for compound 31, compound 308 is prepared by a procedure analogous to that described for compound 305.


Compound 309

Substituting compound 12 for compound 38, compound 309 is prepared by a procedure analogous to that described for compound 305.


Example 6—Stereoselective Thioalkylation of H-Phosphanates

Objective: To demonstrate that the reaction of MTS reagents to H-phosphonate to generate phosphorothio triester is stereospecific. 31P NMR was used to trace the changes during the course of the reaction.




embedded image


Experimental procedure: In an NMR tube was added compound 100S 5′-O-(4,4′-dimethoxytrityl)thymidin-3′-yl 3′-O-(4,4′-dimethoxytrityl)thymidin-5′-yl H-phosphonate (20 mg, 18 μmol) in 0.8 mL CD3CN and the 31P NMR spectrum was recorded. BSTFA (17 μL, 176 μmol) was added to same NMR tube and after 5 min 31P NMR spectrum was recorded again. Triethylamine (49 μL, 352 μmol) and S-methyl methanethiosulfonate (22 μL, 88 μmol) were added to same NMR tube and 31P NMR spectrum was recorded immediately.


The same procedure was repeated for Rp isomer (compound 100R). The 31P NMR spectrum recorded for the starting material, intermediate and the product show that the stereochemistry at phosphorus atom is retained during the reaction.


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.

Claims
  • 1. A thiosulfonate compound of structure IIa:
  • 2. The thiosulfonate compound of claim 1, wherein R1 is selected from -alkenylenyl-S— and -alkylenyl-S—.
  • 3. The thiosulfonate compound of claim 1, wherein R2 is —S-alkylenyl-heterocyclo.
  • 4. The thiosulfonate compound of claim 1, wherein R1 is selected from:
  • 5. The thiosulfonate compound of claim 1, wherein R2 is selected from:
  • 6. The thiosulfonate compound of claim 1, wherein X is alkyl.
  • 7. The thiosulfonate compound of claim 6, wherein X is methyl.
  • 8. A compound selected from
  • 9. The compound of claim 8, wherein the compound is
  • 10. The compound of claim 8, wherein the compound is
  • 11. The compound of claim 8, wherein the compound is
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation application of U.S. patent application Ser. No. 15/222,910, filed Jul. 28, 2016, which is a continuation of U.S. patent application Ser. No. 14/233,579, filed Feb. 27, 2014 (now U.S. Pat. No. 9,605,019), which is a 371 national phase entry of International Application No. PCT/US12/46805, which claims priority to U.S. Provisional Application No. 61/509,526, filed Jul. 19, 2011. The contents of these priority applications are hereby incorporated by reference in their entirety.

US Referenced Citations (746)
Number Name Date Kind
2878264 Lunsford Mar 1959 A
3135766 Gould Jun 1964 A
3484473 Buckman et al. Dec 1969 A
3687808 Merigan et al. Aug 1972 A
3745162 Helsley Jul 1973 A
4022791 Welch, Jr. May 1977 A
4113869 Gardner Sep 1978 A
4415732 Caruthers et al. Nov 1983 A
4458066 Caruthers et al. Jul 1984 A
4500707 Caruthers et al. Feb 1985 A
4542142 Martel et al. Sep 1985 A
4659774 Webb et al. Apr 1987 A
4663328 Lafon May 1987 A
4668777 Caruthers et al. May 1987 A
4725677 Koster et al. Feb 1988 A
4735949 Domagala et al. Apr 1988 A
4840956 Domagala et al. Jun 1989 A
4845205 Huynh Dinh et al. Jul 1989 A
4923901 Koester et al. May 1990 A
4943629 DeVries et al. Jul 1990 A
4945158 DeVries et al. Jul 1990 A
4973679 Caruthers et al. Nov 1990 A
4981957 Lebleu et al. Jan 1991 A
5047524 Andrus et al. Sep 1991 A
5118800 Smith et al. Jun 1992 A
5130302 Spielvogel et al. Jul 1992 A
5132418 Caruthers et al. Jul 1992 A
5134066 Rogers et al. Jul 1992 A
5138045 Cook et al. Aug 1992 A
5141813 Nelson Aug 1992 A
5151510 Stec et al. Sep 1992 A
5175273 Bischofberger et al. Dec 1992 A
5200553 Nudelman et al. Apr 1993 A
5212295 Cook May 1993 A
5223618 Cook et al. Jun 1993 A
5262530 Andrus et al. Nov 1993 A
5292875 Stec et al. Mar 1994 A
5319080 Leumann Jun 1994 A
5359044 Cook et al. Oct 1994 A
5367066 Urdea et al. Nov 1994 A
5378825 Cook et al. Jan 1995 A
5386023 Sanghvi et al. Jan 1995 A
5432272 Benner Jul 1995 A
5457187 Gmeiner et al. Oct 1995 A
5457191 Cook et al. Oct 1995 A
5459255 Cook et al. Oct 1995 A
5484908 Froehler et al. Jan 1996 A
5489677 Sanghvi et al. Feb 1996 A
5502177 Matteucci et al. Mar 1996 A
5506212 Hoke et al. Apr 1996 A
5512668 Stec et al. Apr 1996 A
5521302 Cook May 1996 A
5525711 Hawkins et al. Jun 1996 A
5541307 Cook et al. Jul 1996 A
5552540 Haralambidis Sep 1996 A
5565488 Braunlich et al. Oct 1996 A
5576302 Cook et al. Nov 1996 A
5587361 Cook et al. Dec 1996 A
5587469 Cook et al. Dec 1996 A
5594121 Froehler et al. Jan 1997 A
5596091 Switzer Jan 1997 A
5599797 Cook et al. Feb 1997 A
5602240 De Mesmaeker et al. Feb 1997 A
5607923 Cook et al. Mar 1997 A
5608046 Cook et al. Mar 1997 A
5610289 Cook et al. Mar 1997 A
5614617 Cook et al. Mar 1997 A
5618704 Sanghvi et al. Apr 1997 A
5620963 Cook et al. Apr 1997 A
5622989 Br aunlich et al. Apr 1997 A
5623070 Cook et al. Apr 1997 A
5635488 Cook et al. Jun 1997 A
5643889 Suhadolnik et al. Jul 1997 A
5643989 Van De Grampel et al. Jul 1997 A
5646267 Stec et al. Jul 1997 A
5654284 Cook et al. Aug 1997 A
5661134 Cook et al. Aug 1997 A
5677437 Teng et al. Oct 1997 A
5681940 Wang et al. Oct 1997 A
5681941 Cook et al. Oct 1997 A
5688941 Cook et al. Nov 1997 A
5708161 Reese Jan 1998 A
5712378 Wang Jan 1998 A
5734041 Just et al. Mar 1998 A
5750692 Cook et al. May 1998 A
5777092 Cook et al. Jul 1998 A
5783682 Cook et al. Jul 1998 A
5792844 Sanghvi et al. Aug 1998 A
5795765 Izu et al. Aug 1998 A
5808023 Sanghvi et al. Sep 1998 A
5824503 Kurome et al. Oct 1998 A
5834607 Manoharan et al. Nov 1998 A
5846466 Abe et al. Dec 1998 A
5851840 Sluka et al. Dec 1998 A
5852188 Cook Dec 1998 A
5856465 Stec et al. Jan 1999 A
5883237 Stec et al. Mar 1999 A
5892024 Chaturvedula et al. Apr 1999 A
5898031 Crooke Apr 1999 A
5908772 Mitta et al. Jun 1999 A
5914396 Cook et al. Jun 1999 A
5932450 Dattagupta et al. Aug 1999 A
5936080 Stec et al. Aug 1999 A
5965721 Cook et al. Oct 1999 A
5969118 Sanghvi et al. Oct 1999 A
5976855 Svendsen et al. Nov 1999 A
5998148 Bennett et al. Dec 1999 A
5998602 Torrence et al. Dec 1999 A
5998603 Cook et al. Dec 1999 A
6004813 Serlupi-Crescenzi et al. Dec 1999 A
6005107 Nguyen-Ba et al. Dec 1999 A
6015886 Dale et al. Jan 2000 A
6015887 Teng Jan 2000 A
6017700 Horn et al. Jan 2000 A
6025482 Cook et al. Feb 2000 A
6031092 Just et al. Feb 2000 A
6056973 Allen et al. May 2000 A
6057371 Glennon May 2000 A
6066500 Bennett et al. May 2000 A
6080543 Engel et al. Jun 2000 A
6087482 Teng et al. Jul 2000 A
6107094 Crooke Aug 2000 A
6121433 Cook et al. Sep 2000 A
6124445 Imbach et al. Sep 2000 A
6127540 Nguyen-Ba et al. Oct 2000 A
6133438 Cook et al. Oct 2000 A
6140096 Kofod et al. Oct 2000 A
6146829 Cook et al. Nov 2000 A
6147200 Manoharan et al. Nov 2000 A
6159728 Stockley et al. Dec 2000 A
6160109 Just et al. Dec 2000 A
6166197 Cook et al. Dec 2000 A
6172209 Manoharan et al. Jan 2001 B1
6191266 Wang Feb 2001 B1
6194576 Nguyen-Ba et al. Feb 2001 B1
6207646 Krieg et al. Mar 2001 B1
6214551 Sanghvi et al. Apr 2001 B1
6214805 Torrence et al. Apr 2001 B1
6222025 Cook et al. Apr 2001 B1
6232463 Cook et al. May 2001 B1
6235887 Froehler et al. May 2001 B1
6239116 Krieg et al. May 2001 B1
6239265 Cook May 2001 B1
6242589 Cook et al. Jun 2001 B1
6248519 Engel et al. Jun 2001 B1
6265172 St. Clair et al. Jul 2001 B1
6270968 Dalb.o slashed.ge et al. Aug 2001 B1
6271004 Warthoe Aug 2001 B1
6271357 Cook et al. Aug 2001 B1
6300069 Missel et al. Oct 2001 B1
6306627 Decker Oct 2001 B1
6316024 Allen et al. Nov 2001 B1
6316626 Swayze et al. Nov 2001 B1
6320040 Cook et al. Nov 2001 B1
6322985 Kashi et al. Nov 2001 B1
6326199 Cook et al. Dec 2001 B1
6339066 Bennett et al. Jan 2002 B1
6344356 Stemmer Feb 2002 B1
6369209 Manoharan et al. Apr 2002 B1
6369237 Verdine et al. Apr 2002 B1
6372492 Bennett et al. Apr 2002 B1
6380368 Froehler et al. Apr 2002 B1
6383808 Monia et al. May 2002 B1
6407223 Stec et al. Jun 2002 B1
6440739 Bennett et al. Aug 2002 B1
6440943 Cook et al. Aug 2002 B1
6444656 Nguyen-Ba et al. Sep 2002 B1
6451524 Ecker Sep 2002 B1
6455308 Freier Sep 2002 B1
6468983 Silverman et al. Oct 2002 B2
6495677 Ramasamy et al. Dec 2002 B1
6500945 Cook Dec 2002 B2
6506594 Barany et al. Jan 2003 B1
6506894 Reese et al. Jan 2003 B1
6528262 Gilad et al. Mar 2003 B1
6528640 Beigelman et al. Mar 2003 B1
6538126 Cho et al. Mar 2003 B1
6559279 Manoharan et al. May 2003 B1
6562960 Baxter et al. May 2003 B1
6582936 Serafini et al. Jun 2003 B1
6608186 Miculka et al. Aug 2003 B1
6610837 Guzaev et al. Aug 2003 B1
6613873 Buchardt et al. Sep 2003 B1
6617438 Beigelman et al. Sep 2003 B1
6632600 Short Oct 2003 B1
6639022 Michels et al. Oct 2003 B2
6639062 Manoharan et al. Oct 2003 B2
6649750 Capaldi et al. Nov 2003 B1
6670461 Wengel et al. Dec 2003 B1
6682889 Wang et al. Jan 2004 B1
6699979 Cook Mar 2004 B2
6737520 Manoharan et al. May 2004 B2
6762281 Manoharan et al. Jul 2004 B2
6767739 Crooke et al. Jul 2004 B2
6809195 Sanghvi et al. Oct 2004 B1
6811975 Cook et al. Nov 2004 B2
6815542 Hong et al. Nov 2004 B2
6861518 Just et al. Mar 2005 B2
6867294 Sanghvi et al. Mar 2005 B1
6900301 Cook et al. May 2005 B2
6933146 Helliwell et al. Aug 2005 B2
6933288 Migawa et al. Aug 2005 B2
6936432 Gopalan et al. Aug 2005 B2
6943240 Bauer et al. Sep 2005 B2
6949520 Hartmann et al. Sep 2005 B1
6977245 Klinman et al. Dec 2005 B2
6995259 Vargeese et al. Feb 2006 B1
7015315 Cook et al. Mar 2006 B1
7018793 Short Mar 2006 B1
7019127 Reese et al. Mar 2006 B2
7022833 Reese Apr 2006 B2
7030230 Ross et al. Apr 2006 B2
7045610 Dempcy et al. May 2006 B2
7049122 Chang et al. May 2006 B2
7067497 Hanecak et al. Jun 2006 B2
7101993 Cook et al. Sep 2006 B1
7119184 Manoharan et al. Oct 2006 B2
RE39464 Cook et al. Jan 2007 E
7160920 Garvey et al. Jan 2007 B2
7205399 Vargeese et al. Apr 2007 B1
7214491 Yadav et al. May 2007 B2
7227014 Crooke et al. Jun 2007 B2
7238795 Seela et al. Jul 2007 B2
7247621 Hong et al. Jul 2007 B2
7259150 Crooke et al. Aug 2007 B2
7264932 Latham et al. Sep 2007 B2
7268119 Cook et al. Sep 2007 B2
7271156 Krieg et al. Sep 2007 B2
7285658 Cook et al. Oct 2007 B2
7288376 Sarma et al. Oct 2007 B2
7303895 O'Regan et al. Dec 2007 B1
7304081 Yao et al. Dec 2007 B2
7354909 Klinman et al. Apr 2008 B2
7381527 Sarma et al. Jun 2008 B2
7399845 Seth et al. Jul 2008 B2
7407943 Crooke et al. Aug 2008 B2
7407965 Chen et al. Aug 2008 B2
7410975 Lipford et al. Aug 2008 B2
7414116 Milton et al. Aug 2008 B2
7425545 Crooke et al. Sep 2008 B2
7427672 Imanishi et al. Sep 2008 B2
7429565 Boojamra et al. Sep 2008 B2
7432249 Crooke Oct 2008 B2
7432250 Crooke Oct 2008 B2
7432261 Cannizzaro et al. Oct 2008 B2
7452901 Boojamra et al. Nov 2008 B2
7470724 Cannizzaro et al. Dec 2008 B2
7495088 Brakel et al. Feb 2009 B1
7501091 Munoz et al. Mar 2009 B2
7507808 Dobie Mar 2009 B2
7507811 Khvorova et al. Mar 2009 B2
7511131 Crooke et al. Mar 2009 B2
7517520 Manolova et al. Apr 2009 B2
7534879 van Deutekom May 2009 B2
7537767 Bachmann et al. May 2009 B2
7547684 Seth et al. Jun 2009 B2
7585847 Bratzler et al. Sep 2009 B2
7598031 Liew Oct 2009 B2
7598227 Crooke et al. Oct 2009 B2
7598230 Cook et al. Oct 2009 B2
7608594 Blagg et al. Oct 2009 B2
7622451 Blagg et al. Nov 2009 B2
7629321 Crooke Dec 2009 B2
7645747 Boojamra et al. Jan 2010 B2
7662558 Liew Feb 2010 B2
7666854 Seth et al. Feb 2010 B2
7666888 Bartberger et al. Feb 2010 B2
7683036 Esau et al. Mar 2010 B2
7695902 Crooke Apr 2010 B2
7696345 Allerson et al. Apr 2010 B2
7713941 Cook et al. May 2010 B2
7718623 Kitagawa et al. May 2010 B2
7723508 Crooke et al. May 2010 B2
7732590 Bhanot et al. Jun 2010 B2
7732660 Helliwell et al. Jun 2010 B2
7741305 Crooke et al. Jun 2010 B2
7741457 Seth et al. Jun 2010 B2
7749700 Baird et al. Jul 2010 B2
7750131 Seth et al. Jul 2010 B2
7750141 Crooke et al. Jul 2010 B2
7750731 Poulsen et al. Jul 2010 B2
7759318 Perera et al. Jul 2010 B1
7776344 Hartmann et al. Aug 2010 B2
7776874 Yao et al. Aug 2010 B2
7777023 Vargeese et al. Aug 2010 B2
7803930 Crooke et al. Sep 2010 B2
7807653 Cook et al. Oct 2010 B2
7807816 Wilton et al. Oct 2010 B2
7811998 Blagg et al. Oct 2010 B2
7812003 Safe et al. Oct 2010 B2
7838287 Goldsmith et al. Nov 2010 B2
7863252 Crooke et al. Jan 2011 B2
7884086 Bennett et al. Feb 2011 B2
7884117 Zhang et al. Feb 2011 B2
7888324 Crooke et al. Feb 2011 B2
7893039 Swayze et al. Feb 2011 B2
7919472 Monia et al. Apr 2011 B2
7947658 Aronin et al. May 2011 B2
7951934 Freier May 2011 B2
7960353 Blagg Jun 2011 B2
7960541 Wilton et al. Jun 2011 B2
7973015 van Ommen et al. Jul 2011 B2
8003619 Hartmann et al. Aug 2011 B2
8008011 Schmutz et al. Aug 2011 B2
8008459 Goldsmith et al. Aug 2011 B2
8022083 Boojamra et al. Sep 2011 B2
8039235 Lin et al. Oct 2011 B2
8057997 Seela et al. Nov 2011 B2
8058288 Yao et al. Nov 2011 B2
8067173 Liew Nov 2011 B2
8076303 Iyer et al. Dec 2011 B2
8084437 Freier et al. Dec 2011 B2
8084600 Natt et al. Dec 2011 B2
8088582 Sampath et al. Jan 2012 B2
8093222 Freier et al. Jan 2012 B2
8093225 Mamet Jan 2012 B2
8101348 Tuschl et al. Jan 2012 B2
8101358 Liew Jan 2012 B2
8101585 Yu et al. Jan 2012 B2
8101743 Brown-Driver et al. Jan 2012 B2
8106025 Bennett et al. Jan 2012 B2
8110358 Liew Feb 2012 B2
8110558 Bennett et al. Feb 2012 B2
8114597 Liew Feb 2012 B2
8124745 Allerson et al. Feb 2012 B2
8133674 Liew Mar 2012 B2
8133675 Liew Mar 2012 B2
8133876 Bennett et al. Mar 2012 B2
8138328 Crooke et al. Mar 2012 B2
8143230 Bhanot et al. Mar 2012 B2
8148072 Liew Apr 2012 B2
8158598 Bhanot et al. Apr 2012 B2
8163707 Qiu et al. Apr 2012 B2
8178506 Lollo et al. May 2012 B2
8188059 Bhanot et al. May 2012 B2
8206923 Garza Gonzalez et al. Jun 2012 B2
8207263 Popot et al. Jun 2012 B2
8212011 Blagg Jul 2012 B2
8212012 Blagg Jul 2012 B2
8226759 Shin et al. Jul 2012 B2
8232384 Wilton et al. Jul 2012 B2
8257922 Liew Sep 2012 B2
8258289 Bhanot et al. Sep 2012 B2
8350022 Meier et al. Jan 2013 B2
8361977 Baker et al. Jan 2013 B2
8383660 Chang et al. Feb 2013 B2
8410070 Miller et al. Apr 2013 B2
8415465 Freier Apr 2013 B2
8431693 Manoharan et al. Apr 2013 B2
8450474 Wilton et al. May 2013 B2
8455634 Wilton et al. Jun 2013 B2
8455635 Wilton et al. Jun 2013 B2
8455636 Wilton et al. Jun 2013 B2
8470987 Wada et al. Jun 2013 B2
8476423 Wilton et al. Jul 2013 B2
8481710 Davidson et al. Jul 2013 B2
8486907 Wilton et al. Jul 2013 B2
8501414 Danzer et al. Aug 2013 B2
8524880 Wilton et al. Sep 2013 B2
8557549 Chang et al. Oct 2013 B2
8557844 Platt et al. Oct 2013 B2
8592566 Iwamura et al. Nov 2013 B2
8632963 Shah et al. Jan 2014 B2
8633206 Promo et al. Jan 2014 B2
8647742 Dendukuri et al. Feb 2014 B2
8648186 Monteleone Feb 2014 B2
8653254 Umemoto et al. Feb 2014 B2
8669058 Liew Mar 2014 B2
8674044 Popot et al. Mar 2014 B2
8679750 Hayden et al. Mar 2014 B2
8680063 Aronin et al. Mar 2014 B2
8729036 Zamore et al. May 2014 B2
8735417 Altman et al. May 2014 B2
8750507 Roosta et al. Jun 2014 B2
8754107 George et al. Jun 2014 B2
8759507 Van Deutekom Jun 2014 B2
8802659 Thomas et al. Aug 2014 B2
8809516 Manoharan et al. Aug 2014 B2
8815817 Hessel et al. Aug 2014 B2
8822671 Shimizu et al. Sep 2014 B2
8859755 Wada et al. Oct 2014 B2
8865146 Fukuhara et al. Oct 2014 B2
8871785 Boojamra et al. Oct 2014 B2
8877435 Helliwell et al. Nov 2014 B2
8883752 Swayze et al. Nov 2014 B2
8883969 Ide et al. Nov 2014 B2
8927513 Manoharan et al. Jan 2015 B2
8952145 Freier Feb 2015 B2
8957040 Bennett et al. Feb 2015 B2
8957042 Safe et al. Feb 2015 B2
8975389 Manoharan et al. Mar 2015 B2
8980853 Bennett et al. Mar 2015 B2
8987222 Aronin et al. Mar 2015 B2
8987435 Swayze et al. Mar 2015 B2
8993738 Prakash et al. Mar 2015 B2
9006198 Bennett et al. Apr 2015 B2
9018368 Wilton et al. Apr 2015 B2
9024007 Wilton et al. May 2015 B2
9035040 Wilton et al. May 2015 B2
9040674 Benson et al. May 2015 B2
9057066 Hung et al. Jun 2015 B2
9120774 Blagg et al. Sep 2015 B2
9121020 Feinstein et al. Sep 2015 B2
9126927 Yao et al. Sep 2015 B2
9127033 Prakash et al. Sep 2015 B2
9127123 Livingston et al. Sep 2015 B2
9132289 Kawai Sep 2015 B2
9139604 Boojamra et al. Sep 2015 B2
9175286 Wilton et al. Nov 2015 B2
9186367 Thomas et al. Nov 2015 B2
9249416 Wilton et al. Feb 2016 B2
9260716 Davidson et al. Feb 2016 B2
9273315 Hung et al. Mar 2016 B2
9284344 Kim et al. Mar 2016 B2
9308252 Suckow et al. Apr 2016 B2
9321799 Prakash et al. Apr 2016 B2
9353372 Freier May 2016 B2
9382540 Prakash et al. Jul 2016 B2
9382575 Eom et al. Jul 2016 B2
9394333 Wada et al. Jul 2016 B2
9422555 Wilton et al. Aug 2016 B2
9428541 Platt et al. Aug 2016 B2
9441229 Wilton et al. Sep 2016 B2
9447415 Wilton et al. Sep 2016 B2
9453228 Kandimalla et al. Sep 2016 B2
9476044 Tuschl et al. Oct 2016 B2
9480740 Reed et al. Nov 2016 B2
9481704 Clarke Nov 2016 B2
9572824 Thomas et al. Feb 2017 B2
9598458 Shimizu et al. Mar 2017 B2
9605019 Verdine et al. Mar 2017 B2
9605262 Wilton et al. Mar 2017 B2
9605263 Rigo Mar 2017 B2
9611472 Zamore et al. Apr 2017 B2
9617547 Gemba Apr 2017 B2
9695211 Wada et al. Jul 2017 B2
9725474 Murata et al. Aug 2017 B2
9738895 Swayze et al. Aug 2017 B2
9744183 Verdine et al. Aug 2017 B2
9809616 Amblard et al. Nov 2017 B2
9827258 Thomas et al. Nov 2017 B2
9885082 Hrdlicka Feb 2018 B2
9896688 Chang et al. Feb 2018 B2
9982257 Butler May 2018 B2
20010055761 Kanemoto et al. Dec 2001 A1
20020013792 Imielinski et al. Jan 2002 A1
20020082227 Henry Jun 2002 A1
20020137921 Cook Sep 2002 A1
20020183502 Mesmaeker et al. Dec 2002 A1
20030045705 Cook et al. Mar 2003 A1
20030049662 Monia et al. Mar 2003 A1
20030050261 Krieg et al. Mar 2003 A1
20030159938 Hradil Aug 2003 A1
20030198982 Seela et al. Oct 2003 A1
20030232978 Seeberger et al. Dec 2003 A1
20030235845 van Ommen et al. Dec 2003 A1
20040002596 Hong et al. Jan 2004 A1
20040023901 Cook et al. Feb 2004 A1
20040023921 Hong et al. Feb 2004 A1
20040059104 Cook et al. Mar 2004 A1
20040063647 Johnson Apr 2004 A1
20040149587 Hradil Aug 2004 A1
20040203145 Zamore et al. Oct 2004 A1
20040213780 Krainc Oct 2004 A1
20050042646 Davidson et al. Feb 2005 A1
20050059619 Krieg et al. Mar 2005 A1
20050096284 McSwiggen May 2005 A1
20050159375 Srivastava et al. Jul 2005 A1
20050169888 Hartmann et al. Aug 2005 A1
20050176045 Fedorov et al. Aug 2005 A1
20050203044 Zinnen Sep 2005 A1
20050215513 Boojamra et al. Sep 2005 A1
20050227947 Chen et al. Oct 2005 A1
20050239102 Verdine et al. Oct 2005 A1
20050261237 Boojamra et al. Nov 2005 A1
20050277133 McSwiggen Dec 2005 A1
20050277609 Krieg et al. Dec 2005 A1
20060003962 Ahluwalia et al. Jan 2006 A1
20060035858 Geary et al. Feb 2006 A1
20060035866 Cannizzaro et al. Feb 2006 A1
20060041115 Ravikumar Feb 2006 A1
20060063730 Monia et al. Mar 2006 A1
20060079478 Boojamra et al. Apr 2006 A1
20060099616 van Ommen et al. May 2006 A1
20060147952 van Ommen et al. Jul 2006 A1
20060166910 Tuschl et al. Jul 2006 A1
20060199776 Blagg et al. Sep 2006 A1
20060199788 Cannizzaro et al. Sep 2006 A1
20060211644 Krieg et al. Sep 2006 A1
20060257912 Kaemmerer et al. Nov 2006 A1
20060264404 Boojamra et al. Nov 2006 A1
20070027116 Cho et al. Feb 2007 A1
20070099851 Linn May 2007 A1
20070099860 Sah et al. May 2007 A1
20070123484 Bhat et al. May 2007 A1
20070135363 Cook et al. Jun 2007 A1
20070149462 Iyer et al. Jun 2007 A1
20070161547 Bhat et al. Jul 2007 A1
20070161590 Van Bilsen et al. Jul 2007 A1
20070196852 Heindl et al. Aug 2007 A1
20070249589 Aebi et al. Oct 2007 A1
20070259832 Cook et al. Nov 2007 A1
20070265224 Cook et al. Nov 2007 A1
20070270452 Blagg et al. Nov 2007 A1
20070282097 Ohgi et al. Dec 2007 A1
20070287831 Seth et al. Dec 2007 A1
20070299027 Hung et al. Dec 2007 A1
20080015158 Ichiro et al. Jan 2008 A1
20080015162 Bhanot et al. Jan 2008 A1
20080039418 Freier Feb 2008 A1
20080045473 Uhlmann et al. Feb 2008 A1
20080064867 Leuck et al. Mar 2008 A1
20080119426 Dale May 2008 A1
20080200409 Wilson et al. Aug 2008 A1
20080200423 Cook et al. Aug 2008 A1
20080209581 van Ommen et al. Aug 2008 A1
20080221055 Sah et al. Sep 2008 A1
20080221303 Katzhendler et al. Sep 2008 A1
20080249291 Kwon et al. Oct 2008 A1
20080274989 Davidson et al. Nov 2008 A1
20090012120 Borhan et al. Jan 2009 A1
20090023675 McSwiggen et al. Jan 2009 A1
20090053148 Kandimalla et al. Feb 2009 A1
20090053205 Kandimalla et al. Feb 2009 A1
20090060898 Kandimalla et al. Mar 2009 A1
20090062224 Kim et al. Mar 2009 A1
20090076246 van Deutekom Mar 2009 A1
20090093425 Dowdy et al. Apr 2009 A1
20090131372 Chen et al. May 2009 A1
20090162316 Verdine et al. Jun 2009 A1
20090163709 Blagg Jun 2009 A1
20090186410 Aronin et al. Jul 2009 A1
20090187014 Blagg Jul 2009 A1
20090227543 Cannizzaro et al. Sep 2009 A1
20090228998 Van Ommen et al. Sep 2009 A1
20090247488 Cannizzaro et al. Oct 2009 A1
20090263413 Iwamura et al. Oct 2009 A1
20090275535 Boojamra et al. Nov 2009 A1
20090306176 Schlingensiepen et al. Dec 2009 A1
20100008937 Peer et al. Jan 2010 A1
20100008981 Kaemmerer et al. Jan 2010 A1
20100022467 Boojamra et al. Jan 2010 A1
20100022620 Crispin et al. Jan 2010 A1
20100038543 Toda et al. Feb 2010 A1
20100048882 Blagg et al. Feb 2010 A1
20100069472 Hung et al. Mar 2010 A1
20100074889 Qiu et al. Mar 2010 A1
20100105630 Blagg Apr 2010 A1
20100120900 van Bilsen et al. May 2010 A1
20100186626 Shin et al. Jul 2010 A1
20100203002 Fukuhara et al. Aug 2010 A1
20100204162 Platt et al. Aug 2010 A1
20100215642 Lan et al. Aug 2010 A1
20100273999 Jung et al. Oct 2010 A1
20100299768 Perrin et al. Nov 2010 A1
20100311684 Cook et al. Dec 2010 A1
20100325746 Kaemmerer et al. Dec 2010 A9
20110009477 Yu et al. Jan 2011 A1
20110015253 Wilton et al. Jan 2011 A1
20110015258 Wilton et al. Jan 2011 A1
20110021365 Seela et al. Jan 2011 A1
20110039334 Bennett et al. Feb 2011 A1
20110046203 Wilton et al. Feb 2011 A1
20110071101 Boojamra et al. Mar 2011 A1
20110105587 Fishcher et al. May 2011 A1
20110111491 Davidson et al. May 2011 A1
20110136765 Promo et al. Jun 2011 A1
20110178284 Wada et al. Jul 2011 A1
20110201599 Bahceci et al. Aug 2011 A1
20110212520 Davidson et al. Sep 2011 A1
20110213010 Hayden et al. Sep 2011 A1
20110257251 Gude-Rodriguez et al. Oct 2011 A1
20110263686 Wilton et al. Oct 2011 A1
20110269814 Manoharan et al. Nov 2011 A1
20110269821 Swayze et al. Nov 2011 A1
20110288053 Boojamra et al. Nov 2011 A1
20110294124 Wada et al. Dec 2011 A1
20110294869 Petersen Dec 2011 A1
20110306652 Freier Dec 2011 A1
20110312086 Van Deutekom Dec 2011 A1
20120022144 Wilton et al. Jan 2012 A1
20120022145 Wilton et al. Jan 2012 A1
20120029057 Wilton et al. Feb 2012 A1
20120029058 Wilton et al. Feb 2012 A1
20120029059 Wilton et al. Feb 2012 A1
20120029060 Wilton et al. Feb 2012 A1
20120041050 Wilton et al. Feb 2012 A1
20120059045 Prakash et al. Mar 2012 A1
20120064137 Kawai Mar 2012 A1
20120095076 Sah et al. Apr 2012 A1
20120108800 Murata et al. May 2012 A1
20120136039 Aronin et al. May 2012 A1
20120156138 Smith Jun 2012 A1
20120157511 Manoharan et al. Jun 2012 A1
20120190649 Thomas et al. Jul 2012 A1
20120208864 Bhanot et al. Aug 2012 A1
20120214865 Bennett et al. Aug 2012 A1
20120216823 Fukuhara et al. Aug 2012 A1
20120246747 Tuschl et al. Sep 2012 A1
20120252745 Blagg et al. Oct 2012 A1
20120252879 Hung et al. Oct 2012 A1
20120276037 Suzuki et al. Nov 2012 A1
20120308609 Gibbon et al. Dec 2012 A1
20120316224 Verdine et al. Dec 2012 A1
20130005794 Kaemmerer et al. Jan 2013 A1
20130046008 Bennett et al. Feb 2013 A1
20130072671 Van Deutekom Mar 2013 A1
20130084576 Prakash et al. Apr 2013 A1
20130116310 Wilton et al. May 2013 A1
20130116420 Prakash et al. May 2013 A1
20130156845 Manoharan et al. Jun 2013 A1
20130178612 Wada et al. Jul 2013 A1
20130184450 Wada et al. Jul 2013 A1
20130189782 Hung et al. Jul 2013 A1
20130197061 Hohjoh et al. Aug 2013 A1
20130217755 Wilton et al. Aug 2013 A1
20130236536 Phiasivongsa et al. Sep 2013 A1
20130243725 Clarke Sep 2013 A1
20130253033 Wilton et al. Sep 2013 A1
20130253178 Shimizu et al. Sep 2013 A1
20130253180 Wilton et al. Sep 2013 A1
20130274313 Wilton et al. Oct 2013 A1
20130281684 Freier Oct 2013 A1
20130302806 Van Deutekom Nov 2013 A1
20130316969 Boojamra et al. Nov 2013 A1
20130323836 Manoharan et al. Dec 2013 A1
20130331438 Wilton et al. Dec 2013 A1
20140080769 Platt et al. Mar 2014 A1
20140080896 Nelson et al. Mar 2014 A1
20140080898 Wilton et al. Mar 2014 A1
20140107330 Freier et al. Apr 2014 A1
20140120088 Carpentier May 2014 A1
20140142160 Lee et al. May 2014 A1
20140155587 Wilton et al. Jun 2014 A1
20140163213 Debelak et al. Jun 2014 A1
20140194610 Verdine et al. Jul 2014 A1
20140213635 Van Deutekom Jul 2014 A1
20140220573 Hrdlicka Aug 2014 A1
20140221395 Dhanoa Aug 2014 A1
20140235566 Amblard et al. Aug 2014 A1
20140243515 Wilton et al. Aug 2014 A1
20140243516 Wilton et al. Aug 2014 A1
20140255936 Rademakers et al. Sep 2014 A1
20140256578 Hayden et al. Sep 2014 A1
20140275212 van Deutekom Sep 2014 A1
20140303238 Linsley et al. Oct 2014 A1
20140309190 Thomas et al. Oct 2014 A1
20140309283 Wilton et al. Oct 2014 A1
20140309284 Wilton et al. Oct 2014 A1
20140309285 Wilton et al. Oct 2014 A1
20140316121 Prakash et al. Oct 2014 A1
20140323707 Seth et al. Oct 2014 A1
20140350076 van Deutekom Nov 2014 A1
20140357698 Van Deutekom et al. Dec 2014 A1
20140357855 Van Deutekom et al. Dec 2014 A1
20140373188 Zamore et al. Dec 2014 A1
20140378527 van Deutekom Dec 2014 A1
20150025039 Boojamra et al. Jan 2015 A1
20150051389 Seth et al. Feb 2015 A1
20150057330 Wilton et al. Feb 2015 A1
20150080457 Manoharan et al. Mar 2015 A1
20150080563 van Deutekom Mar 2015 A2
20150096064 Tuschl et al. Apr 2015 A1
20150126725 Swayze et al. May 2015 A1
20150148404 de Visser et al. May 2015 A1
20150159163 Prakash et al. Jun 2015 A1
20150166999 Gemba Jun 2015 A1
20150167006 Swayze et al. Jun 2015 A1
20150197540 Shimizu et al. Jul 2015 A1
20150211006 Butler et al. Jul 2015 A1
20150218559 Van Deutekom et al. Aug 2015 A1
20150259679 Bennett et al. Sep 2015 A1
20150267197 Bennett et al. Sep 2015 A1
20150275208 Oestergaard et al. Oct 2015 A1
20150291636 Atamanyuk et al. Oct 2015 A1
20150292015 Bennett et al. Oct 2015 A1
20150307877 Freier Oct 2015 A1
20150315594 Prakash et al. Nov 2015 A1
20150322434 van Deutekom Nov 2015 A1
20150329859 Bennett et al. Nov 2015 A1
20150335708 Froelich et al. Nov 2015 A1
20150353931 Wilton et al. Dec 2015 A1
20150361424 van Deutekom Dec 2015 A1
20150376615 Wilton et al. Dec 2015 A1
20150376616 Wilton et al. Dec 2015 A1
20150376624 Gryaznov et al. Dec 2015 A1
20150376625 Oestergaard et al. Dec 2015 A1
20160002281 Mayes et al. Jan 2016 A1
20160002631 Wilton et al. Jan 2016 A1
20160002632 Wilton et al. Jan 2016 A1
20160002635 Wilton et al. Jan 2016 A1
20160017327 Rudnicki et al. Jan 2016 A1
20160024496 Bennett et al. Jan 2016 A1
20160040161 Packard et al. Feb 2016 A1
20160046939 Prakash et al. Feb 2016 A1
20160050929 Benfatti et al. Feb 2016 A1
20160050930 Benfatti et al. Feb 2016 A1
20160053256 Hung et al. Feb 2016 A1
20160068837 Chang et al. Mar 2016 A1
20160076033 Torii et al. Mar 2016 A1
20160108396 Jensen et al. Apr 2016 A1
20160122761 Prakash et al. May 2016 A1
20160128928 Fukuhara et al. May 2016 A1
20160129023 Thomas et al. May 2016 A1
20160138022 Kandimalla et al. May 2016 A1
20160159846 Prakash et al. Jun 2016 A1
20160168570 Van Deutekom et al. Jun 2016 A1
20160186175 Seth et al. Jun 2016 A1
20160186178 Radovic-Moreno et al. Jun 2016 A1
20160186185 Prakash et al. Jun 2016 A1
20160194349 Prakash et al. Jul 2016 A1
20160194636 Van Deutekom et al. Jul 2016 A1
20160214974 Schaetzer et al. Jul 2016 A1
20160230172 Rigo Aug 2016 A1
20160237108 Fraley et al. Aug 2016 A1
20160237432 Bennett et al. Aug 2016 A1
20160251653 Davidson et al. Sep 2016 A1
20160251655 Freier et al. Sep 2016 A1
20160251658 Van Deutekom et al. Sep 2016 A1
20160264964 Cancilla et al. Sep 2016 A1
20160312217 Hung et al. Oct 2016 A1
20160331835 Gemba et al. Nov 2016 A1
20160331836 Gemba et al. Nov 2016 A1
20160333349 Gemba et al. Nov 2016 A1
20160347780 Wada et al. Dec 2016 A1
20160347784 Verdine et al. Dec 2016 A1
20160355810 Van Deutekom Dec 2016 A1
20160369273 Freier Dec 2016 A1
20170009233 Wilton et al. Jan 2017 A1
20170009234 Wilton et al. Jan 2017 A1
20170029445 Shimizu et al. Feb 2017 A1
20170029457 Verdine et al. Feb 2017 A1
20170037399 Meena et al. Feb 2017 A1
20170044526 Wan et al. Feb 2017 A1
20170067050 Tuschl et al. Mar 2017 A1
20170114086 Clarke Apr 2017 A1
20170114340 Mueller et al. Apr 2017 A1
20170130224 Oestergaard et al. May 2017 A1
20170197903 Hoashi Jul 2017 A1
20170239280 Thomas et al. Aug 2017 A1
20170275621 Butler et al. Sep 2017 A1
20170327824 Oestergaard et al. Nov 2017 A1
20170349897 Rigo Dec 2017 A1
20180111958 Wada et al. Apr 2018 A1
20180216107 Frank-Kamenetsky et al. Aug 2018 A1
20180216108 Vargeese et al. Aug 2018 A1
Foreign Referenced Citations (434)
Number Date Country
102675386 Sep 2012 CN
1144279 Feb 1963 DE
01934150 Jan 1970 DE
0 002 322 Jun 1979 EP
192521 Aug 1986 EP
269258 Jun 1988 EP
0506242 Sep 1992 EP
0531447 Mar 1993 EP
0604409 Jul 1994 EP
0655088 May 1995 EP
0779893 Jun 1997 EP
0831854 Apr 1998 EP
0973945 Jan 2000 EP
1097162 May 2001 EP
1100807 May 2001 EP
1185305 Mar 2002 EP
1244682 Oct 2002 EP
1311526 May 2003 EP
1418179 May 2004 EP
1499627 Jan 2005 EP
1539188 Jun 2005 EP
1556077 Jul 2005 EP
1560840 Aug 2005 EP
1562971 Aug 2005 EP
1670810 Jun 2006 EP
1670896 Jun 2006 EP
1795536 Jun 2007 EP
1957507 Aug 2008 EP
1984381 Oct 2008 EP
2021472 Feb 2009 EP
2062980 May 2009 EP
2066684 Jun 2009 EP
2149571 Feb 2010 EP
2161038 Mar 2010 EP
2170917 Apr 2010 EP
2173760 Apr 2010 EP
2176280 Apr 2010 EP
2282744 Feb 2011 EP
2285819 Feb 2011 EP
2316967 May 2011 EP
2360166 Aug 2011 EP
1 866 319 Nov 2011 EP
2399588 Dec 2011 EP
2422819 Feb 2012 EP
2428227 Mar 2012 EP
2458005 May 2012 EP
2462153 Jun 2012 EP
2479182 Jul 2012 EP
1606407 Dec 2013 EP
14193887.8 Nov 2014 EP
14198167.0 Dec 2014 EP
15182401.8 Aug 2015 EP
15191074.2 Oct 2015 EP
15191075.9 Oct 2015 EP
15191076.7 Oct 2015 EP
2982758 Feb 2016 EP
2125852 Apr 2016 EP
2370451 Nov 2016 EP
2 534 262 Dec 2016 EP
1448437 Sep 1976 GB
2016273 Sep 1979 GB
H03-074398 Mar 1991 JP
3072345 Jul 2000 JP
2003238586 Aug 2003 JP
2009-190983 Aug 2009 JP
4348044 Oct 2009 JP
04348077 Oct 2009 JP
2011088935 May 2011 JP
WO-9110671 Jul 1991 WO
WO-9116331 Oct 1991 WO
WO-9117755 Nov 1991 WO
WO-9203452 Mar 1992 WO
WO-9220822 Nov 1992 WO
WO-9220823 Nov 1992 WO
WO-9308296 Apr 1993 WO
WO-9417093 Aug 1994 WO
WO-9422886 Oct 1994 WO
WO-9422888 Oct 1994 WO
WO-9422890 Oct 1994 WO
WO-9602555 Feb 1996 WO
WO-9607392 Mar 1996 WO
WO-9614329 May 1996 WO
WO-9619572 Jun 1996 WO
WO-9636627 Nov 1996 WO
WO-9637504 Nov 1996 WO
WO-9639413 Dec 1996 WO
WO-9706183 Feb 1997 WO
WO-9709443 Mar 1997 WO
WO-9714710 Apr 1997 WO
WO-9747637 Dec 1997 WO
WO-9802582 Jan 1998 WO
WO-9803542 Jan 1998 WO
WO-9807734 Feb 1998 WO
WO-98016535 Apr 1998 WO
WO-9818810 May 1998 WO
WO-9839334 Sep 1998 WO
WO-9846794 Oct 1998 WO
WO-9853801 Dec 1998 WO
WO-9900377 Jan 1999 WO
WO-9905160 Feb 1999 WO
WO-9912034 Mar 1999 WO
WO-9956755 Nov 1999 WO
WO-9958118 Nov 1999 WO
WO-0000499 Jan 2000 WO
WO-0004034 Jan 2000 WO
WO-0006588 Feb 2000 WO
WO-0009159 Feb 2000 WO
WO-0023444 Apr 2000 WO
WO-0031110 Jun 2000 WO
WO-0037658 Jun 2000 WO
WO-0055179 Sep 2000 WO
WO-0058329 Oct 2000 WO
WO-0076554 Dec 2000 WO
WO-0102415 Jan 2001 WO
WO-01022990 Apr 2001 WO
WO-0127126 Apr 2001 WO
WO-0140515 Jun 2001 WO
WO-0149701 Jul 2001 WO
WO-0164702 Sep 2001 WO
WO-2001068663 Sep 2001 WO
WO-0181303 Nov 2001 WO
WO-0185751 Nov 2001 WO
WO-0188198 Nov 2001 WO
WO-0212263 Feb 2002 WO
WO-0214340 Feb 2002 WO
WO-0215410 Feb 2002 WO
WO-0220544 Mar 2002 WO
WO-0222635 Mar 2002 WO
WO-0224906 Mar 2002 WO
WO-0232450 Apr 2002 WO
WO-0257425 Jul 2002 WO
WO-2002051716 Jul 2002 WO
WO-0297134 Dec 2002 WO
WO-02099317 Dec 2002 WO
WO-03002065 Jan 2003 WO
WO-03004602 Jan 2003 WO
WO-03011887 Feb 2003 WO
WO-03012057 Feb 2003 WO
WO-03014306 Feb 2003 WO
WO-03014307 Feb 2003 WO
WO-03018600 Mar 2003 WO
WO-03066633 Aug 2003 WO
WO-2003071001 Aug 2003 WO
WO-2003072757 Sep 2003 WO
WO-2003073989 Sep 2003 WO
WO-03097662 Nov 2003 WO
WO-03099840 Dec 2003 WO
WO-03100017 Dec 2003 WO
WO-03106477 Dec 2003 WO
WO-2004000351 Dec 2003 WO
WO-2004003228 Jan 2004 WO
WO-2004007718 Jan 2004 WO
WO-2004014312 Feb 2004 WO
WO-2004014933 Feb 2004 WO
WO-2004016805 Feb 2004 WO
WO-2004010956 Feb 2004 WO
WO-2004024919 Mar 2004 WO
WO-2004039829 May 2004 WO
WO-2004041889 May 2004 WO
WO-2004044134 May 2004 WO
WO-2004044136 May 2004 WO
WO-2004044141 May 2004 WO
WO-2004044181 May 2004 WO
WO-2004048522 Jun 2004 WO
WO-2004055162 Jul 2004 WO
WO-2004080466 Sep 2004 WO
WO-2004083432 Sep 2004 WO
WO-2004083446 Sep 2004 WO
WO-2004085454 Oct 2004 WO
WO-2004096233 Nov 2004 WO
WO-2004096235 Nov 2004 WO
WO-2004096286 Nov 2004 WO
WO-2004093783 Nov 2004 WO
WO-2005002626 Jan 2005 WO
WO-2005000201 Jan 2005 WO
WO-2005005599 Jan 2005 WO
WO-2005014609 Feb 2005 WO
WO-2005013901 Feb 2005 WO
WO-2005019236 Mar 2005 WO
WO-2005019237 Mar 2005 WO
WO-2005021568 Mar 2005 WO
WO-2005023828 Mar 2005 WO
WO-2005028494 Mar 2005 WO
WO-2005019418 Mar 2005 WO
WO-2005023825 Mar 2005 WO
WO-2005023995 Mar 2005 WO
WO-2005039630 May 2005 WO
WO-2005042018 May 2005 WO
WO-2005042716 May 2005 WO
WO-2005040180 May 2005 WO
WO-2005063976 Jul 2005 WO
WO-2005070859 Aug 2005 WO
WO-2005085272 Sep 2005 WO
WO-2005092909 Oct 2005 WO
WO-2006000057 Jan 2006 WO
WO-2006020676 Feb 2006 WO
WO-2006022323 Mar 2006 WO
WO-2006029258 Mar 2006 WO
WO-2006031267 Mar 2006 WO
WO-2006031461 Mar 2006 WO
WO-2006044531 Apr 2006 WO
WO-2006049454 May 2006 WO
WO-2006050501 May 2006 WO
WO-2006053861 May 2006 WO
WO-2006065751 Jun 2006 WO
WO-2006066260 Jun 2006 WO
WO-2006070284 Jul 2006 WO
WO-2006080596 Aug 2006 WO
WO-2006091915 Aug 2006 WO
WO-2006117400 Nov 2006 WO
WO-2006121960 Nov 2006 WO
WO-2007002904 Jan 2007 WO
WO-2007005941 Jan 2007 WO
WO-2007027775 Mar 2007 WO
WO-2007041045 Apr 2007 WO
WO-2007051045 May 2007 WO
WO-2007059041 May 2007 WO
WO-2007064291 Jun 2007 WO
WO-2007070598 Jun 2007 WO
WO-2007064954 Jun 2007 WO
WO-2007089584 Aug 2007 WO
WO-2007089611 Aug 2007 WO
WO-2007090071 Aug 2007 WO
WO-2007095316 Aug 2007 WO
WO-2007131232 Nov 2007 WO
WO-2007131237 Nov 2007 WO
WO-2007131238 Nov 2007 WO
WO-2007134014 Nov 2007 WO
WO-2007136988 Nov 2007 WO
WO-2007139190 Dec 2007 WO
WO-2007143315 Dec 2007 WO
WO-2007143316 Dec 2007 WO
WO-2007143317 Dec 2007 WO
WO-2007146511 Dec 2007 WO
WO-2008005562 Jan 2008 WO
WO-2008008476 Jan 2008 WO
WO-2008021136 Feb 2008 WO
WO-2008017081 Feb 2008 WO
WO-2008049085 Apr 2008 WO
WO-2008051763 May 2008 WO
WO-2008068638 Jun 2008 WO
WO-2008073959 Jun 2008 WO
WO-2008066776 Jun 2008 WO
WO-2008098104 Aug 2008 WO
WO-2008118883 Oct 2008 WO
WO-2008139262 Nov 2008 WO
WO-2008148801 Dec 2008 WO
WO-2008151833 Dec 2008 WO
WO-2009001097 Dec 2008 WO
WO-2009007855 Jan 2009 WO
WO-2009014237 Jan 2009 WO
WO-2009046141 Apr 2009 WO
WO-2009086264 Jul 2009 WO
WO-2009089659 Jul 2009 WO
WO-2009098197 Aug 2009 WO
WO-2009117589 Sep 2009 WO
WO-2009124238 Oct 2009 WO
WO-2009135322 Nov 2009 WO
WO-2009143387 Nov 2009 WO
WO-2009143390 Nov 2009 WO
WO-2009143391 Nov 2009 WO
WO-2009143463 Nov 2009 WO
WO-2009146123 Dec 2009 WO
WO-2009148605 Dec 2009 WO
WO-2010003133 Jan 2010 WO
WO-2010030858 Mar 2010 WO
WO-2010039543 Apr 2010 WO
WO-2010042636 Apr 2010 WO
WO-2010048549 Apr 2010 WO
WO-2010048585 Apr 2010 WO
WO-2010036696 Apr 2010 WO
WO-2010036698 Apr 2010 WO
WO-2010048552 Apr 2010 WO
WO-2010064146 Jun 2010 WO
WO-2010072831 Jul 2010 WO
WO-2010080953 Jul 2010 WO
WO-2010096650 Aug 2010 WO
WO-2010091301 Aug 2010 WO
WO-2010107838 Sep 2010 WO
WO-2010113937 Oct 2010 WO
WO-2010118263 Oct 2010 WO
WO-2010120262 Oct 2010 WO
WO-2010129853 Nov 2010 WO
WO-2010141471 Dec 2010 WO
WO-2010146784 Dec 2010 WO
WO-2010150789 Dec 2010 WO
WO-2011005761 Jan 2011 WO
WO-2011005764 Jan 2011 WO
WO-2011005860 Jan 2011 WO
WO-2011010706 Jan 2011 WO
WO-2011015572 Feb 2011 WO
WO-2011015573 Feb 2011 WO
WO-2011017521 Feb 2011 WO
WO-2011017561 Feb 2011 WO
WO-2011034072 Mar 2011 WO
WO-2011038288 Mar 2011 WO
WO-2011045702 Apr 2011 WO
WO-2011062210 May 2011 WO
WO-2011064974 Jun 2011 WO
WO-2011085271 Jul 2011 WO
WO-2011097643 Aug 2011 WO
WO-2011097644 Aug 2011 WO
WO-2011108682 Sep 2011 WO
WO-2011133871 Oct 2011 WO
WO-2011127175 Oct 2011 WO
WO-2011127307 Oct 2011 WO
WO-2011139699 Nov 2011 WO
WO-2011139911 Nov 2011 WO
WO-2011135396 Nov 2011 WO
WO-2012030683 Mar 2012 WO
WO-2012073857 Jun 2012 WO
WO-2012092367 Jul 2012 WO
WO-2012109395 Aug 2012 WO
WO-2012151324 Nov 2012 WO
WO-2013012758 Jan 2013 WO
WO-2013013068 Jan 2013 WO
WO-2013022984 Feb 2013 WO
WO-2013022990 Feb 2013 WO
WO-2013022966 Feb 2013 WO
WO-2013022967 Feb 2013 WO
WO-2013033223 Mar 2013 WO
WO-2013030588 Mar 2013 WO
WO-2013089283 Jun 2013 WO
WO-2013138236 Sep 2013 WO
WO-2014010250 Jan 2014 WO
WO-2014010718 Jan 2014 WO
WO-2014012081 Jan 2014 WO
WO-2014025805 Feb 2014 WO
WO-2014028739 Feb 2014 WO
WO-2014059356 Apr 2014 WO
WO-2014062686 Apr 2014 WO
WO-2014062691 Apr 2014 WO
WO-2014062736 Apr 2014 WO
WO-2014067904 May 2014 WO
WO-2014069520 May 2014 WO
WO-2014076195 May 2014 WO
WO-2014076196 May 2014 WO
WO-2014080004 May 2014 WO
WO-2014070771 May 2014 WO
WO-2014099941 Jun 2014 WO
WO-2014118267 Aug 2014 WO
WO-2014118272 Aug 2014 WO
WO-2014130607 Aug 2014 WO
WO-2014132671 Sep 2014 WO
WO-2014154486 Oct 2014 WO
WO-2014154488 Oct 2014 WO
WO-2014179626 Nov 2014 WO
WO-2014188001 Nov 2014 WO
WO-2014192310 Dec 2014 WO
WO-2014203518 Dec 2014 WO
WO-2014205451 Dec 2014 WO
WO-2014207232 Dec 2014 WO
WO-2015010135 Jan 2015 WO
WO-2015017675 Feb 2015 WO
WO-2015032617 Mar 2015 WO
WO-2015051169 Apr 2015 WO
WO-2015051214 Apr 2015 WO
WO-2015051366 Apr 2015 WO
WO-2015054676 Apr 2015 WO
WO-2015057727 Apr 2015 WO
WO-2015057738 Apr 2015 WO
WO-2015070212 May 2015 WO
WO-2015071388 May 2015 WO
WO-2015089511 Jun 2015 WO
WO-2015107425 Jul 2015 WO
WO-2015108046 Jul 2015 WO
WO-2015108047 Jul 2015 WO
WO-2015108048 Jul 2015 WO
WO-2015143078 Sep 2015 WO
WO-2015168172 Nov 2015 WO
WO-2015168589 Nov 2015 WO
WO-2015171932 Nov 2015 WO
WO-2015179525 Nov 2015 WO
WO-2016011226 Jan 2016 WO
WO-2016020399 Feb 2016 WO
WO-2016021683 Feb 2016 WO
WO-2016027168 Feb 2016 WO
WO-2016037191 Mar 2016 WO
WO-2016079181 May 2016 WO
WO-2016079183 May 2016 WO
WO-2016081444 May 2016 WO
WO-2016096938 Jun 2016 WO
WO-2016102664 Jun 2016 WO
WO-2016112132 Jul 2016 WO
WO-2016126995 Aug 2016 WO
WO-2016127000 Aug 2016 WO
WO-2016127002 Aug 2016 WO
WO-2016130589 Aug 2016 WO
WO-2016130806 Aug 2016 WO
WO-2016138017 Sep 2016 WO
WO-2016141236 Sep 2016 WO
WO-2016145142 Sep 2016 WO
WO-2016154096 Sep 2016 WO
WO-2016161374 Oct 2016 WO
WO-2016164896 Oct 2016 WO
WO-2016167780 Oct 2016 WO
WO-2016168592 Oct 2016 WO
WO-2016209862 Dec 2016 WO
WO-2017004261 Jan 2017 WO
WO-2017011276 Jan 2017 WO
WO-2017011286 Jan 2017 WO
WO-2017015109 Jan 2017 WO
WO-2017015555 Jan 2017 WO
WO-2017015575 Jan 2017 WO
WO-2017019660 Feb 2017 WO
WO-2017023660 Feb 2017 WO
WO-2017032726 Mar 2017 WO
WO-2017035340 Mar 2017 WO
WO-2017040078 Mar 2017 WO
WO-2017-048620 Mar 2017 WO
WO-2017055423 Apr 2017 WO
WO-2017059411 Apr 2017 WO
WO-2017059446 Apr 2017 WO
WO-2017062862 Apr 2017 WO
WO-2017067970 Apr 2017 WO
WO-2017068087 Apr 2017 WO
WO-2017079291 May 2017 WO
WO-2017081223 May 2017 WO
WO-2017157672 Sep 2017 WO
WO-2017157899 Sep 2017 WO
WO-2017160741 Sep 2017 WO
WO-2017165489 Sep 2017 WO
WO-2017178656 Oct 2017 WO
WO-2017180835 Oct 2017 WO
WO-2017192664 Nov 2017 WO
WO-2017192679 Nov 2017 WO
WO-2017194498 Nov 2017 WO
WO-2017194664 Nov 2017 WO
WO-2017198775 Nov 2017 WO
WO-2017210647 Dec 2017 WO
WO-2017221883 Dec 2017 WO
WO-2018022473 Feb 2018 WO
WO-2018067973 Apr 2018 WO
WO-2018098264 May 2018 WO
Non-Patent Literature Citations (728)
Entry
Aaronson, J.G. et al., Rapid HATU-Mediated Solution Phase siRNA Conjugation, Bioconjugate. Chem., 22: 1723-1728 (2011).
Aartsma-Rus, A. et al., Antisense-Induced Multiexon Skipping for Duchenne Muscular Dystrophy Makes More Sense, Am. J. Hum. Genet., 74:83-92 (2004).
Aartsma-Rus, A. et al., Targeted exon skipping as a potential gene correction therapy for Duchenne muscular dystrophy, Neuromuscular Disorders, 12: S71-S77 (2002).
Aartsma-Rus, A. et al., Therapeutic antisense-induced exon skipping in cultured muscle cells from six different DMD patients, Human Molecular Genetics, 12(8):907-914 (2003).
Adams, S.P. et al., Hindered dialkylamino nucleoside phosphite reagents in the synthesis of two DNA 51-mers, Journal of the American Chemical Society, 105(3): 661-663 (1983).
Adarsh, et al., Organelle Specific Targeted Drug Delivery—A Review, International Journal of Research in Pharmaceutical and Biomedical Sciences, 2(3): 895-912 (2011).
Ager, D.J. The Peterson olefination reaction, Organic Reactions, 38:1-223 (2004).
Agrawal, S. and Kandimalla, E.R., Antisense and/or Immunostimulatory Oligonucleotide THerapeutics, Current Cancer Drug Targets, Bentham Science, 1(3): 1 page. URL: <http:www.eurekaselect.com/65087/article> [Retrieved Apr. 3, 2016].
Agrawal, S. and Tang, J.Y., GEM 91—an antisense oligonucleotide phosphorothioate as a therapeutic agent for AIDS, Antisense Research and Development, 2(4):261-266 (1992).
Agrawal, S. et al., Mixed-backbone oligonucleotides as second generation antisense oligonucleotides: In vitro and in vivo studies, Proc. Natl. Acad. Sci. USA, 94: 2620-2625 (1997).
Aldaye, F.A. et al., Assembling materials with DNA as the guide, Science, 321(5897): 1795-1799 (2008).
Aldrich Chemical Co. Catalog, 2007-2008 Issue, only p. 1719 supplied: see first full entry at col. 1 (S-methyl methanethiosulfonate), Milwaukee, WI.
Almer et al., Synthesis of Stereochemically Homogeneous Oligoribonucleoside All-Rp-Phosphorothioates by Combining H-Phosphonate Chemistry and Enzymatic Digestion, J. Chem. Soc., Chem. Commun., 1459-1460 (1994).
Almer, et al. A New Approach to Stereospecific Synthesis of P-chiral Phosphorothioates. Preparation of Diastereomeric Dithymidyl-(3′-5′) Phosphorothioates, Chem. Commun., (3): 290-1 (2004).
Almer, et al. Solid Support Synthesis of all-Rp-oligo(ribonucleoside phosphorothioate)s, Nucleic Acids Research 24(19): 3811-3820 (1996).
Almer, H. et al., Synthesis of Diribonucleoside Phosphorothioates via Sterospecific Sulfurization of H-Phosphonate Diesters, J. Org. Chem., 57(23): 6163-6169 (1992).
Altschul, S.F. et al., Basic local alignment search tool, Journal of Molecular Biology, 215(3):403-410 (1990).
Altschul, S.F. et al., Gapped BLAST and PSI-BLAST: a new generation of protein database search programs, Nucleic Acids Research, 25(17):3389-3402 (1997).
Alul, R.H. et al., Oxalyl-CPG: a labile support for synthesis of sensitive oligonucleotide, Nucleic Acids Research, 19(7):1527-1532 (1991).
Alvarez, K. et al., Photocleavable Protecting Groups as Nucleobase Protections Allowed the Solid-Phase Synthesis of Base-Sensitive SATE-Prooligonucleotides, Journal of Organic Chemistry, 64(17): 6319-6328(1999).
Amarzguioui et al., Tolerance for mutations and chemical modifications in a siRNA, Nucleic Acids Research 31(2): 589-595 (2003).
Anthony, K. et al., Exon Skipping Quantification by Quantitative Reverse-Transcription Polymerase Chain Reaction in Duchenne Muscular Dystrophy Patients Treated with the Antisense Oligomer Eteplirsen, Human Gene Therapy Methods, 23: 336-345 (2012).
Arai, K. et al., Synthesis and properties of novel 2′-O-alkoxymethyl-modified nucleic acids, Bioorganic & Medicinal Chemistry Letters, 21(21): 6285-6287 (2011).
Aristarkhova, L.N. et al., Investigation in the field of thiosulfonic acids. 28. alkyl esters of cyclopentane- and cyclohexanethiosulfonic acids, Journal of Organic Chemistry of the USSR, 6: 2454-2458 (1970).
Athyros, V.G. et al., Antisense technology for the prevention or the treatment of cardiovascular disease: the next blockbuster?, Expert Opin. Investig. Drugs, 17(7): 969-72 (2008).
Ausin, C. et al., Assesment of heat-sensitive thiophosphate protecting groups in the development of thermolytic DNA oligonucleotide prodrugs, Tetrahedron, 66(1):68-79 (2010).
Bachelin et al., Structure of a Stereoregular Phosphorothioate DNA/RNA duplex, Nat. Struct. Biol., 5(4): 271-276 (1998).
Baek, M-S. et al., In Vitro Metabolic Stabilities and Metabolism of 2′-O-(Methoxyethyl) Partially Modified Phosphorothioate Antisense Oligonucleotides in Preincubated Rat or Human Whole Liver Homogenates, Oligonucleotides, 20(6): 309-316 (2010).
Ballas, Z.K. et al., Induction of NK Activity in Murine and Human Cells by CpG Motifs in Oligodeoxynucleotides and Bacterial DNA, J. Immunoll., 57: 1840-1845 (1996).
Barber, I. et al., the Prooligonucleotides Approach I: Esterase-Mediated Reversibility of Dithymidine S-Alkyl Phosphorothiolates to Dithymidine Phosphorothioates, Bioorganic and Medicinal Chemistry Letters, 5(6):563-568 (1995).
Barber, I. et al., the Prooligonucleotides Approach II: Synthesis and stability studies of chimeric oligonucleotide models, Bioorganic and Medicinal Chemistry Letters, 5(14):1441-1444 (1995).
Barnes, P.J. and Peterson, S. Efficacy and Safety of Inhaled Corticosteroids in Asthma, Am. Rev. Respir. Dis., 148: SI-S26 (1993).
Bartz, H. et al., Poly-guanosine strings improve cellular uptake and stimulatory activity of phosphodiester CpG oligonucleotides in human leukocytes, Vaccine, 23: 148-155 (2004).
Battistini et al., Stereoselective Synthesis of Cyclic Dinucloetide Phosphorothioates, Tetrahedron, 49(5): 1115-1132 (1993).
Bayever, E. et al., Systematic administration of a phosphorothioate oligonucleotide with a sequence complementary to p53 for acute myelogenous leukemia and myelodysplastic syndrome: intial results of a phase I trial, Antisense Research Development, 3(4):383-390 (1993).
Beal, P.A. et al., Second Structural Motif for Recognition of DNA by Oligonucleotide-Directed Triple-Helix Formation, Science, 251: 1360-1363 (1991).
Beaucage, S.L. and Iyer, R.P., Advances in the Synthesis of Oligonucleotides by the Phosphoramidite Approach, Tetrahedron, 48(12):2223-2311 (1992).
Benner, S.A. and Sismour, A.M., Synthetic biology, Nature Reviews Genetics, 6(7):533-543 (2005).
Berge, S.M. et al., Pharmaceutical salts, J. Pharm. Sci., 66(1):1-19 (1997).
Besch, R. et al, Specific Inhibition of ICAM-1 Expression Mediated by Gene Targeting with Triplex-forming Oligonucleotides, J. Biol. Chem., 277(26): 32473-32479 (2002).
Birts, C.N. et a., Transcription of Click-Linked DNA un Human Cells, Angew. Chem. Int. Ed., 53:2362-2365 (2014).
Bisbal, C. and Silverman, R.H., Diverse functions of RNase L and implication in pathology, Biochimie, 89(6-7):789-798 (2007).
Blade, H. et al., Modular Synthesis of Constrained Ethyl (cEt) Purine and Pyrimidine Nucleosides, J. Org. Chem., 80: 5337-5343 (2015).
Block, E. et al., Allium Chemistry: Synthesis and Sigmatropic Rearrangements of Alk(en)yl 1-Propenyl Disulfide S-Oxides from Cut Onion and Garlic, Journal of the Ameican Chemical Society, 118(12): 2799-2810 (1996).
Block, S.S. and Weidner, J.P, Vibrational Behavior and Structure of Disulfide Dioxides (Thiolsulfonates), Applied spectroscopy, 20(2): 73-79 (1966).
Bobkov, G.V. et al., Phosphoramidite building blocks for efficient incorporation of 2′-O-aminoethoxy(and propoxy)methyl nucleosides into oligonucleotides, Tetrahedron, 64: 6238-6251 (2008).
Bock, L.C. et al., Selections of single-stranded DNA molecules that bind and inhibit human thrombin, Nature, 355: 564-566 (1992).
Boczkowska, M. et al., Stereodefined Phosphorothioate Analogues of DNA: Relative Thermodynamic Stability of the Model PS-DNA/DNA and PS-DNA/RNA complexes, Biochemistry, 41: 12483-12487 (2002).
Bode, C. et al. CpG DNA as a vaccine adjuvant, Expert Rev. Vaccines, 10(4): 499-511 (2011).
Bodor, N. et al., A convenient synthesis of (acyloxy)alkyl .alpha.-ethers of phenols, The Journal of Organic Chemistry, 48(26):5280-5284 (1983).
Bohringer, M. et al., Why Pentose and not Hexose Nucleic Acids? Part II: Oligonucleotides of 2′3′-dideoxy-β-d-glucopyranosyl (‘homo-DNA’) production, Helvetica Chimica Acta, 75:1416-1477 (1992).
Bologna, J. et al., Uptake and Quantification of Intracellular Concentration of Lipophilic Pro-Oligonucleotides in HeLa Cells, Antisense and Nucleic Acid Drug Development, 12(1):33-41 (2002).
Bonora, G.M. et al., Large scale, liquid phase synthesis of oligonucleotides by the phosphoramidite approach, Nucleic Acids Research, 21(5): 1213-1217 (1993).
Boudreau, R.L. et al., Nonallele-specific silencing of mutant and wild-type huntingtin demonstrates therapeutic efficacy in Huntington's disease mice, 17(6): 1053-1063 (2009).
Braasch et al., RNA Interference in Mammalian Cells by Chemically-Modified RNA, Biochemistry 42(26): 7967-7975 (2003).
Brill, W. et al., Thioalkylation of Nucleoside-H-Phosphonates and Its Application to Solid Phase Synthesis of Oligonucleotides, Tetrahedron Letters, 36(5):703-706 (1995).
Brooks, P.C. et al., Insulin-like Growth Factor Receptor Cooperates with Integrin ανβ5 to Promote Tumor Cell Dissemination in Vivo, The Journal of Clinical Investigation, 99(6):1390-1398 (1997).
Brown, J.W.S. and Simpson, C.G., Splice Site Selection in Plant Pre-mRNA Splicing, Ann. Rev. Plant Physiol. Plant Mol. Biol., 49: 77-95 (1998).
Bumcrot, D et al., RNAi therapeutics: a potential new class of pharmaceutical drugs, Nat. Chem. Biol., 2: 711-9 (2006).
Bundgaard, H., (C) Means to Enhance Penetration. (1) Prodrugs as a means to improve the delivery of peptide drugs, Advanced Drug Delivery Reviews, 8:1-38 (1992).
Bundgaard, H., Design and Application of Prodrugs, A Textbook of Drug Design and Development, Edited by Krogsgaard-Larsen, P. and Bundgaard, H., Chapter 5: 113-191 (1991).
Bundgaard, H., Design of Prodrugs, Elsevier, 7-9 and 21-24 (Chapter 1) (1985).
Bunnell. B.A. et al., Targeted Delivery of Antisense Oligonucleotides by Molecular Conjugates, Somatic Cell and Molecular Genetics, 18(6):559-569 (1992).
Burgers et al., Absolute configuration of the diastereomers of adenosine 5′-O-(1-thiaotriphosphate): Consequences for the stereochemistry of polymerization by DNA-dependent RNA polymerase from Escherichia coli, Proceedings of the National Academy of Sciences of the United States of America 75(10): 4798-4800 (1978).
Burgers, P. M. J. et al., Stereochemistry of Hydrolysis by Snake Venom Phosphodiesterase, J. Biol. Chem., 254(16): 7476-7478 (1979).
Burgers, P.M.J. and Eckstein, F., A Study of the Mechanism of DNA Polymerase I from Escherichia coli with Diastereomeric Phosphorothioate Analogs of Deoxyadenosine Triphosphate, J. Biol. Chem., 254(15): 6889-6893 (1979).
Burgers, P.M.J. and Eckstein, F., Diastereomers of 5′-O-adenosyl 3′-O-uridyl phosphorothioate: chemical synthesis and enzymatic properties, Biochemistry, 18: 592-596 (1979).
Campbell, J. et al., Hybrid polymer/MOF membranes for Organic Solvent Nanofiltration (OSN): Chemical modification and the quest for perfection, Journal of Membrance Science, 503: 166-176 (2016).
Cankurtaran, E.S. et al., Clinical Experience with Risperidone and Memantine in the Treatment of Huntington's Disease, Journal of the National Medical Association, 98(8): 1353-1355 (2006).
Carbone, G.M. et al., Selective inhibition of transcription of the Ets2 gene in prostate cancer cells by a triplex-forming oligonucleotide, Nucl. Acid. Res., 31: 833-843 (2003).
Carrillo, H., and Lipman, D.J., The multiple sequence alignment problem in biology, SIAM J. Appl. Math., 48:1073-1082 (1988).
CAS Registry No. 1225524-67-3; STN Entry Date May 28, 2010; α-[(2-methylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1225524-68-4; STN Entry Date May 28, 2010; α-[(4-methylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1225545-00-5; STN Entry Date May 28, 2010; α-[(2,4,6-trimethylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1225554-20-0; STN Entry Date May 28, 2010; α-[(4-ethylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1225594-74-0; STN Entry Date May 28, 2010; α-[(2-chloro-6-fluorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1225682-42-7; STN Entry Date May 30, 2010; α-[(4-chlorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226037-41-7; STN Entry Date May 30, 2010; α-[(3-chlorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226118-97-3; STN Entry Date May 30, 2010; α-[(3-bromophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226119-02-3; STN Entry Date May 30, 2010; α-[(4-bromophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226146-65-1; STN Entry Date May 30, 2010; α-[(2,4-dimethylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226160-20-8; STN Entry Date May 30, 2010; α-[(2,5-dimethylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226178-36-4; STN Entry Date May 30, 2010; α-[(2-fluorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226188-06-2; STN Entry Date May 30, 2010; α-[[4-(1-methylethyl)phenyl]methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226204-20-1; STN Entry Date May 30, 2010; α-[(3-methylphenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226231-44-2; STN Entry Date May 30, 2010; α-[(2-chlorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226352-28-8; STN Entry Date May 30, 2010; α-[(2,4-dichlorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226352-38-0; STN Entry Date May 30, 2010; α-[(3,4-dichlorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1226413-27-9; STN Entry Date May 30, 2010; α-(phenylmethyl)- 2-Pyrrolidinemethanol.
CAS Registry No. 1226419-15-3; STN Entry Date May 30, 2010; α-[(4-fluorophenyl)methyl]-2-Pyrrolidinemethanol.
CAS Registry No. 1263282-82-1 ; STN Entry Date Feb. 21, 2011; (S)-[(diphenyl)methyl]-2-Pyrrolidinemethanol.
CAS RN 78-96-6, Entered STN: Nov. 16, 1984.
Chak, L-L, and Okamura, K., Argonaute-dependent small RNAs derived from single-stranded, non-structured precursors, Frontiers in Genetics, 5(172): 1-15 (2014).
Chan, J.H.P. et al., Antisense Oligonucleotides: From Design to Therapeutic Application, Clinical and Experimental Pharmacology and Physiology, 33: 544-540 (2006).
Chang, W. et al., Systematic chemical modifications of single stranded siRNAs significantly improved CTNNB1 mRNA silencing, Bioorg. Med. Chem. Lett., 1-5 (2016), http://dx.doi.org/10.1016/j.bmcl.2016.07.064.
Chappell, C. et al., Involvement of human polynucleotide kinase in double-strand break repair by non-homologous end joining, The EMBO Journal, 21(11): 2827-2832 (2002).
Chatgilialoglu, C. and Snieckus, V., Chemical Synthesis: Gnosis to Prognosis, Kluwer Academic, 293-340 (1996).
Check, E., RNA interference: hitting the on switch, Nature, 448(7156): 855-858 (2007).
Cheloufi, S. et al., A Dicer-independent miRNA biogenesis pathway that requires Ago catalysis, Nature, 465(7298): 584-589 (2010).
Chen, B. and Bartlett, M., A One-Step Solid Phase Extraction Method for Bioanalysis of a Phosphorothioate Oligonucleotide and Its 3′ n-1 Metabolite from Rat Plasma by uHPLC-MS/MS, The AAPS Journal, 14(4): 772-780 (2012).
Chiu, Y. and Rana, T.M., siRNA function in RNAi: A chemical modification analysis, RNA, 9(9):1034-1048 (2003).
Chmielewski, M.K. and Markiewicz, W.T., Novel Method of Synthesis of 5″-Phosphate 2′-O-ribosyl-ribonucleosides and Their 3′-Phosphoramidites, Molecules, 18:14780-14796 (2013).
Cieslak, J. et al., 31P NMR Study of the Desulfurization of Oligonucleoside Phosphorothioates Effected by “Aged” Trichloroacetic Acid Solutions, J. Org. Chem., 70: 3303-3306 (2005).
Cieslak, J. et al., Thermolytic 4-methylthio-1-butyl group for phosphate/thiophosphate protection in solid-phase synthesis of DNA oligonucleotides, Journal of Organic Chemistry, 69(7):2509-2515 (2004).
Clark, J.H, Flouride IOn as a Base in Organic Synthesis, Chemical Reviews, 1980 American Chemical Society 80(5): 429-452 (1980).
Communication Relating to the Results of the Partial International Search of PCT/IB2015/000395, Annex to Form PCT/ISA/206, 3 pages (dated Aug. 24, 2015).
Conway, N., The introduction of reporter groups at multiple and/or specific sites in DNA containing phosphorothioate diesters, Nucleic Acids Research, 43-44 (1989).
Cooney, M., et al., Site-Specific Oligonucleotide Binding Represses Transcription of the Human c-myc Gene in Vitro, Science, 241: 456-459 (1988).
Cosstick, R. and Eckstein, F., Synthesis of d(GC) and d(CG) Octamers Containing Alternating Phosphorothioate Linkages: Effect of the Phosphorothioate Group on the B-Z Transition, Biochemistry, 24: 3630-3638 (1985).
Coughlin, J.E. et al., Orally bioavailable anti-HBV dinucleotide acyloxyalkyl prodrugs, Bioorganic and Medicinal Chemistry Letters, 20(5):1783-1786 (2010).
Cox, J.R. and Ramsay, O.B., Mechanisms of Nucleophilic Substitution in Phosphate Esters, Chemical Reviews, 64(4): 317-352, (1964).
Crary, S.M. et al., Specific phosphorothioate substitutions probe the active site of Bacilus subtilis ribonuclease P, RNA, 8:933-947 (2002).
Crooke, S.T. and Geary, R.S. Clinical pharmacological properties of mipomersen (Kynamro), a second generation antisense inhibitor of apolipoprotein B, Br. J. Clin. Pharmacol., 76: 269-276 (2012).
Crooke, S.T., Antisense Strategies, Current Molecular Medicine, 4: 465-487 (2004).
Crooke, S.T., Molecular mechanisms of action of antisense drugs, Biochemica et Biophysica Acta, 1489: 31-44 (1999).
Crooke, S.T., Progress in Antisense Technology , Annu. Rev. Med., 55: 61-95 (2004).
Cullen, K.A. et al., Ambulatory surgery in the United States, 2006, National Health Statistics Reports, 11: 1-28 (Jan. 28, 2009—Revised Sep. 4, 2009).
Current Protocols in Nucleic Acid Chemistry, Edited by Beaucage, S.L. et al., Chapter 2: Protection of Nucleosides for Oligonucleotide Synthesis, 2.0.1.-2.16.31 (2012).
Davis, B.G. et al., Altering the specificity of subtilisin bacillus lentus through the introduction of positive charge at single amino acid sites, Bioorganic & Medicinal Chemistry, 7(11): 2303-2311 (1999).
De Koning, M.G. et al., Simple and Efficient Solution-Phase Synthesis of Oligonucleotides Using Extractive Work-Up, Organic Process Research & Developmen, 10: 1238-1245 (2006).
Dejesus-Hernandez, M. et al., Expanded GGGGCC hexanucleotide repeat in non-coding region of C9ORF72 causes chromosome 9p-linked frontotemporal dementia and amyotrophic lateral sclerosis, Neuron, 72(2): 245-256 (2011).
Deleavey, G.F. and Damha, M.J., Designing chemically modified oligonucleotides for targeted gene silencing. Chem. Biol., 19: 937-54 (2012).
Dellinger, D.J. et al., Streamlined Process for the Chemical Synthesis of RNA Using 2′-O-Thionocarbamate-Protected Nucleoside Phosphoramidites in the Solid Phase, J. Am. Chem. Soc., 133: 11540-11556 (2011).
Devereux, J. et al., A comprehensive set of sequence analysis programs for the VAX, Nucleic Acids Research, 12(1):387-395 (1984).
Dias, N. and Stein, C.A., Antisense Oligonucleotides: Basic Concepts and Mechanisms, Molecular Cancer Therapeutics, 1: 347-355 (2002).
Dietz, G.P.H. et al., Delivery of bioactive molecules into the cell: the Trojan horse approach, Molecular and Cellular Neuroscience, 27(2): 85-131 (2004).
Dikfidan, A. et al., RNA Specificity and Regulation of Catalysis in the Eukaryotic Polynucleotide Kinase Clp1, Molecular Cell, 54: 975-986 (2014).
Djukanovic, R. et al., Mucosal Inflammation in Asthma, Am. Rev. Respir. Dis., 142: 434-457 (1990).
Documents submitted to and/or received from the United States Securities and Exchange Commission; downloaded from EDGAR (Feb. 2, 2015 to Dec. 10, 2015).
Documents submitted to and/or received from the United States Securities and Exchange Commission; downloaded from EDGAR (Dec. 17, 2015 to Oct. 4, 2016).
Documents submitted to and/or received from the United States Securities and Exchange Commission; downloaded from EDGAR (Nov. 9, 2016 to May 10, 2017).
Donnelly, C.J. et al., RNA Toxicity from the ALS/FTD C90RF72 Expansion Is Mitigated by antisense Intervention, Neuron, 80:415-428 (2013).
Dorman et al., Synthesis of Oligodeoxynucleotides and Oligodeoxynucleotide Analogs using Phosphoramidite Intermediates, Tetrahedron, 40(1):95-102 (1984).
Dua, P. et al., Patents on SELEX and therapeutic aptamers, Recent Patents on DNA & Gene Sequences, 2(3):172-186 (2008).
Eaton, W.A. et al., Submillisecond kinetics of protein folding, Curr. Opin. Chem. Biol., 1:10-14 (1997).
Eckstein, F. et al., Stereochemistry of polymerization by DNA-dependent RNA-polymerase from Escherichia coli: an investigation with a diastereomeric ATP-analogue, Proc. Natl. Acad. Sci. USA, 73: 2987-90 (1976).
Eckstein, F. Phosphorothioates, Essential Components of Therapeutic Oligonucleotides, Nucleic Acid Therapeutics, 24(6): 374-387 (2014).
Eckstein, F., Oligonucleotides and Analogues A Practical Approach, IRL Press, 1-24 (1991).
Efimov, V.A. et al., Rapid synthesis of long-chain deoxyribooligonucleotides by the N-methylimidazolide phosphotriester method, Nucleic Acids Research, 11(23): 8369-8387 (1983).
Egholm, M. et al., PNA hybridizes to complementary oligonucleotides obeying the Watson-Crick hydrogen-bonding rules, Nature, 365, 566-568 (1993).
Egli, M. et al., Crystal structure of homo-DNA and nature's choice of pentose over hexose in the genetic system, Journal of the American Chemical Society, 128(33):10847-56 (2006).
Egli, M. et al., Probing the Influence of Stereoelectronic Effects on the Biophysical Properties of Oligonucleotides: Comprehensive Analysis of the RNA Affinity, Nuclease Resistance, and Crystal Structure of Ten 2′-0-Ribonucleic Acid Modifications, Biochemistry, 44: 9045-9057 (2005).
El Harchaoui, K. et al., Current and future pharmacologic options for the management of patients unable to achieve low-density lipoprotein-cholesterol goals with statins, Am. J. Cardiovasc. Drugs, 8(4): 233-242 (2008).
El-Sagheer, A.H. and Brown, T., Efficient RNA synthesis by in vitro transcription of a triazole-modified DNA template, Chem. Commun., 47(44):12057-12058 (2011).
El-Sagheer, A.H. and Brown, T., New strategy for the synthesis of chemically modified RNA constructs exemplified by hairpin and hammerhead ribozymes, PNAS, 107(35):15329-15334 (2010).
El-Sagheer, A.H. et al., Biocompatible artificial DNA linker that is read through by DNA polymerases and is functional in Escherichia coli, PNAS, 108(28):11338-11343 (2011).
Elbashir, S.M. et al., Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells, Nature, 411: 494-498 (2001).
Elbashir, S.M. et al., Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate, The EMBO Journal, 20(23): 6877-6888 (2001).
Ellington, A.D. and Szostak, J.W., In vitro selection of RNA molecules that bind specific ligands, Nature, 346: 818-822 (1990).
Engelhardt, J.A. et al., Scientific and Regulatory Policy Committee Points-to-consider Paper: Drug-induced Vascular Injury Associated with Nonsmall Molecule Therapeutics in Preclinical Development: Part 2. Antisense Oligonucleotides, Toxicologic Pathology, XX: 1-10 (2015).
Epton, R., Innovation and Perspectives in Solid Phase Synthesis, Peptides, Proteins and Nucleic Acids, 21:157-162 (1994).
Erler, W. et al., Patient Advisory Board Meeting, Wave Life Sciences, London, 46 pages (Mar. 2, 2017).
Erler, W., Stereopure Exon 51-Skipping Oligonucleotide as a Potential Disease-Modifying Therapy for Duchenne Muscular Dystrophy, WAVE Life Sciences, 10 pages (2017).
Eschenmoser, A. et al., Why pentose- and not hexose-nucleic acids? Introduction to the problem, conformational analysis of oligonucleotide single strands containing 2′, 3′-dideoxyglucopyranosyl building blocks (‘homo-DNA’), and reflections on the conformation of A- and B-DNA, Helvetica Chimica Acta, 75:218-259 (1992).
Eschenmoser, A., Chemical etiology of nucleic acid structure, Science, 284(5423):2118-24 (1999).
Eschenmoser, A., Towards a Chemical Etiology of the Natural Nucleic Acids' Structure, Chemical Synthesis, Edited by Chatgilialoglu, C. and Snieckus, V., Kluwer Academic Publishers, 293-340 (1996).
Ewles, M. et al, Quantification of oligonucleotides by LC-MS/MS: the challenges of quantifying a phosphorothioate oligonucleotide and multiple metabolites, Bioanalysis, 6(4), 447-464 (2014).
Exiqon, Locked Nucleic Acid (LNA), Custom Oligonucleotides for RNA and DNA Research, 16 pages (Aug. 2009).
Famulok, M. Oligonucleotide aptamers that recognize small molecules, Curr. Opin. Struct. Biol., 9: 324-329 (1999).
Fearon, K. et al., Phosphorothioate oligodeoxynucleotides: large-scale synthesis and analysis, impurity characterization, and the effect of phosphorus stereochemistry, Oligonucleotides as Therapeutic Agents, Ciba Found. Symp. 209: 19-31 (1997).
Fendrich et al., Determination of the Absolute P-configuration of a Phthalidyl└ Phosphonate Thymidine-Thymidine Dimer, Nucleosides Nucleotides Nucleic Acids., 22(5-8): 1127-1129 (2003).
Ferreira, F. et al., Lewis acid deprotection of silyl-protected oligonucleotides and base-sensitive oligonucleotide analogues, Tetrahedron Letters, 45(33):6287-6290 (2004).
File Registry on STN, RN 18217-60-2, Entered STN: Nov. 16, 1984.
File Registry on STN, RN 871246-91-2, Entered STN: Jan. 5, 2006.
Fire, A. et al., Potent and specific RNA interference by double-stranded RNA in Caenorhadbditis elegans, Nature, 391: 806-811 (1998).
Forster, A.G. and Symons, R.H. Self-cleavage of plus and minus RNAs of a virusoid and a structural model for the active sites, Cell, 49(2): 211-220 (1987).
Forster, A.G. and Symons, R.H. Self-Cleavage of Virusoid RNA is performed by the Proposed 55-Nucleotide Active Site, Cell, 50: 9-16 (1987).
Frank-Kamenetsky, M. et al., Therapeutic RNAi targeting PCSK9 acutely lowers plasma cholesterol in rodents and LDL cholesterol in nonhuman primates. Proc. Natl. Acad. Sci. USA., 105(33): 11915-11920 (2008).
Frazier, K. et al., Potential Mechanisms of vascular toxicity in Monkeys with antisense oligonucleotides, TIDES oligo conference, 1-25 (May 15, 2014).
Frazier, K.S. Antisense Oligonucleotide Therapies: The Promise and the Challenges from a Toxicologic Pathologist's Perspective, Toxicology Pathology, 43: 78-89 (2015).
Frederiksen, J.K. et al., Separation of RNA Phosphorothioate Oligonucleotides by HPLC, Methods of Enzymology, 468:289-309 (2009).
Freier, S.M. et al., Improved free-energy parameters for predictions of RNA duplex stability, Proc. Nat. Acad. Sci. USA, 83: 9373-9377 (1986).
Freschauf, G., Identification of Small Molecule Inhibitors of the Human DNA Repair Enzyme Polynucleotide Kinase/Phosphatase, Master of Science in Experimental Oncology Thesis, University of Alberta, 155 pages (2011).
Froehler, B.C. et al., Synthesis of DNA via deoxynucleoside H-phosphonate intermediates, Nucleic Acids Research, 14(13): 5399-5407 (1986).
Fujii et al., Acylphosphonates. 5.1A new method for stereospecific generation of phosphorothioate via aroylphosphonate intermediate, Tetrahedron Letters, 27(8): 935-938 (1986).
Fujii et al., Acylphosphonates. 7.1 A New Method for Stereospecific and Stereoselective Generation of Dideoxyribonucleoside Phosphorothioates via the Acylphosphonate Intermediates, Tetrahedron, 43: 3395-3407 (1987).
Gaffney, P.R.J. et al., Liquid-Phase Synthesis of 2′-Methyl-RNA on a Homostar Support through Organic-Solvent Nanofiltration, Chem. Eur. J., 21:1-10 (2015).
Gallier, F. et al., 5′,6′-Nucleoside Phosphonate Analogues Architecture: Synthesis and Comparative Evaluation towards Metabolic Enzymes, Chem Med Chem, 6: 1094-1106 (2011).
Ganguly, A.K. et al., Structure of Halomicin B, J.C.S. Chem. Comm., 395-396 (1974).
Garegg, P.J. et al., Nucleoside H-Phosphonates. III. Chemical Synthesis of Oligodeoxyribonucleotides by the Hydrogenphosphonate Approach, Tetrahedron Letters, 27(34): 4051-4054 (1986).
Gauglitz, G.G. et al., Hypertrophic Scarring and Keloids: Pathomechanisms and Current Emerging Treatment Strategies, Mol. Med., 17(1-2): 113-125 (2011).
Giacometti, R.D. et al., Design, synthesis, and duplex-stabilizing properties of conformationally constrained tricyclic analogues of LNA, Org. Biomol. Chem., 14: 2034-2040 (2016).
Gijsen, H.J.M et al., Development of two diastereoselective rougtes towards trans-4-aminomethyl-piperidin-3-o1 building blocks, Tetrahedron 64(10): 2456-2464 (2008).
Goraczmiak, R. et al., Gene silencing by synthetic U1 Adaptors, Nature Biotechnology 27(3): 257-263 (2008).
Gosselin, G. et al., New insights regarding the potential of the pronucleotide approach in antiviral chemotherapy, 43(1):195-208 (1996).
Gough, G.R. et al., Recovery and recycling of synthetic units in the construction of oligodeoxyribonucleotides on solid supports, Tetrahedron Letters, 22(42): 4177-4180 (1981).
Gould, W.A. et al., Pyrrolidines. IX. 3-Aryl-3-pyrrolidinols, Journal of Medicinal Chemistry, 7(1): 60-67 (1964).
Graham, M.J. et al., Antisense inhibition of proprotein convertase subtilisin/kexin type 9 reduces serum LDL in hyperlipidemic mice, J. Lipid Res., 48(4): 763-767 (2007).
Grajkowski, A. et al., Design and Development of Thermolytic DNA Oligonucleotide Prodrugs, Annals of the New York Academy of Sciences, 1058:26-38 (2005).
Grajkowski, A. et al., Solid-Phase Synthesis of Thermolytic DNA Oligonucleotides Functionalized with a Single 4-Hydroxy-1-butyl or 4-Phosphato-/Thiophosphato-1-butyl Thiophosphate Protecting Group, Journal of Organic Chemistry, 72(3): 805-815 (2007).
Grajkowski, A. et al., Thermolytic CpG-containing DNA oligonucleotides as potential immunotherapeutic prodrugs, Nucleic Acids Research, 33(11):3550-3560 (2005).
Green, L.S. et al., Inhibitory DNA Ligands to Platelet-Derived Growth Factor B-Chain, Biochemistry, 35: 14413-14424 (1996).
Green, L.S. et al., Nuclease-resistant nucleic acid ligands to vascular permeability factor/vascular endothelial growth factor, Chem. Biol., 2(10): 683-695 (1995).
Griffiths-Jones, S. et al., miRBase: microRIVA sequences, targets and gene nomenclature, Nucleic Acids Research, 34 (Database Issue): D140-D144 (2006).
Griffiths-Jones, S. The microRNA Registry, Nucleic Acids Research, 32 (Database Issue): D109-D111 (2004).
Groebke, K. et al., Why pentose and not hexose nucleic acids? Part V. Purine-purine pairing in homo-DNA: guanine, isoguanine, 2,6-diaminopurine and xanthine. Helvetica Chimica Acta. 81: 375-474 (1998).
Gryaznov, S. and, Chen, J.-K., Oligodeoxyribonucleotide N3′4P5′ Phosphoramidates: Synthesis and Hybridization Properties, J. Am. Chem. Soc., 116: 3143-3144 (1994).
Gude, L. et al., Mapping Targetable Sites on Human Telomerase RNA Pseudoknot/Template Domain Using 2′-OMe RNA-interacting Polynucleotide (RIPtide) Microarrays, J. Biol. Chem., 287(22): 18843-18853 (2012).
Guerciolini, R., Allele-selective Silencing of Mutant Huntingtin by Stereopure Oligonucleotides, WAVE Life Sciences, Huntington's Disease Society of America, HDSA Presentation 2016 (Jun. 3, 2016).
Guerlavais-Dagland, T et al., Fluoride-labile protecting groups for the synthesis of base-sensitive methyl-SATE oligonucleotide prodrugs, European Journal of Organic Chemistry, 2003(12):2327-2335 (2003).
Guga et al., Oxathiaphospholane Approach to the Synthesis of P-Chiral, Isotopomeric Deoxy(ribonucleoside phosphorothioate)s and Phosphates Labeled with an Oxygen Isotope. Angew Chem., 113(3): 630-633 (2001).
Guga et al., Unusual Thermal Stability of RNA/[RP-PS]-DNA/RNA Triplexes Containing a Homopurine DNA Strand, Biophys J., 92(7): 2507-2515 (2007).
Guga, P. and Stec, W.J., Synthesis of Phosphorothioate Oligonucleotides with Stereodefined Phsphorothioate Linkages, Current Protocols in Nucleic Acid Chemistry, Unit 4.17: 4.17.1-4.17.28 (2003).
Guga, P., P-chiral oligonucleotides in biological recognition processes, Current Topics in Medicinal Chemistry, 7:695-713 (2007).
Guo, M. et al., Solid-phase stereoselective synthesis of 2′-0-methyl-oligo-ribonucleoside phosphorothioates using nucleoside bicyclic oxazaphospholidines, Biorganic & Medicinal Chemistry Letters, 8(18):2539-2544 (1998).
Guzaev, A.P., Reactivity of 3H-1,2,4-dithiazole-3-thiones and 3H-1,2-dithiole-3-thiones as sulfurizing agents for oligonucleotide synthesis, Tetrahedron Letters, 52: 434-437 (2011).
Hacia, J.G. et al., Phosphorothioate oligonucleotide-directed triple helix formation, Biochemistry, 33:5367-5369 (1994).
Hagedorn, P.H. et al., Locked nucleic acid: modality, diversity, and drug discovery, Drug Discovery, 1-14 (Oct. 2017).
Hammond, S.M. and Wood, M.J. Genetic therapies for RNA mis-splicing diseases, Trends Genet., 27: 196-205 (2011).
Hanagata, N., Structure-dependent immunostimulatory effect of CpG oligodeoxynucleoties and their delivery system, Int. J. Nanomedicine, 7: 2181-95 (2012).
Hansen et al., Azaribofuranoside Analogues as Designed Inhibitors of Purine Nucleoside Phosphorylase, Synthesis and Biological Evaluation, Acta Chemis Scandinavica 52: 1214-1222 (1998).
Haringsma, H.J. et al., mRNA knockdown by single strand Rna is improved by chemical modifications, Nucleic Acids Research, 40(9): 4125-4136 (2012).
Harper, S.Q. et al., RNA interference improves motor and neuropathological abnormalities in a Huntington's disease mouse model, Proc. Natl. Acad. Sci. USA, 102(16): 5820-5825 (2005).
Hartmann, B. et al., Sequence effects on energetic and structural properties of phosphorothioate DNA: a molecular modelling study, Nucleic Acids Research, 27(16): 3342-3347 (1999).
Hartmann, G. et al., Delineation of a CpG Phosphorothioate Oligodeoxynucleotide for Activating Primate Immune Responses In Vitro and In Vivo, The Journal of Immunology, 164(3): 1617-1624 (2000).
Hau, P. et al., Results of G004, a phase lib actively controlled clinical trial with the TGF-b2 targeted compound AP 12009 for recurrent anaplastic astrocytoma, Journal of Clinical Oncology, 2006 ASCO Annual Meeting Proceedings (Post-Meeting Edition), 24(18, Jun. 20 Supplement): 1566 (2006).
Hayashi, S. et al., Studies on Antitumor Substances, Chemical & Pharmaceutical Bulletin, 12(11): 1271-1276 (1964).
Heemskerk, H.A. et al., In vivo comparison of 2′-O-methyl phosphorothioate and morpholino antisense oligonucleotides for Duchenne muscular dystrophy exon skipping, The Journal of Gene Medicine, 11:257-266 (2009).
Heger, W. et al., Embryotoxic effects of thalidomide derivatives on the non-human primate Callithrix jacchus; 3. Teratogenic potency of the EM 12 enantiomers, Arch. Toxicol., 62: 205-208 (1988).
Hendrix, C. et al., 1′,5′-Anhydrohexitol Oligonucleotides: Synthesis, Base Pairing and Recognition by Regular Oligodeoxyribonucleotides and Oligoribonucleotides, Chem. Eur. J., 3(1): 110-120 (1997).
Henry, A.A. and Romesberg, F.E., Beyond A, C, G and T: augmenting nature's alphabet, Current Opinion in Chemical Biology, 7(6): 727-733 (2003).
Henry, S.P. et al., Activation of the Alternative Pathway of Complement by a Phosphorothioate Oligonucleotide: Potential Mechanism of Action, The Journal of Pharmacology and Experimental Therapeutics, 281(2): 810-816 (1997).
Herbert, B-S. et al., Nonradioactive detection of telomerase activity using the telomeric repeat amplification protocol, Nat. Protoc., 1(3): 1583-1590 (2006).
Herdewijn, Oligonucleotide Synthesis, Methods in Molecular Biology, 288: 1-435 (2005).
Heuberger, B.D. and Switzer, C., A Pre-RNA Candidate Revisited: Both Enantiomers of Flexible Nucleoside Triphosphates are DNA Polymerase Substrates, Journal of the American Chemical Society, 130(2):412-413 (2008).
Higuchi, T. et al., Pro-drugs as Novel Delivery Systems, ACS Symposium Series, 14 (1975).
Hirama, T. et al., PCR-Based Rapid Identification System Using Bridged Nucleic Acids for Detection of Clarithromycin-Resistant Mycobacterium avium-M. intracellulare Complex Isolates, Journal of Clinical Microbiology, 54(3): 699-704 (2016).
Hirao, I., Unnatural base pair systems for DNA/RNA-based biotechnology, Current Opinion in Chemical Biology,10:622-627 (2006).
Hirose, M. et al., MDM4 expression as an indicator of TP53 reactivation by combined targeting of MDM2 and MDM4 in cancer cells without TP53 mutation, Oncoscience, 1(12): (2014).
Hohjoh, H., Disease-Causing Allele-Specific Silencing by RNA Interference, Pharmaceuticals, 6: 522-535 (2013).
Hu, J. et al., Allele-Selective Inhibition of Huntingtin Expression by Switching to an miRNA-like RNAi Mechanism, Chemistry & Biology 17: 1183-1188 (2010).
Hu, J. et al., Exploring the Effect of Sequence Length and Composition on Allele-Selective Inhibition of Human Huntingtin Expression by Single-Stranded Silencing RNAs, Nucleic Acid Therapeutics, 24(3): 199-209 (2014).
Hu, J. et al., Recognition of c9orf72 Mutant RNA by Single-Stranded Silencing RNAs, Nucleic Acid Therapeutics, 8 (2016). Supplementary Figure, 1 page.
Hunziker, J. et al., Why Pentose—And Not Hexose-Nucleic Acids? Part III. Oligo(2′,3′-dideoxy-(3-D-glucopyranosyl)nucleotides. (‘Homo-DNA’): Base-Pairing Properties, Helvetica Chimica Acta, 76(1):259-352 (1993).
Hyrup., B. and Nielsen, P.E., Peptide Nucleic Acids (PNA): Synthesis, Properties and Potential Applications, Bioorg. Med. Chem., 4(1): 5-23 (1996).
Inagawa, T. et al., Inhibition of human immunodeficiency virus type 1 replication by P-stereodefined oligo(nucleoside phosphorothioate)s in a long-term infection model, FEBS Letters, 528(1-3): 48-52 (2002).
International Preliminary Report on Patentability for PCT/JP2010/065900, 6 pages (dated Mar. 29, 2012).
International Preliminary Report on Patentability for PCT/JP2010/065900, English Translation, 7 pages (dated Apr. 19, 2012).
International Preliminary Report on Patentability for PCT/JP2011/055018, English Translation, 5 pages (dated Oct. 11, 2012).
International Preliminary Report on Patentability for PCT/JP2011/071559, English Translation, 7 pages (dated Apr. 25, 2014).
International Preliminary Report on Patentability for PCT/JP2013/004303, 7 pages (dated Jan. 13, 2015).
International Preliminary Report on Patentability for PCT/JP2013/069107, English Translation, 10 pages (dated Jan. 15, 2015).
International Search Report for PCT/IB2009/007923, 4 pages (dated Sep. 6, 2010).
International Search Report for PCT/IB2015/000395, 7 pages (dated Oct. 30, 2015).
International Search Report for PCT/JP2010/065900, 1 page (dated Sep. 15, 2010).
International Search Report for PCT/JP2011/055018, 2 pages (dated Mar. 29, 2011).
International Search Report for PCT/JP2011/071559, 3 pages (dated Dec. 20, 2011).
International Search Report for PCT/JP2011/077313, 2 pages (dated Jan. 10, 2012).
International Search Report for PCT/JP2013/004303, 3 pages (dated Aug. 13, 2013).
International Search Report for PCT/JP2013/069107, 2 pages (dated Oct. 1, 2013).
International Search Report for PCT/JP2015/050714, and English Translation, 8 pages (dated Apr. 21, 2015).
International Search Report for PCT/JP2015/050716 and English Translation, 8 pages (dated Apr. 21, 2015).
International Search Report for PCT/JP2015/050718 and English Translation, 8 pages (dated Apr. 21, 2015).
International Search Report for PCT/US2010/041068, 1 page (dated Sep. 1, 2010).
International Search Report for PCT/US2011/064287, 2 pages (dated Apr. 12, 2012).
International Search Report for PCT/US2012/046805, 2 pages (dated Sep. 19, 2012).
International Search Report for PCT/US2013/050407, 5 pages (dated Jan. 9, 2014).
International Search Report for PCT/US2016/043542, 6 pages (dated Dec. 28, 2016).
International Search Report for PCT/US2016/043598, 4 pages (dated Nov. 28, 2016).
International Search Report for PCT/US2016/056123, 5 pages (dated Mar. 17, 2017).
International Search Report for PCT/US2017/022135, 3 pages (dated Jun. 6, 2017).
International Search Report for PCT/US2017/030753, 6 pages (dated Sep. 26, 2017).
International Search Report for PCT/US2017/030777, 5 pages (dated Oct. 2, 2017).
International Search Report for PCT/US2017/035837, 4 pages (dated Aug. 24, 2017).
International Search Report for PCT/US2017/043431, ISA/US, 5 pages (dated Dec. 21, 2017).
International Search Report for PCT/US2017/045218, 3 pages (dated Sep. 27, 2017).
International Search Report for PCT/US2017/055601, ISR/US, 6 pages (dated Feb. 15, 2018).
International Search Report for PCT/US2017/062996, 4 pages (dated Mar. 9, 2018).
Ionis Pharmaceuticals, Inc., Ionis Pharmaceuticals Licenses IONIS-HTT Rx to Partner Following Successful Phase 1/2a Study in Patients with Huntington's Disease, Press Release, 2 pages (Dec. 11, 2017).
Isis Pharmaceuticals, Inc. 2014 Annual Report, Improving Patients' Lives by Treating Disease Through Targeting RNA, 192 pages (2014).
Isis Pharmaceuticals, Inc. v. Santaris Pharma A/S Corp., Order Denying Defendants' Motion for Summary Judgment Without Prejudice, Case No. 11cv02214 BTM (KSC), United States District Court, S.D. California, 5 pages (Sep. 19, 2012).
Isis Pharmaceuticals, Intellectual Property: Capturing Value From Innovation, Isis' Annual Meeting of Stockholders and Open House, Intellectual Property Poster, 1 page (2011). Received from Internet <http://www.isispharm.com/Site_Gfx/pdf/11-AnMtg_IntellectualProperty_TAB.pdf>.
Isis Pharmaceuticals, Intellectual Property: Capturing Value From Innovation, Isis' Annual Meeting of Stockholders and Open House, Intellectual Property Poster, 1 page (2012). Received from Internet <http://www.isispharm.com/Site_Gfx/pdf/2012_Annual_Meeting_IP_Poster.pdf>.
Iwamoto et al., Stereocontrolled Synthesis of H-phosphonate DNA, Nucleic Acids Symposium Series, (50):159-60 (2006).
Iwamoto, N. et al., Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides, Nature Biotechnology, Life Sciences Reporting Summary, 6 pages (2017).
Iwamoto, N. et al., Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides, Nature Biotechnology, pp. 1-9 (2017).
Iwamoto, N. et al., Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides, Nature Biotechnology, Supplementary Methods, Supplementary Tables 1-4, and Supplementary Note, 23 pages (2017).
Iwamoto, N. et al., Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides, Nature Biotechnology, Supplementary Text and Figures 1-9, 13 pages (2017).
Iwamoto, N. et al., Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides, Nature Biotechnology, with Supplemental Data, 19 pages (2017).
Iwamoto, N. et al., Optimization of Therapeutic Phosphorothioate Oligonucleotides by P-Chirality Control, WAVE Life Sciences, PSJ Congress: The Pharmaceutical Society of Japan, (Mar. 25, 2015-Mar. 28, 2016).
Iwamoto, N. et al., Stereocontrolled solid-phase synthesis of oligonucleoside H-phosphonates by an oxazaphospholidine approach, Angewandte Chemie International Edition, 48(3):496-499 (2009).
Iyer, R.P. et al., A novel nucleoside phosphoramidite synthon derived from 1R, 2S-ephedrine, Tetrahedron Asymmetry 6(5):1051-1054 (1995).
Iyer, R.P. et al., Acyloxyaryl prodrugs of oligonucleoside phosphorothioates, Bioorganic and Medicinal Chemistry Letters, 6(16):1917-1922 (1996).
Iyer, R.P. et al., Bioreversible oligonucleotide conjugates by site-specific derivatization, Bioorganic and Medicinal Chemistry Letters, 7:871-876 (1997).
Iyer, R.P. et al., Stereospecific Bio-Reversibility of Dinucleoside S-Alkyl Phosphorothiolates to Dinucleoside Phosphorothioates, Bioorganic & Medicinal Chemistry Letter, 4(20):2471-2476 (1994).
Iyer, R.P., et al., 3H-1,2-Benzodithiole-3-one 1,1-Dioxide as an Improved Sulfurizing Reagent in the Solid-Phase Synthesis of Oligodeoxyribonucleoside Phosphorothioates, Journal of the American Chemical Society, 112(3):1253-1254 (1990).
Iyer, R.P., et al., Prodrugs of Oligonucletides: The Acyloxyalkyl Esters of Oligodeoxyribonucleoside Phosphorothioates, Bioorganic Chemistry, 23:1-21 (1995).
Iyer, R.P., et al., Solid-phase stereoselective synthesis of oligonucleoside phosphorothioates: The nucleoside bicyclic oxazaphospholidines as novel synthons, Tetrahedron Letters, 39:2491-2494 (1998).
Jahns, H., et al., Stereochemical bias introduced during RNA synthesis modulates the activity of phosphorothioate siRNAs, Nat. Commun., 6: 6317 (2015).
Jepsen, J.S. et al., LNA-Antisense Rivals Sirna for Gene Silencing, Current Opinion in Drug Discovery and Development, 7(2): 188-194 (2004).
Jepsen, J.S. et al., Locked Nucleic Acid: A Potent Nucleic Acid Analog in Therapeutics and Biotechnology, Oligonucleotides,14: 130-146 (2004).
Jiang, J. et al., Allele-Specific Silencing of Mutant Myh6 Transcripts in Mice Suppresses Hypertrophic Cardiomyopathy, Science, 342: 111-114 (2013).
Jin et al., A Stereoselective Synthesis of Dinucleotide Boranophosphate, Using Chiral Indole-Oxazaphosphorine Intermediates, Tetrahedron Letters, 39: 6433-6436 (1998).
Jin et al., Stereoselective Synthesis of Dithymidine Phosphorothioates Using Xylose Derivatives as Chiral Auxiliaries, J. Org. Chem., 63(11): 3647-3654 (1998).
Johansson et al., Studies towards synthesis of dinucleoside arylphosphonates with metal complexing properties, Nucleosides Nucleotides & Nucleic Acids, 22(5-8): 1459-61 (2003).
Johansson et al., Synthesis of dinucleoside pyridylphosphonates involving palladium(o)-catalysed phosphorus-carbon bond formation as a key step, Chem. Commun., 2564-2565 (2001).
Johansson et al., The case for configurational stability of H-phosphonate diesters in the presence of diazabicyclo[5.4.0]undec-7-ene (DBU), Bioorg Med Chem., 9(9): 2315-22 (2001).
Jones, R.J. et al., Synthesis and binding properties of pyrimidine oligodeoxynucleoside analogs containing neutral phosphodiester replacements: The Formacetal and 3′-Thioformacetal Internucleoside Linkages, J. Org. Chem., 58: 2983-2991 (1993).
Jopling, C.L. et al., Modulation of Hepatitis C Vicus RNA Abundance by a Liver-Specific MicroRNA, Science, 309: 1577-1581 (2005).
Joyce, G.F. et al., The case for an ancestral genetic system involving simple analogues of the nucleotide, Proceedings of the National Academy of Sciences, 84:4398-4402 (1987).
Joyce, G.F. The antiquity of RNA-based evolution, Nature, 418(6894): 214-221 (2002).
Kakeya, N. et al., Studies on Prodrugs of Cephalosporins. I. Synthesis and Biological Properties of Glycyloxybenzoyloxymethyl and Glycylaminobenzoyloxymethyl Esters of 7-[2-(2- Aminothiazol-4-yl)-(Z)-2-methoxyiminoacetamido]-3-methyl-3-cephem-4-carboxylic Acid, Chem. Pharm. Bull., 32(2): 692-698 (1984).
Kamada, A.K. et al., Issues in the Use of Inhaled Glucocorticoids, Am. J. Respir. Crit. Care. Med., 153: 1739-1748 (1996).
Karwowski, B. et al., Stereocontrolled Synthesis of LNA Dinucleoside Phosphorothioate by the Oxathiaphospholane Approach, Bioorganic & Medicinal Chemistry Letters, 11: 1001-1003 (2001).
Kashida, H. et al., Acyclic artificial nucleic acids with phosphodiester bonds exhibit unique functions, Polymer Journal, 1-6 (2016).
Kaur, H. et al., Activation of natural killer-like YT-INDY cells by oligodeoxynucleotides and binding by homologous pattern recognition proteins, Scandinavian Journal of Immunology, 62: 361-370 (2005).
Kawasaki, A et. al., Uniformly Modified 2′-Deoxy-2′-fluoro Phosphorothioate Oligonucleotides as Nuclease-Resistant Antisense Compounds with High Affinity and Specificity for RNA Targets, J. Med. Chem., 36: 831-841 (1993).
Kay, C. et al., Huntingtin Haplotypes Provide Prioritized Target Panels for Allele-Specific Silencing in Huntington Disease Patients of European Ancestry, Molecular Therapy, Accepted Article Preview Online (Jul. 23, 2015).
Kay, C. et al., Huntingtin Haplotypes Provide Prioritized Target Panels for Allele-specific Silencing in Huntington Disease Patients of European Ancestry, The American Society of Gene & Cell Therapy, 1-13 (2015).
Kay, C. et al., Personalized gene silencing therapeutics for Huntington disease, Clinical Genetics, 1-8 (2014).
Kers et al., A new type of nucleotide analogue with 4-pyridylphosphonate internucleotide linkage, Tetrahedron Letters, 40(22): 4263-4266 (1999).
Kihara, M et al., New norepinephrine potentiators: synthesis and structure-actvity relastionships of a series of 4-phenyl-1,2,3,4-tetrahydroisoquinolin-4-ols, Chemical & Pharmaceutical Bulletin 42(1): 67-73 (1994).
Kim, D. et al., Immunostimulation and anti-DNA antibody production by backbone modified CpG-DNA, Biochemical and Biophysical Research Communications, 379(2): 362-367 (2009).
Kim, M., Beta conformation of polyglutamine track revealed by a crystal structure of Huntingtin N-terminal region with insertion of three histidine residues, Prion, 7(3): 221-228 (2013).
Kim, N.W. et al., Specific Association of Human Telomerase Activity with Immortal Cells and Cancer, Science, 226: 2011-2015 (1994).
Kim, S-H. and Cech, T.R., Three-dimensional model of the active site of the selfsplicing rRNA precursor of Tetrahymena, Proc. Natl. Acad. Sci. U S A., 84(24): 8788-8792 (1987).
Kim, S-K. et al., Bridged Nucleic Acids (BNAs) as Molecular Tools, J Biochem Mol Biol Res., 1(3): 67-71 (2015).
Kim, S. et al., Liquid-Phase RNA Synthesis by Using Alkyl-Chain-Soluble Support, Chem. Eur. J., 19: 8615-8620 (2013).
Kiviniemi, a. et al., Solid-Supported 2′-O-Glycoconjugation of Oligonucleotides by Azidation and Click Reactions, Bioconjugate Chemistry, 22(6): 1249-1255 (2011).
Klose, J. et al., Preparation of 2-(2-Cyanoethyl)-sulfany1-1H-isoindole-1,3-(2H)-dione and related sulfur transfer reagents, Tetrahedron, 53(42):14411-14416 (1997).
Koch, T., A New Dimension in LNA Therapeutics, Roche Innovation Center, Copenhagen, Denmark, Presentation, 39 pages (May 3, 2017).
Koizumi, M. et al., Triplex formation with 2′-O,4′-C-ethylene-bridged nucleic acids (ENA) having C3′-endo conformation at physiological pH, Nuc. Acids Res., 31(12): 3267-3273 (2003).
Kool, E.T., Replacing the Nucleobases in DNA with Designer Molecules, Accounts of Chemical Research, 35:936-943 (2002).
Kordasiewicz, H.B. et al., Sustained therapeutic reversal of Huntington's disease by transient repression of huntingtin synthesis, Neuron, 74(6): 1031-1044 (2012).
Koseoglu, M. et al., Effects of hemolysis interference on routine biochemistry parameters. Biochemia Medica., 21(1): 79-85 (2011). Retrieved May 18, 2017, URL: <http://www.biochemia-medica.com/2011/21/79>.
Koshkin, A.A. et al., LNA (Locked Nucleic Acids): Synthesis of the Adenine, Cytosine, Guanine, 5-Methylcytosine, Thymine and Uracil Bicyclonucleoside Monomers, Oligomerisation, and Unprecedented Nucleic Acid Recognition, Tetrahedron 54: 3607-3630 (1998).
Kozikowski, A.P. et al., Chemistry of the main group metals: A stereoselective synthesis of allyl vinyl thioethers for the thio-claisen reaction, Journal of Organometallic Chemistry, 164(3): C33-C37 (1979).
Koziolkewicz et al., Stability of Stereoregular Oligo-(nucleoside Phosphorothioate)s in Human Plasma: Diastereoselectiviy of Plasma 3′-Exonuclease, Antisense Nucl. Acid Drug Dev., 7: 43-48 (1997).
Koziolkewicz et al., Stereodifferentiation-the effect of P chirality of oligo(nucleoside phosphorothioates) on the activity of bacterial RNase H, Nucl. Acids Res., 23(24): 5000-5005 (1995).
Koziolkiewicz, M. et al., Effect of P-chirality of oligo(deoxyribonucleoside phosphorothioate)s) on the activity of terminal deoxyribonucleotidyl transferase, FEBS Letters, 434(1-2): 77-82 (1998).
Kraszewski et al., Studies on Reactions of Nucleoside H-phosphonates with Bifunctional Reagents. Part 1. Reaction with amino alcohols, J. Chem. Soc., Perkin Trans., 1: 1699-1704 (1993).
Kremer, B. et al., A Worldwide Study of the Huntington's Disease Mutation, The New England Journal of Medicine, 330(20): 1401-1406 (1994).
Kretschmer-Kazemi Far, R. and Sczakiel, G., The activity of siRNA in mammalian cells is related to structural target accessibility: a comparison with antisense oligonucleotides, Nucleic Acids Research, 31(15):4417-4424 (2003).
Krieg, A.M. et al., CpG motifs in bacterial DNA trigger direct B-cell activation, Nature, 374: 546-549 (1995).
Krieg, A.M. et al., P-Chirality-Dependent Immune Activiation by Phosphorothioate CpG Oligodeoxynucleotides, Oligonucleotides, 13:491-499 (2003).
Krieg, A.M., Development of TLR9 agonists for cancer therapy, The Journal of Clinical Investigation, 117(5): 1184-1194 (2007).
Krieg, A.M., Therapeutic potential of Toll-like receptor 9 activation, Nature Reviews, 471-484 (2006).
Krotz, A.H. et al., Phosphorothioate Oligonucleotides with Low Phosphate Diester Content: Greater than 99.9% Sulfurization Efficiency with “Aged” Solutions of Phenylacetyl Disulfide (PADS), Organic Process Research & Development, 8: 852-858 (2004).
Krueger, A.T. et al., Synthesis and properties of size-expanded DNAs: toward designed, functional genetic systems, Accounts of Chemical Research, 40:141-150 (2007).
Krutzfeldt, J. et al., Silencing of microRNAs in vivo with ‘antagomirs’, Nature, 438: 685-689 (2005).
Kumar, R. et al., The First Analogues of LNA (Locked Nucleic Acids): Phosphorothioate-LNA and 2′-THIO-LNA, Bioo. Med. Chem. Let., 8: 2219-2222 (1998).
Kungurtsev, V. et al., Solution-Phase Synthesis of Short Oligo-2′-deoxyribonucleotides by Using Clustered Nucleosides as a Soluble Support, Eur. J. Org. Chem., 6687-6693 (2013).
Kuramoto, Y. et al., Mannosylated cationic liposomes/CpG DNA complex for the treatment of hepatic metastasis after intravenous administration in mice, Journal of Pharmaceutical Science, 98(3): 1193-1197 (2009).
Kwon, H-J. et al., NF-kappaB-dependent regulation of tumor necrosis factor-alpha gene expression by CpG-oligodeoxynucleotides, Biochem. Biophys. Res. Commun., 311(1): 129-138 (2003).
Lahiri, N., Shooting the messenger with single-stranded RNA gene silencing, edited by Wild, E., HDBuzz, 7 pages (Sep. 24, 2012). Retrieved Oct. 7, 2015. URL: http://en.hdbuzz.net/099.
LaPlanche, L.A. et al., Phosphorothioate-modified oligodeoxyribonucleotides. III. NMR and UV spectroscopic studies of the Rp-Rp, Sp-Sp, and Rp-Sp duplexes, [d(GGsAATI'CC)2, derived from diastereomeric 0-ethyl phosphorothioates, Nucleic Acids Research, 14(22): 9081-9093 (1986).
Latimer, L.J.P. et al, Synthetic repeating sequence DNAs containing phosphorothioates: nuclease sensitivity and triplex formation, Nucleic Acids Research, 17(4): 1549-1561 (1989).
Laurent et al., Chiral and steric effects in the efficient binding of alpha-anomeric deoxyoligonucleoside N-alkylphosphoramidates to ssDNA and RNA, Nucleic Acids Res., 27(21): 4151-9 (1999).
Lauritsen, A. et al., Methylphosphonate LNA: A Locked Nucleic Acid with a Methylphosphonate Linkage, Bioo. Med. Chem. Lett., 13: 253-256 (2003).
Lauritsen, A. et al., Oligodeoxynucleotides containing amide-linked LNA-type dinucleotides: synthesis and high-affinity nucleic acid hybridization, Chem. Comm., 5: 530-531 (2002).
Lavergne, T. et al., A Base-Labile Group for 2′-OH Protection of Ribonucleosides: A Major Challenge for RNA Synthesis, Chem. Eur. J, 14, 9135-9138 (2008).
Lee, K-W et al., CG sequence- and phosphorothioate backbone modification-dependent activation of the NF-κB-responsive gene expression by CpG-oligodeoxynucleotides in human RPMI 8226 B cells, Molecular Immonulogy, 41: 955-964 (2004).
Lesnikowski et al., Studies on Stereospecific Formation of P-Chiral Internucleotide Linkage. Synthesis of (RP, RP)- and (SP, SP)-Thymidylyl (3′, 5′) Thymidylyl (3′, 5′) Thymidine DI (O,O-Phosphorothioate) Using 2-Nitrobenzyl Group as a New S-Protection, Tetrahedron Letters 30(29) 3821-3824 (1989).
Lesnikowski, Z. J. et al., Octa(thymidine methanephosphonates) of partially defined sterochemistry: synthesis and effect of chirality at phosphorus on binding to pentadecadeoxyriboadenylic acid, Nucleic Acids Research, 18(8): 2109-2115 (1990).
Levin, A.A. et al., Basic Principles of the Pharmacokinetics of Antisense Oligonucleotide Drugs, Antisense Drug Technology: Principles, Strategies, and Applications, Second Edition, Chapter 7: 183-215 (2008).
Leviten, M., Wave's Purity Progress, Biocentury, 1-6 (Sep. 28, 2017).
Li L.C., Small RNA Mediated Gene Activation, RNA and the Regulation of Gene Expression: A Hidden Layer of Complexity, Edited by Kevin V. Morris, Chapter 13, Caister Academic Press (2008).
Li, L-C. et al., Small dsRNAs induce transcriptional activation in human cells, PNAS, 103(46): 17337-17342 (2006).
Li, M. et al., Synthesis and cellular activity of stereochemically-pure 2′-O-(2-methoxyethyl)-phosphorothioate oligonucleotides, Chem. Commun., 53: 541-544 (2017).
Li-Tsang, C.W. et al., Prevalence of hypertrophic scar formation and its characteristics among the Chinese population, Burns, 31: 610-616 (2005).
Liang, X-h. et al., Identification and characterization of intracellular proteins that bind oligonucleotides with phosphorothioate linkages, Nucleic Acids Research, 43(5): 2927-2945, Supplemental Data pp. 1-20 (2015).
Lima, W. et al., Single-Stranded ssRNAi Activate RNAi in Animals, Cell, 150: 883-894 (2012).
Lima, W.F. et al., The influence of antisense oligonucleotide-induced RNA structure on Escherichia coli RNase H1 activity, J. Biol. Chem., 272(29):18191-9 (1997).
Lima, W.F., et al., Human RNase H1 discriminates between subtle variations in the structure of the heteroduplex substrate, Mol. Pharmacol., 71: 83-91 (2007).
Limbach, P.A. et al., Summary: the modified nucleosides of RNA, Nucleic Acids Research, 22(12):2183-2196 (1994).
Lin et al., Synthesis and resolution of dinucleotide (TpAZT) phosphoramidates, Synthetic Commun., 33(14): 2553-2562 (2003).
Linton, M.F., et al., Transgenic Mice Expressing High Plasma Concentrations of Human Apolipoproteins B100 and Lipoprotein (a), J. Clin. Invest., 92: 3029-37 (1993).
Liu, J. et al., Modulation of Splicing by Single-Stranded Silencing RNAs, Nucleic Acid Therapeutics, 25(3): 113-120 (2015).
Liu, W. et al., Increased Steady-State Mutant Huntingtin mRNA in Huntington's Disease Brain, Journal of Huntington's Disease 2: 491-500 (2013).
Lopez, C. et al., Inhibition of AAC(6′)-Ib-Mediated Resistance to Amikacin in Acinetobacter baumannii by an Antisense Peptide-Conjugated 2′,4′- Bridged Nucleic Acid-NC-DNA Hybrid Oligomer, Antimicrobial Agents and Chemotherapy, 59(9): 5798-5803 (2015).
Lu, X. et al., Antisense-Mediated Inhibition of Human Immunodeficiency Virus (HIV) Replication by Use of an HIV Type 1-Based Vector Results in Severely Attenuated Mutants Incapable of Developing Resistance, Journal of Virology, 78(13): 7079-7088 (2004).
Lu, Y. And Just, G., Stereoselective synthesis of dithymidine phosphorothioates using d-xylose derived chiral auxiliaries, Tetrahedron, 57(9):1677-1687 (2001).
Lu, Y. et al., Stereoselective Synthesis of R(P)- and S(P)-Dithymidine Phosphorothioates via Chiral Indolooxazaphosphorine Intermediates Derived from Tryptophan This work was financially supported by Natural Science and Engineering Research Council of Canada (NSERC). We thank Nadim Saadeh and Dr. Orval Mamer, McGill University biomedical mass spectroscopy unit, for recording mass spectra, Angewandte Chemie International Edition, 39(24):4521-4524 (2000).
Lu, Y., Recent advances in the stereocontrolled synthesis of antisense phosphorothioates, Mini Reviews in Medicinal Chemistry, 6(3): 319-330 (2006).
Machine Translation of JP 2010-265304 (2010). <http://dossier1.ipdl.inpit.go.jp/AIPN/odse_top_dn.ipdl?NOOOO=7400>.
Machytka et al., Extension of the Applicability of &I-Values for the Configurational Assignment of Diastereomeric Phosphate-Modified Dideoxynucleotides, Nucleosides and Nucleotides, 17(12): 2311-2322 (1998).
Machytka et al., Synthesis and NMR characterization of diastereomeric CPSMeG derivatives, Nucleosides Nucleotides Nucleic Acids., 19(5-6): 903-15 (2000).
Maher III, L.J., et al., Inhibition of DNA Binding Proteins by Oligonucleotide-Directed Triple Helix Formation, Science, 245: 725-730 (1989).
Mann, M.J. et al., Therapeutic applications of transcription factor decoy oligonucleotides, J. Clin. Invest., 106:1071-1075 (2000).
Mannironi, C. et al., In Vivo Selection of Dopamine RNA Ligands, Biochemistry, 36: 9726-9734 (1997).
Martin, P., A New Access to 2′-O-alkylated Ribonucleosides and Properties of 2′-O-Alkylated Oligoribonucleotides, Helv. Chim. Acta., Abstract Only, 78: 486-504 (1995).
Martin, P., Stereoselective Synthesis of 2′-O-(2-Methoxyethyl)ribonucleosides: Neighboring-Group Participation of the Methoxyethoxy Group in the Ribosylation Step, Helv. Chim. Acta, 79: 1930-1938 (1996).
Martinez, J. et al., Single-Stranded Antisense siRNAs Guide Target RNA Cleavage in RNAi, Cell, 110: 563-574 (2002).
Martinez-Montero, S. et al., Locked 2′-Deoxy-2′,4′-Difluororibo Modified Nucleic Acids: Thermal Stability, Structural Studies, and siRNA Activity, ACS Chem. Biol., 10: 2016-2023 (2015).
Masahiro, T. et al., Nematicidal and antimicrobial constituents from Allium grayi Regel and Allium fistulosum L. var. caespitosum, Agricultural and Biological Chemistry, 52(9): 2383-2385 (1988).
Matranga, C. et al., Passenger-Strand Cleavage Facilitates Assembly of siRNA into Ago2-Containing RNAi Enzyme Complexes, Cell, 123: 607-620 (2005). Supplemental Data, 6 pages.
Matsui, M. et al., Argonaute 2-dependent Regulation of Gene Expression by Single-stranded miRNA Mimics, Molecular Therapy, 10 pages (2016).
Matsui, M. et al., Transcriptional Silencing by Single-Stranded RNAs Targeting a Noncoding RNA That Overlaps a Gene Promoter, ACS Chem. Biol., 8: 122-126 (2013).
Matsuno, Y. et al., Synthetic Method for Oligonucleotide Block by Using Alkyl-Chain-Soluble Support, Org. Lett., 18: 800-803 (2016).
Matysiak, S et al., Acetals as New 2′-O-Protecting Functions for the Synthesis of the Oligoribonucleotides: Synthesis of Uridine Building Blocks and Evaluatino of Their Relative Acid Stability, Helvetica Chimica Acta 81: 1545-1566 (1998).
Maung, J. et al., Alternatives to 1-H-tetrazole in the preparation of phosphonate diesters and phosphonamidates from phosphonyl dichlorides, Tetrahedron Lett., 45: 6497-6499 (2004).
Mauritz, R.P. et al., Elucidation of the Hydrolytical Properties of α-Hydroxybenzylphosphonates as a New Potential Pro-Oligonucleotide Concept, Nucleosides and Nucleotides, 18(6-7):1417-1418 (1999).
Mauritz, R.P. et al., Synthesis of 3′,5′-Dithymidylyl-α-hydroxyphosphonate Dimer Building Blocks for Oligonucleotide Synthesis—A New Pro-oliguncleotide, Nucleosides and Nucleotides, 16(7-9):1209-1212 (1997).
McBride, J.L. et al., Prelinical Safety of RNAi-Mediated HTT Suppression in the Rhesus Macaque as a Potential Therapy for Huntington's Disease, Molecular Therapy, 19:1-11 (2011).
Meade, M.F., et al., Efficient delivery of RNAi prodrugs containing reversible charge-neutralizing phosphotriester backbone modifications, Nat. Biotech., 32: 1256-61 (2014).
Medical News Today, AVI BioPharma Announces FDA Clears IND Applications for Clinical Trials of RNA Therapeutic Agents for Treatment of Ebola and Marburg Viruses, Accessed Apil 2, 2015, 2 pages (Dec. 30, 2008).
Meena, Control of Human RNase H Mediated Cleavage by Stereopure Phosphorothioate Oligonucleotides, WAVE Life Sciences, TIDES Meeting, 23 pages (May 3-6, 2015).
Meena, Development of Allele Specific Antisense Oligonucleotides, WAVE Life Sciences, ACS Central Regional Meeting (CERM), Covington, KY (May 19, 2016).
Meena, Development of Allele Specific Antisense Oligonucleotides, WAVE Life Sciences, TIDES Meeting (May 11, 2016).
Meena, et al., Discovery and Early Clinical Development of the First Allele-Specific Stereopure ASO Drug Candidate with Disease-Modifying Potential for the Treatment of Huntington's Disease, WAVE Life Sciences, Poster, 1 page (2016).
Meena, et al., Therapeutic Implications of Controlling P-Chirality in Phosphorothioate Oligonucleotides, TIDES Poster (May 12-15, 2014).
Meena, et al., Therapeutic Implications of Controlling P-Chirality in Phosphorothioate Oligonucleotides, TIDES, San Diego, WAVE Life Sciences, Poster, 1 page (May 3-6, 2014).
Meena, Optimization of Antisense Drugs by P-Stereochemistry Control, WAVE Life Sciences, OTS Annual Meeting 2014, Oligonucleotide Therapeutics Society, 13 pages (Oct. 12-14, 2014).
Merki, E. et al., Antisense oligonucleotide directed to human apolipoprotein B-100 reduces lipoprotein(a) levels and oxidized phospholipids on human apolipoprotein B-1 00 particles in lipoprotein(a) transgenic mice, Circulation, 118(7): 743-53 (2008).
Mesmaeker, A.D. Backbone modifications in oligonucleotides and peptide nucleic acid systems, Current Opinion in Structural Biology, 5: 343-355 (1995).
Mesmaeker, A.D. et al. Amides as a New Type of Backbone Modification in Oligonucleotides, Angew. Chem., Int. Ed. Engl., 33: 226-229 (1994).
Methods in Enzymology, Edited by Widder, K. and Green, R., Drug and Enzyme Targeting, Academic Press, 112: 309-396 (1985).
Midturi, J. et al., Spectrum of Pulmonary Toxicity Associated with the Use of Interferon Therapy for Hepatitis C: Case Report and Review of the Literature, Clinical Infectious Diseases, 39(11): 1724-1729 (2004).
Mignet, N. et al., Synthesis and evaluation of glucuronic acid derivatives as alkylating agents for the reversible masking of internucleoside groups of antisense oligonucleotides, Carbohydrate Research, 303:17-24 (1997).
Mignet, N. et al., The Prooligonucleotide Approach. V: Influence of the phosphorus atom environment on the hydrolysis of enzymolabile dinucleoside phosphotriesters, Bioorganic and Medicinal Chemistry Letters, 7(7):851-854 (1997).
Milkowski, J.D. et al., Thiol Protection with the Acetamidomethyl Group: S-Acetamidomethyl-l-cysteine Hydrochloride, Organic Syntheses, 6: 5 (1988).
Misaki, S et al., Dehydration of 2-Trifluoromethyl-3,3,3-Trifluoropropanil with Base, Journal of Flourine Chemistry 24: 531-533 (1984).
Molenkamp, B.G. et al., Local Administration of PF-3512676 CpG-B Instigates Tumor-Specific CD8+ T-Cell Reactivity in Melanoma Patients , Clin. Cancer Res., 14(14): 4532-4542 (2008).
Molina, a.G. et al., Acetylated and Methylated β-Cyclodextrins asViable Soluble Supports for the Synthesis of Short 2′-Oligodeoxyribo-nucleotides in Solution, Molecules, 17: 12102-12120 (2012).
Molina, A.G. et al., Assembly of Short Oligoribonucleotides from Commercially Available Building Blocks on a Tetrapodal Soluble Support, Current Organic Synthesis, 12:1-6 (2015).
Molina, A.G. et al., Solution phase synthesis of short oligoribonucleotides on a precipitative tetrapodal support, Beilstein Journal of Organic Chemistry, 10: 2279-2285 (2014).
Molina, A.G., Synthesis of Short Oligonucleotides on a Soluble Support by the Phosphoramidite Method, University of Turku, 1-66 (2015).
Monteys, A.M. et al., Artificial miRNAs Targeting Mutant Huntingtin Show Preferential Silencing In Vitro and In Vivo, Molecular THerapy—Nucleic Acids, 4: e234 1-11 (2015).
Monteys, A.M. et al., Single nucleotide seed modification restores in vivo tolerability of a toxic artificial miRNA sequence in the mouse brain, Nucleic Acids Res., 42(21): 13315-13327 (2014).
Morales-Rojas, H. and Kool, E.T., A porphyrin C-nucleoside incorporated into DNA, Organic Letters, 4(25):4377-4380 (2002).
Morcos, P.A., Achieving targeted and quantifiable alteration of mRNA splicing with Morpholino oligos, Biochem. Biophys. Res. Commun., 358(2): 521-527 (2007).
Morita , K. et al., 2′-O,4′-C-Ethylene-bridged nucleic acids (ENA) with nuclease-resistance and high affnity for RNA, Nucl. Acids Res., Supp. 1: 241-242 (2001).
Morita , K. et al., 20-O,40-C-Ethylene-Bridged Nucleic Acids (ENA): Highly Nuclease-Resistant and Thermodynamically Stable Oligonucleotides for Antisense Drug, Bioo. Med. Chem. Lett., 12: 73-76 (2002).
Morita, K. et al., Synthesis and properties of 2′-O,4′-C-Ethylene-bridged nucleic acids (ENA) as effective antisense oligonucleotides, Bioorganic & Medicinal Chemistry, 11(10): 2211-2226 (2003).
Morvan, F. et al., Cellular uptake and intracellular quantification of fluorescent labeled T20 Me-SATE prooligonucleotides, Nucleosides Nucleotides Nucleic Acids, 20(4-7):1165-1168 (2001).
Morvan, F. et al., Kinetics study of the biotransformation of an oligonucleotide prodrug in cells extract by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, Nucleosides, Nucleotides and Nucleic Acids, 20(2-4):1159-1163 (2001).
Morvan, F. et al., The Oligonucleotide Prodrug Approach: The Pro-Oligonucleotides, Pharmaceutical Aspects of Oligonucleotides, 79-97 (2000).
Moser, H. E. et al., Sequence-Specific Cleavage of Double Helical DNA by Triple Helix Formation, Science, 238: 645-650 (1987).
Nawrot et al., DNA Oligonucleotides Containing Stereodefined Phosphorothioate Linkages in Selected Positions, Current Protocols in Nucleic Acid Chemistry, UNIT 4.34: 4.34.1-4.34.15 (2009).
Nencka, R. et al., Novel Conformationally Locked Nucleosides and Nucleotides, Collection Symposoim Series, 14: 119-122 (2014).
Nielsen, J. and Caruthers, M.H., Directed Arbuzov-type reactions of 2-cyano-1,1-dimethylethyl deoxynucleoside phosphites, J. Am. Chem. Soc., 110: 6275-6 (1988).
Nielsen, N.M. and Bundgaard, H. Glycolamide Esters as Biolabile Prodrugs of Carboxylic Acid Agents: Synthesis, Stability, Bioconversion, and Physicochemical Properties, Journal of Pharmaceutical Sciences, 77(4): 285-298 (1988).
Nielsen, P.E. and Haaima, G., Peptide nucleic acid (PNA). A DNA mimic with a pseudopeptide backbone, Chem. Soc. Rev., 73-78 (1997).
Nielsen, P.E. et al., Sequence-Selective Recognition of DNA by Strand Displacement with a Thymine-Substituted Polyamide, Science, 254(5037): 1497-1500 (1991).
Nielsen, P.E. et al., Synthesis of 29-O,39-C-linked bicyclic nucleosides and bicyclic Oligonucleotides, J. Chem. Soc. Perkins Trans., 1: 3423-3433 (1997).
Nieuwlandt, D. et al., In Vitro Selection of RNA Ligands to Substance P, Biochemistry, 34: 5651-5659 (1995).
Nilsson et al., Chemical and Stereochemical Aspects of Oxidative Coupling of H-Phosphonate and H-Phosphonothioate Diesters. Reactions with N,N-,N,O and O,O-Binucleophiles, Letters in Organic Chemistry, 2(2): 188-197 (2005).
Nilsson et al., Controlling Stereochemistry During Oxidative Coupling. Preparation of Rp or Sp Phosphoramidates from One P-chiral Precursor, Chem. Commun., (22): 2566-7 (2004).
Nilsson, J. et al., Chemoselectivity in oxidative coupling of bifunctional nucleophiles with dinucleoside H-phosphonate and dinucleoside H-phosphonothioate diesters, Nucleosides, Nucleotides & Nucleic Acids, 22(5-8):1467-1469 (2003).
Nowotny, M. et al., Structure of human RNase H1 complexed with an RNA/DNA hybrid: insight into HIV reverse transcription, Mol Cell, 28(2):264-76 (2007).
Nukaga, Y. et al., Stereocontrolled Solid-Phase Synthesis of Phosphate/Phosphorothioate (PO/PS) Chimeric Oligodeoxyribonucleotides on an Automated Synthesizer Using an Oxazaphospholidine-Phosphoramidite Method, J. Org. Chem., A-J, 10 pages (Publication Date (Web): Mar. 3, 2016).
Nukaga, Y. et al., Stereocontrolled Solid-Phase Synthesis of Phosphorothioate Oligoribonucleotides Using 2′-O-(2-Cyanoethoxymethyl)-nucleoside 3′-O-Oxazaphospholiidine Monomers, Journal of Organic Chemistry, 77(18):7913-7922 (2012).
O'Connell, D. et al., Calcium-dependent oligonucleotide antagonists specific for L-selectin, Proc. Natl. Acad. Sci. USA, 93: 5883-5887 (1996).
Obika et al. Stability and structural features of the duplexes containing nucleoside analogues with a fixed N-type conformation, 2′-O,4′-C-methyleneribonucleosides, Tetrahedron Lett. 39: 5401-5404 (1998).
Obika, S. et al., Synthesis of 2′-O,4′-C-Methyleneuridine and -cytidine. Novel Bicyclic Nucleosides Having a Fixed C a ,-endo Sugar Puckering, Tetrahedron Lett., 38(50): 8735-8 (1997).
Ohgi, T. et al., A New RNA Synthetic Method with a 2′-O-(2-Cyanoethoxymethyl) Protecting Group, Organic Letters, 7(16): 3477-3480 (2005).
Ohkubo et al., Synthesis of oligodeoxyribonucleotides containing hydroxymethylphosphonate bonds in the phosphoramidite method and their hybridization properties, Tetrahedron Letters, 46(51): 8953-8957 (2005).
Oka, N. and Wada, T., Stereocontrolled synthesis of oligonucleotide analogs containing chiral internucleotidic phosphorus atoms, Chemical Society Reviews, 40(12):5829-5843 (2011).
Oka, N. et al., An oxazaphospholidine approach for the stereocontrolled synthesis of oligonucleoside phosphorothioates, Journal of the America Chemical Society, 125(27):8307-8317 (2003).
Oka, N. et al., Diastereocontrolled Synthesis of Dinucleoside Phosphorothioates Using a Novel Class of Activators, Dialkyl(cyanomethyl)ammonium Tetrafluoroborates, Journal of the American Chemical Society, 124(18):4962-4963 (2002).
Oka, N. et al., Solid-Phase Synthesis of Stereoregular Oligodeoxyribonucleoside Phosphorothioates Using Bicyclic Oxazaphospholidine Derivatives as Monomer Units, Journal of the American Chemical Society, 130(47):16031-16037 (2008).
Oka, N. et al., Stereocontrolled synthesis of dinucleoside boranophosphates by an oxazaphospholidine method, Nucleic Acids Symposium Series, (49): 131-132 (2005).
Oka, N. et al., Stereocontrolled synthesis of oligonucleoside phosphorothioates and PO/PS-chimeric oligonucleotides by using oxazaphospholidine derivaties, Nucleic Acids Symposium Series, 52: 335-336 (2008).
Oka, N. et al., Stereocontrolled Synthesis of Oligoribonucleoside Phosphorothioates by an Oxazaphospholidine Approach, Organic Letters, 11(4):967-970 (2009).
Onizuka, K. et al., Short Interfering RNA Guide Strand Modifiers from Computational Screening, J. Am. Chem. Soc., 135: 17069-17077 (2013).
Osawa, T. et al., Synthesis and Properties of the 5-Methyluridine Derivative of 3,4-Dihydro-2H-pyran-Bridged Nucleic Acid (DpNA), J. Org. Chem., 80: 10474-10481 (2015).
Ostergaard, M. et al., Rational design of antisense oligonucleotides targeting single nucleotide polymorphisms for potent and allele selective suppression of mutant Huntingtin in the CNS, Nucleic Acids Research, 41(21), 9634-9650 (2013).
Ostergaard, M.E. et al., Efficient Synthesis and Biological Evaluation of 5?-GaINAc Conjugated Antisense Oligonucleotides, Bioconjugate. Chem., 26: 1452-1455 (2015).
Otting, G. et al., Why Pentose- and Not Hexose-Nucleid Acids? Part IV. ‘Homo-DNA’: 1 H-, 13C-, 31P-, and 15N-NMR-Spectroscopic Investigation of ddGlc(A-A-A-A-A-T-T-T-T-T) in Aqueous Solution, Helvetica Chimica Acta, 76(8):2701-2756 (1993).
Padmanabhan, S. et al., Anti-HBV nucleotide prodrug analogs: Synthesis, bioreversibility, and cytotoxicity studies, Bioorganic and Medicinal Chemistry Letters, 16(15):1491-1494 (2006).
Pallan, P.S. et al., Structure and nuclease resistance of 20,40-constrained 20-O-methoxyethyl (cMOE) and 20-O-ethyl (cEt) modified DNAs, Chem. Comm., 48: 8195-8197 (2012).
Pan, Q-W. et al., New therapeutic opportunities for Hepatitis C based on small RNA, World J. Gastroenterol., 13(33): 4431-4436 (2007).
Panzara, M. et al., Duchenne Muscular Dystrophy Advisory Board Meeting, WAVE Life Sciences, 70 pages (Mar. 3, 2017).
Parmer, R. et al., 5′-(E)-Vinylphosphonate: A Stable Phosphate Mimic Can Improve the RNAi Activity of siRNA-GaINAc Conjugates, Chem. Bio. Chem., 17: 1-6 (2016).
Parrish et al., Functional Anatomy of a dsRNA Trigger: Differential Requirement for the Two Trigger Strands in RNA Interference, Molecular Cell, 6:1077-1087 (2000).
Patil et al., Syntheses and properties of oligothymidylate analogs containing stereoregulated phosphorothioate and phosphodiester linkages in an alternating manner, Bioorganic & Medicinal Chemistry Letters, 4(22): 2663-2666 (1994).
Pedersen, L. et al, A Kinetic Model Explains Why Shorter and Less Affine Enzyme-recruiting Oligonucleotides Can Be More Potent, Mol Ther Nucleic Acids, 3: e149 1-8 (2014).
Pendergraff, H.M. et al., Single-Stranded Silencing RNAs: Hit Rate and Chemical Modification, Nucleic Acid Therapeutics, 1-7 (2016).
Perrino, E. et al., New sulfurated derivatives of valproic acid with enhanced histone deacetylase inhibitory activity, Bioorganic & Medicinal Chemistry Letters, 18(6): 1893-1897 (2008).
Petersen, M. and Wengel, J., LNA: a versatile tool for therapeutics and genomics, Trends in Biotechnology, 21(2): 74-81 (2003).
Peyrottes, S. et al., SATE pronucleotide approaches: an overview, Mini-Reviews Medicinal Chemistry, 4(4):395-408 (2004).
Pfister, E.L. et al., Five siRNAs targeting three SNPs may provide therapy for three-quarters of Huntington's disease patients, 19(9): 774-778 (2009).
Pharmacology Review(s), Application No. 203568Orig1s000, Center for Drug Evaluation and Research, Food and Drug Administration, Department of Health & Human Services, 2013.
Pitsch, S. et al., Reliable Chemical Synthesis of Oligoribonucleotides (RNA) with 2′-O-[(Triisopropylsilypoxy]methyl(2′-O-tom)-Protected Phosphoramidites, Helvetica Chimica Acta, 84: 3773-3795 (2001).
Poijarvi, P. et al., 2,2-Bis(ethoxycarbonyl)- and 2-(Alkylaminocarbonyl)-2-cyano-Substituted 3-(Pivaloyloxy)propyl Groups as Biodegradable Phosphate Protections of Oligonucleotides, Bioconjugate Chemistry, 16(6):1564-1571 (2005).
Poijarvi, P. et al., The chemical stability of S-(2-acylthioethyl) and S-acyloxymethyl protected thymidylyl-3′,5′-thymidine phosphoromonothiolates and their deacylation products in aqueous solution, Nucleosides Nucleotides and Nucleic Acids, 20(1-2):77-91 (2001).
Poijarvi, P. et al., Towards Nucleotide Prodrugs Derived from 2,2-Bis(hydroxymethyl)malonate and Its Congeners: Hydrolytic Cleavage of 2-Cyano-2-(hydroxymethyl)-3-methoxy-3-oxopropyl and 3-(Alkylamino)-2-cyano-2-(hydroxymethyl)-3-oxopropyl Protections from the Internucleosidic Phosphodiester and Phosphorothioate Linkages, Helvetica Chimica Acta, 85(7):1869-1876 (2002).
Poijarvi, P. et al., Towards Oligonucleotide Pro-Drugs: 2,2-Bis(ethoxycarbonyl) and 2-(Alkylaminocarbonyl)-2-cyano Substituted 3-(Pivaloyloxy)Propyl Groups as Biodegradable Protecting Groups for Internucleosidic Phosphoromonothioate Linkages, Letters in Organic Chemistry, 1(2):183-188 (2004).
Poijarvi, P., Prodrug Approaches of Nucleotides and Oligonucleotides, Current Medicinal Chemistry, 13(28):3441-3465 (2006).
Pon, R. T., Solid-Phase Supports for Oligonucleotide Synthesis, Current Protocols in Nucleic Acid Chemistry, 3.1.1-3.1.28 (2000).
Pontarollo, R.A. et al., Monocytes are required for optimum in vitro stimulation of bovine peripheral blood mononuclear cells by non-methylated CpG motifs, Veterinary Immunology and Immunopathology, 84(1-2): 43-59 (2002).
Pontiggia, R. et al., 2-C-Methyluridine modified hammerhead riboxyme against the estrogen receptor, Bioorganic & Medicinal Chemistry Letters, 20: 2806-2808 (2010).
Pontiggia, R. et al., DNAzymes and ribozymes carrying 2′-C-methyl nucleotides, Nucleic Acids Sumposium Series, 52: 521-522 (2008).
Potter et al, Stereospecificity of nucleases towards phosphorothioate-substituted RNA: stereochemistry of transcription by T7 RNA polymerase, Nucleinc Acids Research, 15(10): 4145-4162 (1987).
Potter, B.V.L. et al., Synthesis and Configurational Analysis of Dinucleoside Phosphate Isotopically Chiral at Phosphorus. Stereochmical Course of Penicillium citrum Nuclease P1 Reaction, Biochemistry, 22: 1369-1377 (1983).
Prakash, T.P. et al., 2′-O-[2-(Methylthio )ethyl]-Modified Oligonucleotide: An Analogue of 2′-O-[2-(Methoxy)-ethyl]-Modified Oligonucleotide with Improved Protein Binding Properties and High Binding Affinity to Target RNA, Biochemistry, 41: 11642-11648 (2002).
Prakash, T.P. et al., Identification of metabolically stable 5-phosphate analogs that support single-stranded siRNA activity, Nucleic Acids Research, 43(6): 2993-3011 (2015). Supplementary Data, 80 pages.
Prakash, T.P. et al., Lipid Nanoparticles Improve Activity of Single-Stranded siRNA and Gapmer Antisense Oligonucleotides in Animals, ACS Chem. Biol., 5 pages (2013), DOI: 10.1021/cb4001316.
Prakash, T.P. et al., Synergistic effect of phosphorothioate, 50-vinylphosphonate and GaINAc modifications for enhancing activity of synthetic siRNA, Bioorg. Med. Chem. Lett., 26: 2817-2820 (2016).
Prakash, T.P. et al., Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice, Nucleic Acids Res., 42(13): 8796-807 (2014).
Prhavc, M. et al., 2′-O-[2-[2-(N,N-Dimethylamino)ethoxy]ethyl] Modified Oligonucleotides: Symbiosis of Charge Interaction Factors and Stereoelectronic Effects, Organic Letters, 5(12): 2017-2020 (2003).
Pubchem, Substance Record for SID 174316404, Available Date: Mar. 31, 2014 (retrieved on Feb. 26, 2018). Retrieved from the Internet: <URL: https://pubchem.ncbi.nlm.nih.gov/substance/174316404>.
Pubchem, Substance Record for SID 174316700, Available Date: Mar. 31, 2014 (retrieved on Feb. 26, 2018). Retrieved from the Internet: <URL: https://pubchem.ncbi.nlm.nih.gov/substance/174316700>.
Pubchem, Substance Record for SID 174316999, Available Date: Mar. 31, 2014 {retrieved on Feb. 26, 2018). Retrieved from the Internet: <URL: https://pubchem.ncbi.nlm.nih.gov/substance/174316999>.
Puri, N. et al, Targeted Gene Knockout by 2′-O-Aminoethyl Modified Triplex Forming Oligonucleotides, J. Biol. Chem., 276: 28991-28998 (2001).
Puri, N. et al., The Synthesis and Reactivity of New 2-(N,N-Diisoprophylamino)-3-Methylsulfonyl-1,3,2-Benzoxazaphospholes. The Utility of the 5-Chloro analogue in the One-Pot Synthesis of Oligothiophosphates: [ApsppA, ApspppA, ppp5′A2′ps5′A, m7GpsppA, Apspppp, Apspp], Tetrahedron 51(10): 2991-3014 (1995).
Perez, B. et al., Antisense Mediated Splicing Modulation for Inherited Metabolic Diseases: Challenges for Delivery, Nucleic Acid Therapies, 24(1): 48-56 (2014).
Rajwanshi, V.K. et al., Lna stereoisomers: xylo-LNA (b-d-xylo configured locked nucleic acid) and a-l-LNA (a-l-ribo configured locked nucleic acid), Chem. Commun., 1395-1396 (1999).
Ravikumar, V.T. et al., Unylinker: An Efficient and Scaleable Synthesis of Oligonucleotides Utilizing a Universal Linker Molecule: A Novel Approach to Enhance the Purity of Drugs, Org. Process Res. Dev., 12(3): 399-410 (2008).
Ravn, J. et al., Stereodefined LNA Phosphorthioate Oligonucleotides, Roche Pharma Research and Early Development, RTR Research, Roche Innovation Center Copenhagen, RNA & Oligonucleotide Therapeutics Meeting, Poster, 1 page (Mar. 29-Apr. 1, 2017).
Reese, C.B. and Yan, H., Solution phase synthesis of ISIS 2922 (Vitravene) by the modified H-phophane approach, J. Chem. Soc., Perkin Trans. I, 2619-2633 (2002).
Regan, J.F. et al., A Rapid Molecular Approach for Chromosomal Phasing, PLOS ONE, 1-15 (2015).
Reither, S. and Jeltsch, A., Specificity of DNA triple helix formation analyzed by a FRET assay, BMC Biochemistry, 3: 9 pages (2002).
Revankar, G. R. and Rao, T.S., DNA with Altered Bases, DNA and Aspects of Molecular Biology, Comprehensive Natural Products Chemistry, 7.09: 313-339 (1999).
Robinson, D.S. et al., Predominant TH2-Like Bronchoalveolar T-Lymphocyte Population in Atopic Asthma, The New England Journal of Medicine, 326: 298-304 (1992).
Rossetti, G., Structural aspects of the Huntingtin protein investigated by biocomputing methods, Thesis, RWTH Aachen University, Forschungszentrum Juelich, 173 pages (2011).
Rozners, E. et al., Evaluation of 2′-hydroxyl protection in RNA-synthesis using the H-phosphonate approad, Nucleic Acids Research, 22(1): 94-99 (1994).
Saetrom, P., Predicting the efficacy of short oligonucleotides in antisense and RNAi experiments with boosted genetic programming, Bioinformatics, 20(17): 3055-3063 (2004).
Sakatsume, O. et al., Solid Phase Synthesis of Oligoribonucleotides by the Phosphoramidite Approach Using 2′-O-1-(2-Chloroethoxy)Ethyl Protection, Tetrahedron, 47(41): 8717-8728 (1991).
Saneyoshi, H. et al., A General Method for the Synthesis of 2′-0-Cyanoethylated Oligoribonucleotides Having Promising Hybridization Affinity for DNA and RNA and Enhanced Nuclease Resistance, The Journal of Organic Chemistry, 70(25): 10453-10460 (2005).
Sanhueza, C.A. et al., Efficient Liver Targeting by Polyvalent Display of a Compact Ligand for the Asialoglycoprotein Receptor, J. Am. Chem. Soc., 9 pages (2016).
Schirle, N. T. and Macrae, I.J., The Crystal Structure of Human Argonaute2, Science, 336(6084): 1037-1040 (2012).
Schirle, N.T. et al., Structural analysis of human Argonaute-2 bound to a modified siRNA guide, J. Am. Chem. Soc., 1-6 (2016).
Schirle, N.T. et al., Structural Basis for microRNA Targeting, Science, 346(6209): 608-613 (2014).
Schirle, N.T. et al., Water-mediated recognition of t1-adenosine anchors Argonaute2 to microRNA targets, eLife, 4: e07646 1-16 (2015).
Schmitz, C. et al., Synthesis of P-Stereogenic Phosphoramidite and Phosphorodiamidite Ligands and Their Application in Asymmetric Catalysis, Eur. J. Org. Chem., 6205-6230 (2015).
Schoning, K.-U. et al., Chemical Etiology of Nucleic Acid Structure: The α-Threofuranosyl-(3′->2′) Oligonucleotide System, Science, 290(5495):1347-1351 (2000).
Schultz, C., Prodrugs of Biologically Active Phospate Esters, Bioorganic and Medicinal Chemistry, 11(6):885-898 (2003).
Schultz, R.G. and Gryaznov, S.M., Oligo-24-fluoro-24-deoxynucleotide N34_P54 phosphoramidates: synthesis and properties, Nucleic Acids Res., 24(15): 2966-2973 (1996).
Schulz, W.G. and Cai, S.L., Synthetic Genetics, Chemical and Engineering News, 5 (2012).
Scrimgeour, E.M. Huntington Disease (Chorea) in the Middle East, SQU. Med. J., 9(1): 16-23 (2009).
Seela et al, Diastereomerically pure Rp and Sp dinucleoside H-phosphonates. The stereochemical course of their conversion into P-methylphosphonates, phosphorothioates and [18O] chiral phosphates, Journal of Organic Chemistry, 56(12): 3861-3869 (1991).
Seidman, M.M. and Glazer, P.T. The potential for gene repair via triple helix formation, The Journal of Clinical Investigation, 112(4): 487-494 (2003).
Senn, J.J. et al., Non-CpG-Containing Antisense 2-Methoxyethyl Oligonucleotides Activate a Proinflammatory Response Independent of Toll-Like Receptor 9 or Myeloid DifferentiationFactor 88, The Journal of Pharmacology and Experimental Therapeutics, 314: 972-979 (2005).
Sergueeva et al., Synthesis of Dithymidine Boranophosphates via Stereospecific Boronation of H-phosphonate Diesters and Assignment of their Configuration, Tetrahedron Letters, 40: 2041-2044 (1999).
Seth, P., and Olson, R., Nucleic Acid Therapeutics—Making Sense of Antisesnse, 2016 Drug Design and Delivery Symposium, ACS Webinar, 1-36 (Jul. 26, 2016).
Seth, P.P. et al., An Exocyclic Methylene Group Acts as a Bioisostere of the 2′Oxygen Atom in LNA, J. Am. Chem. Soc, 132(42): 14942-14950 (2010).
Seth, P.P. et al., Configuration of the 50-Methyl Group Modulates the Biophysical and Biological Properties of Locked Nucleic Acid (LNA) Oligonucleotides, J. Med. Chem., 53: 8309-8318 (2010).
Seth, P.P. et al., Design, Synthesis and Evaluation of Constrained Methoxyethyl, (cMOE) and Constrained Ethyl (cEt) Nucleoside Analogs, Nucleic Acids Symposium Series, 52(1), 553-554 (2008).
Seth, P.P. et al., Short Antisense Oligonucleotides with Novel 2′-4′ Conformationaly Restricted Nucleoside Analogues Show Improved Potency without Increased Toxicity in Animals, J. Med. Chem., 52: 10-13 (2009).
Seth, P.P. et al., Structural requirements for hybridization at the 50-position are different in a-L-LNA as compared to b-D-LNA, Bioo. Med. Chem. Lett., 22: 296-299 (2012).
Seth, P.P. et al., Structure Activity Relationships of α-I-LNA Modified, Phosphorothioate Gapmer Antisense Oligonucleotides in Animals, Mol. Ther-Nuc. Acids., 1: e47 1-8 (2012).
Seth, P.P. et al., Synthesis and Biophysical Evaluation of 2′,4′-Constrained 2′O-Methoxyethyl and 2′,4′-Constrained 2′O-Ethyl Nucleic Acid Analogues, J. Org. Chem., 75: 1569-1581 (2010).
Sharma, V.K. et al. Antisense oligonucleotides: modifications and clinical trials, Med. Chem. Commun., 5: 1454-71 (2014).
She, X. et al., Synergy between Anti-Endoglin (CD105) Monoclonal Antibodies and TGF-β in Suppression of Growth of Human Endothelial Cells, Int. J. Cancer, 108: 251-257 (2004).
Sheehan, J.P. and Phan, T.M. Phosphorothioate Oligonucleotides Inhibit the Intrinsic Tenase Complex by an Allosteric Mechanism, Biochemistry, 40: 4980-4989 (2001).
Shivalingam, A. et al., Molecular Requirements of High-Fidelity Replication-Competent DNA Backbones for Orthogonal Chemical Ligation, J. Am. Chem. Soc., 139(4):1575-1583 (2017).
Sierzchala et al., Oxathiaphospholane Method of Stereocontrolled Synthesis of Diribonucleoside 3′, 5′-Phosphorotioates, Journal of Organic Chemistry 61(19): 6713-6716 (1996).
Silverman, R.H., A scientific journey through the 2-5A/RNase L system, Cytokine Growth Factor Reviews, 18(5-6):381-388 (2007).
Singh, P.P. et al., Universality of LNA-mediated high-affinity nucleic acid recognition, Chem. Comm., 1247-1248 (1998).
Singh, S.K. et al., Synthesis of 2′-Amino-LNA: A Novel Conformationally Restricted High-Affinity Oligonucleotide Analogue with a Handle, J. Org. Chem., 63: 10035-10039 (1998).
Singh, S.K. et al., Synthesis of Novel Bicyclo[2.2.1] Ribonucleosides: 2′-Amino- and 2′-Thio-LNA Monomeric Nucleosides, J. Org. Chem., 63: 6078-6079 (1998).
Singhrao, S.K. et al., Increased Complement Biosynthesis by Microglia and Complement Activation on Neurons in Huntington's Disease, Experimental Neurology, 159: 362-376 (1999).
Skotte, N.H. et al., Allele-specific suppression of mutant huntingtin using antisense oligonucleotides: providing a therapeutic option for all Huntington disease patients, PLoS One, 9(9): e107434 1-18 (2014).
Small, L.D. et al.,Comparison of Some Properties of Thiolsulfonates and Thiolsulfinates, Journal of the American Chemical Society, 71(10): 3565-3566 (1949).
Smith, A. et al., The murine haemopexin receptor, Biochem. J., 276: 417-425 (1991).
Sobkowski, et al. Stereochemistry of internucleotide bond formation by the H?phosphonate method. 1. Synthesis and 31P NMR analysis of 16 diribonulceoside (3′-5′)-H-phosphonates and the corresponding phosphorothioates, Nucleosides Nucleotides Nucleic Acids, 24(10-12): 1469-84 (2005).
Sobkowski, M. et al., Recent Advances in H-Phosphonate Chemistry. Part 1. H-Phosphonate Esters: Synthesis and Basic Reactions, Top Curr Chem, 361:137-177 (2014).
Sonveaux, E., Protecting Groups in Oligonucleotide Synthesis, Protocols for Oligonucleotide Conjugates, Methods in Molecular Biology, Edited by Agrawal, S., Humana Press, 26:1-71 (1994).
Sorensen, M.D., Functionalized LNA (locked nucleic acid): high-affinity hybridization of oligonucleotides containing N-acylated and N-alkylated 2′-amino-LNA monomers, Chem. Comm., 2130-2131 (2003).
Spinelli, N. et al., Use of Allylic Protecting Groups for the Synthesis of Base-Sensitive Prooligonucleotides, European Journal of Organic Chemistry, 49-56 (2002).
Sproat, B.S., RNA Synthesis Using 2′-O-(Tert-Butyldimethylsilyl) Protection, Methods in Molecular Biology, 288: 17-31 (2005).
Stawinski et al., Nucleoside H-phosphonates. 14. Synthesis of nucleoside phosphoroselenoates and phosphorothioselenoates via stereospecific selenization of the corresponding H-phosphonate and H-phosphonothioate diesters with the aid of new selenium-transfer reagent, 3H-1,2-benzothiaseleno1-3-one, J. Org. Chem., 59(1): 130-136 (1994).
Stawinski et al., Stereospecific oxidation and oxidative coupling of H-phosphonate and H-phosphonothioate diesters, Tetrahedron Letters, 33(22):3185-3188 (1992).
Stawinski, J. and Stromberg, R. Di- and Oligonucleotide Synthesis Using H-Phosphonate Chemistry, Methods in Molecular Biology, 288: 81-100 (2005).
Stawinski, J. and Thelin, M., 3-H-2,1-benzoxathiol-3-one 1-oxide—A New Reagent for Stereospecific Oxidation of Nucleoside H-Phosphonothioate Diesters, Tetrahedron Letters, 33(22): 3189-3192 (1992).
Stawinski, J. and Thelin, M., 3H-1,2-benzothiaseleno1-3-one. A new selenizing reagent for nucleoside H-phosphonate and H-phosphonothioate diesters, Tetrahedron Letters, 33(47): 7255-7258 (1992).
Stec, W.J. and Zon, G., Stereochemical Studies of the Formation of Chiral Internucleotide Linkages by Phosphormadite COupling in the Synthesis of Oligodeocyribonucleotides, Tetrahedron Letters, 25(46): 5279-5282 (1984).
Stec, W.J. et al., Automated Solid-Phase Synthesis, Separation, and Stereochemistry of Phosphorothioate Analogues of Oligodeocyribonucleotides, J. Am. Chem. Soc., 106: 6077-6079 (1984).
Stec, W.J. et al., Deoxyribonucleoside 3′-O-(2-Thio- and 2-Oxo-“spiro”-4,4-pentamethylene-1,3,2-oxathiaphospholane)s:? Monomers for Stereocontrolled Synthesis of Oligo(deoxyribonucleoside phosphorothioate)s and Chimeric PS/PO Oligonucleotides, J. Am. Chem. Soc., 120(29): 7156-7167 (1998).
Stec, W.J. et al., Diastereomers of Nucleoside 3′-O-(2-Thio-1,3,2-oxathia(selena)phospholanes): Building Blocks for Stereocontrolled Synthesis of Oligo(nucleoside phosphorothioate)s, Journal of the American Chemical Society, 117(49):12019-12029 (1995).
Stec, W.J. et al., Novel route to oligo(deoxyribonucleoside phosphorothioates). Stereocontrolled synthesis of P-chiral oligo(deoxyribonucleoside phosphorothioates), Nucleic Acids Research, 19(21):5883-5888 (1991).
Stec, W.J. et al., Stereocontrolled Synthesis of Oligo (nucleoside phosphorothioate)s , Angew. Chem. Int. Ed. Engl., 33:709-722 (1994).
Stec, W.J. et al., Stereodependent inhibition of plasminogen activator inhibitor type 1 by phosphorothioate oligonucleotides: proof of sequence specificity in cell culture and in vivo rat experiments, Antisense Nucleic Acid Drug Dev., 7(6):567-73 (1997).
Stec, W.J. et al., Stereospecific Synthesis of P-Chiral Analogs of Oligonucleotides, Methods in Molecular Biology, 20: 285-313 (1993).
Stec, W.J., Oligo(nucleoside Phosphorothioate)s: The Quest of P-Chirality, in Phosphorus, Sulfur, and Silicon, 177(6): 1775-1778 (2002).
Stein, C.A. and Cheng, Y.C., Antisense oligonucleotides as therapeutic agents—is the bullet really magical?, Science, 261(5124):1004-12 (1993).
Stout, A.K. et al., Inhibition of wound healing in mice by local interferon a/b injection, Int J Exp Pathol, 74 (1): 79-85 (1993).
Sureshbabu, V.V. et al., Synthesis of tetrazole analogues of amino acids using Fmoc chemistry: isolation of amino free tetrazoles and their incorporation into peptides, Tetrahedron Letters, 48(39): 7038-7041 (2007).
Surono, A. et al., Chimeric RNA/Ethylene Bridged Nucleic Acids Promote Dystrophin Expression in Myocytes of Duchenne Muscular Dystrophy by Inducing Skipping of the Nonsense Mutation-Encoding Econ, Human Gene Therapy, 15:749-757 (2004).
Suska, A. et al., Antisense oligonucleotides: Stereocontrolled synthesis of phosphorothioate oligonucleotides, Pure and Applied Chemistry, 65(4):707-714 (1993).
Suter, S.R. et al., Structure-Guided Control of siRNA Off Target Effects, J. Am. Chem. Soc., 1-9 (2016).
Swayze, E.E. and Bhat, B., The medicinal chemistry of oligonucleotides, Crooke, S.T. (ed) Antisense Drug Technology: Principles, Strategies, and Applications, CRC Press, Boca Raton, FL: 143-82 (2007).
Swayze, E.E. et al., Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals, Nucleic Acids Research, 35(20: 687-700 (2007).
Takahashi, D. et al., Novel diphenylmethyl-Derived Amide Protecting Group for Efficient Liquid-Phase Peptide Synthesis: AJIPHASE, Org. Lett., 14(17): 4514-4517 (2012).
Takahashi, T. et al., Interactions between the non-seed region of siRNA and RNA-binding RLC/RISC proteins, Ago and TRBP, in mammalian cells, Nucleic Acids Research, 42(8): 5256-5269 (2014).
Takeno, H. et al., Selection of an RNA Molecule that Specifically Inhibits the Protease Activity of Subtilisin, J. Biochem., 125: 1115-1119 (1999).
Takeshima, Y. et al., Oligonucleotides against a splicing enhancer sequence led to dystrophin production in muscle cells from a Duchenne muscular dystrophy patient, Brain & Development, 23:788-790 (2001).
Tam, Journal of Hematotherapy & Stem Cell Research, 12: 467-471 (2003).
Tamura et al., Preparation of Stereoregulated Antisense Oligodeoxyribonucleoside Phoshorothioate and Interaction with its Complementary DNA and RNA, Nucleosides & Nucleotides,17(1-3): 269-282 (1998).
Tang, J. et al., Enzymatic Synthesis of Stereoregular (All Rp) Oligonucleotide Phosphorothioate and Its Properties, Nucleosides Nucleotides, 14(3-5):985-990 (1995).
Tawarada, R. et al., Mechanistic studies on oxidative condensation of a thymidine 3′-H-phosphonate derivative with 3′-O-acetylthymidine, Archive for Organic Chemistry, (3):264-273 (2009).
Thayer, J.R. et al., Separation of oligonucleotide phosphorothioate distereoisomers by pellicular anion-exchange chromatography, Journal of Chromatography A, 1218: 802-808 (2011).
Tomoskozi et al., Stereospecific conversion of H-phosphonates into phosphoramidates. The use of vicinal carbon-phosphorus couplings for configurational determination of phosphorus, Tetrahedron, 51(24): 6797-6804 (1995).
Tosquellas, G. et al., First synthesis of alternating SATE-phosphotriester/phosphodiester prooligonucleotides on solid support, Bioorganic and Medicinal Chemistry Letters, 8(20): 2913-2918 (1998).
Tosquellas, G. et al., Prooligonucleotides exhibit less serum-protein binding than phosphodiester and phosphorothioate oligonucleotides, Nucleosides, Nucleotides and Nucleic Acids, 19(5-6):995-1003 (2000).
Tosquellas, G. et al., The pro-oligonucleotide approach: solid phase synthesis and preliminary evaluation of model pro-dodecathymidylates, Nucleic Acids Research, 26(9):2069-2074 (1998).
Tosquellas, G. et al., The Prooligonucleotide Approach III: Synthesis and bioreversibility of a chimeric phosphorodithioate prooligonucleotide, Bioorganic and Medicinal Chemistry Letters, 6(4):457-462 (1996).
Tosquellas, G. et al., The Prooligonucleotide Approach IV : Synthesis of chimeric prooligonucleotides with 6 enzymolabile masking groups and unexpected desulfurization side reaction, Bioorganic and Medicinal Chemistry Letters, 7(3):263-268 (1997).
Ts'o, P.O. et al., An Approach to Chemotherapy Based on Base Sequence Information and Nucleic Acid Chemistry, Ann. N. Y. Acad. Sci., 507: 220-241 (1988).
Tsai, C.H. et al., Enzymatic synthesis of DNA on glycerol nucleic acid templates without stable duplex formation between product and template, Proceedings of the National Academy of Science, 104(37):14598-14603 (2007).
Tuerk, C. and Gold, L., Systematic Evolution of Ligans by Exponential Enrichment: RNA Ligands to Bacteriophage T4 DNA Polymerase, Science, 249: 505-510 (1990).
Turner, D.H. et al, Improved Parameters for Prediction of RNA Structure, Cold Spring Harbor Symposia on Quantitative Biology, LII: 123-133 (1987).
Turner, D.H. et al., Free Energy Increments for Hydrogen Bonds in Nucleic Acid Base Pairs, J. Am. Chem. Soc., 109: 3783-3785 (1987).
U.S. Food and Drug Administration, Development of New Stereoisomeric Drugs, 8 pages (May 1, 1992). URL: http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm122883.htm [Retrieved Jun. 15, 2016].
Umemoto, T et al., Oligoribonucleotide Synthesis by the use of 1-(2-cyanoethoxy)ethyl (Cee) as a 2′-hydroxy protecting group, Tetrahedron Letters 45: 9529-9531 (2004).
Uphoff, K.W. et al., in vitro selection of aptamers: the death of pure reason, Curr. Opin. Struct. Biol., 6: 281-288 (1996).
Usman, N et al., Automated Chemical Synthesis of Long Oligoribonucleotides Using 2′-O-Siylylated Ribonucleoside 3′-O-Phosphoramidites on a Controlled-Pore Glass Support, J. Am. Chem. Soc. 109(25): 7845-7854 (1987).
Uznanski, B. et al., Stereochemistry of base-catalyzed ring opening of 1,3,2-oxathiaphospholanes. Absolute configuration of 2-{N-[(Rc)-1-(.alpha.-naphthyl)ethyl]amino}-2-thiono-1,3,2-oxathiaphospholanes and O,S-dimethyl N-[(Rc)-1-(.alpha.-naphthypethyl]phosphoramidothioates, Journal of the American Chemical Society, 114(26):10197-10202 (1992).
Van Aerschot, A. et al., 1,5-Anhydrohexitol Nucleic Acids, a New Promising Antisense Construc, Angew. Chem. Int. Ed. Engl., 34: 1338-1339 (1995).
Van Der Veken, P. et al., Irreversible inhibition of dipeptidyl peptidase 8 by dipeptide-derived diaryl phosphonates, Journal of Medicinal Chemistry, 50(23): 5568-5570 (2007).
Van Deutekom, J.C.T. et al., Antisense-induced exon skipping restores dystrophin expression in DMD patient derived muscle cells, Human Molecular Genetics, 10(15):1547-1554 (2001).
Vasquez, K.M. et al., Chromosomal mutations induced by triplex-forming oligonucleotides in mammalian cells, Nucl. Acids Res. 27(4): 1176-1181 (1999).
Vasseur, J-J. et al., Oligonucleosides: Synthesis of a Novel Methylhydroxylamine-Linked Nucleoside Dimer and Its Incorporation into Antisense Sequences, J. Am. Chem. Soc., 114: 4006-4007 (1992).
Veedu, R.N. et al., Novel Applications of Locked Nucleic Acids, Nucleic Acids Symposium Series, 51: 29-30 (2007).
Verhagen et al., A Conformationally locked Aminomethyl C-Glycoside and Studies on Its N-Pyren-1-ylcarbonyl Derivative Inserted into Oligodeoxynucleotides, European Journal of Organic Chemistry, 2538-2548 (2006).
Verma, S. and Eckstein, F., Modified Oligonucleotides: Synthesis and Strategy for Users, Annu. Rev. Biochem., 67: 99-134 (1998).
Vermeulen, A. et al., Double-Stranded Regions Are Essential Design Components of Potent Inhibitors of RISC Function, RNA, 13: 723-730 (2007).
Vives, E. et al., Lipophilic pro-oligonucleotides are rapidly and efficiently internalized in HeLa cells, Nucleic Acids Research, 27(20):4071-4076 (1999).
Vlassov, V.V. et al., Transport of oligonucleotides across natural and model membranes, Biochimica et Biophysica Acta, 1197: 95-108 (1994).
Vu, H. and Hirschbein, B.L., Internucleotide Phosphite Sulfurization With Tetraethylthiuram Disulfide. Phosphorothioate Oligonucleotide Synthesis Via Phosphoramidite Chemistry, Tetrahedron Letters, 32(26):3005-3008 (1991).
Vuyisich, M. and Beal, P.A., Regulation of the RNA-dependent protein kinase by triple helix formation, Nuc, Acids Res., 28(12): 2369-74 (2000).
Wada et al., Stereocontrolled Synthesis of Phosphorothioate RNA by the Oxazaphospholidine Approach, Nucleic Acids Symp. Ser., 48: 57-58 (2004).
Wada, T. et al., Chemical synthesis and properties of stereoregulated phosphorothioate RNAs, Nucleic Acids Symposium Series, 51:119-120 (2007).
Wada, T. et al., Stereocontrolled synthesis of phosphorothioate DNA by an oxazaphospholidine approach, Nucleic Acids Research Supplement, 3:109-110 (2003).
Wada, Takeshi, Chapter I Development of nucleic acid medicines, 3.3 Chemical synthesis of phosphorous atom-modified nucleic acids, CMC Publication., Fronteir of Development of Nucleic Acid Medicine: 67-75 (2009).
Wagner, C.R. et al., Pronucleotides: toward the in vivo delivery of antiviral and anticancer nucleotides, Medicinal Research Reviews, 20(6):417-451 (2000).
Walker, J.R. et al., Structure of the Ku heterodimer bound to DNA and its implications for double-strand break repair, Nature, 412: 607-614 (2001).
Wan et al., Synthesis of Second Generation Antisense Oligonucleotides Containing Chiral Phosphorothioate Linkages and Evaluation of their Biophysical Properties and Biological Activity, 10th Annual Meeting of the Oligonucleotide Therapeutics Society, abstract received by Applicant Oct. 7, 2014, poster setup prior to presentation (first known to Applicant late Oct. 12, 2014, PST), poster presentation Oct. 13, 2014.
Wan, W.B. and Seth, P.P., The Medicinal Chemistry of Therapeutic Oligonucleotides, J. Med. Chem., 59: 9645-9667 (2016).
Wan, W.B. et al., Synthesis, biophysical properties and biological activity of second generation antisense oligonucleoties containing chiral phosphorothioate linkages, Nucleic Acid Research, 42: 13456-13468 (2014). Supplementary Information, 14 pages.
Wang H, et al., Therapeutic gene silencing delivered by a chemically modified siRNA against mutant SOD 1 slows ALS progression, The Journal of Biological Chemistry, 283(23):15845-15852 (2008).
Wang, J.-C. et al., A stereoselective synthesis of dinucleotide phosphorothioate triesters through a chiral indol-oxazaphosphorine intermediate, Tetrahedron Letters, 38(5):705-708 (1997).
Wang, Y. et al., Structure of an argonaute silencing complex with a seed-containing guide DNA and target RNA duplex, Nature, 456(7224): 921-926 (2008).
Warby, S.C. et al., CAG expansion in the Huntington disease gene is associated with a specific and targetable predisposing haplogroup, Am. J. Hum. Genet., 84(3): 351-366 (2009).
Watts, J.K. and Corey, D.R., Gene silencing by siRNAs and antisense oligonucleotides in the laboratory and the clinic, J. Pathol. 226(2): 365-79 (2012).
WAVE Life Sciences Press Release, WAVE Life Sciences Added to the Russell 2000® Index, 2 pages (Jun. 27, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Announces Plan to Deliver Six Clinical Programs by 2018, 6 pages (Jan. 29, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Announces Pricing of Initial Public Offering, 3 pages (Nov. 11, 2015).
WAVE Life Sciences Press Release, WAVE Life Sciences Appoints Dr. Michael Panzara as Head of Neurology Franchise, 4 pages (Jul. 12, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Appoints Keith Regnante as Chief Financial Officer, 4 pages (Aug. 17, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Appoints Roberto Guerciolini, M. Senior Vice President and Head of Early Development, 2 pages (Apr. 7, 2015).
WAVE Life Sciences Press Release, WAVE Life Sciences Closed $18 Million Series a Financing to Advance Stereopure Nucleic Acid Therapeutics, 3 pages (Feb. 2, 2015).
WAVE Life Sciences Press Release, WAVE Life Sciences Enters Collaboration with Pfizer to Develop Genetically Targeted Therapies for the Treatment of Metabolic Diseases, 5 pages (May 5, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Expands Stereopure Synthetic Chemistry Platform Capabilities, Augments Patent Portfolio with Addition of Single-Stranded RNAi (ssRNAi), 3 pages (Jun. 8, 2015).
WAVE Life Sciences Press Release, WAVE Life Sciences Raises $66 Million in Series B Financing, 3 pages (Aug. 18, 2015).
WAVE Life Sciences Press Release, WAVE Life Sciences Receives Orphan Drug Designation from Fda for its Lead Candidate Designed to Treat Huntington's Disease, 5 pages (Jun. 21, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Reports First Quarter 2016 Financial Results and Provides Business Update, 9 pages (May 16, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Reports Fourth Quarter and Full Year 2015 Financial Results and Provides Business Update, 10 pages (Mar. 30, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences Reports Second Quarter 2016 Financial Results and Provides Business Update, 10 pages (Aug. 15, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences to Advance Next-Generation Nucleic Acid Therapies to Address Unmet Need in Duchenne Muscular Dystrophy, 6 pages (May 9, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences to Present at the Deutsche Bank 41st Annual Health Care Conference, 2 pages (Apr. 29, 2016).
Wave Life Sciences Press Release, Wave Life Sciences to Present at the Jefferies 2016 Healthcare Conference, 2 pp. (Jun. 1, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences to Present at the Jmp Securities Life Sciences Conference, 2 pages (Jun. 15, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences to Present at the Leerink Partner 5th Annual Global Healthcare Conference, 2 pages (Feb. 3, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences to Present at the Leerink Partners Rare Disease & Immuno-Oncology Roundtable, 2 pages (Sep. 14, 2016).
WAVE Life Sciences Press Release, WAVE Life Sciences to Present at the SunTrust Robinson Humphrey 2016 Orphan Drug Day Conference, 2 pages (Feb. 16, 2016).
Weidner, J.P. et al., Alkyl and Aryl Thiolsulfonates, Journal of Medicinal Chemistry, 7(5): 671-673 (1964).
Weiner, G. J. et al., Immunostimulatory oligodeoxynucleotides containing the CpG motif are effective as immune adjuvants in tumor antigen immunization, 94(20): 10833-10837 (1997).
Weinfeld, M., et al., Influence of nucleic acid base aromaticity on substrate reactivity with enzymes acting on single-stranded DNA, Nucleic Acids Res., 21(3): 621-626 (1993).
Weiser, T.G., et al., An estimation of the global volume of surgery: a modeling strategy based on available data, Lancet, 372(9633): 139-144 (2008).
Welz et al., 5-(Benzylmercapto)-1 H-tetrazole as activator for 2′-O-TBDMS phosphoramidite building blocks in RNA synthesis, Tetrahedron Letters, 43: 795-797 (2002).
Wengel, J., Synthesis of 3′-C- and 4′-C-Branched Oligodeoxynucleotides and the Development of Locked Nucleic Acid (LNA), Ace. Chem. Res., 32: 301-310 (1999).
Whittaker, B. et al., Stereoselective synthesis of highly functionalised P-stereogenic nucleosides via palladium-catalysed P-C cross-coupling reactions, Tetrahedron Letters, 49: 6984-6987 (2008).
Widdison, W. C. et al., Semisynthetic Maytansine analogues for the targeted treatment of cancer, Journal of Medicinal Chemistry, 49(14): 4392-4408 (2006).
Wild, E. et al., Quantification of mutant huntingtin protein in cerebrospinal fluid from Huntington's disease patients, The Journal of Clinical Investigation, 125(5): 1979-1986 (2015).
Wilk, A. and Stec, W.J., Analysis of oligo(deoxynucleoside phosphorothioate)s and their diastereomeric composition, Nucleic Acids Research, 23(3):530-534 (1995).
Wilk, A. et al., Deoxyribonucleoside Cyclic N-Acylphosphoramidites as a New Class of Monomers for the Stereocontrolled Synthesis of Oligothymidylyl- and Oligodeoxycytidylyl-Phosphorothioates, Journal of the American Chemical Society, 122(10): 2149-2156 (2000).
Wong, Chui Ming, Synthesis of anisomycin. Part I. The stereospecific synthesis of N-benzoyl-2-(p-methoxybenzyl)-3-hydroxy-4-carboxamido pyrrolidine and the absolute configuration of anisomycin, Canadian journal of Chemistry 46: 1101-1104 (1968).
Woolf, T.M. et al., Specificity of antisense oligonucleotides in vivo, Prov. Natl. Aca. Sci. USA, 89: 7305-7309 (1992).
Wright, P. et al., Large scale synthesis of oligonucleotides via phosphoramidite nucleosides and a high-loaded polystyrene support, Tetrahedron Letters, 34(21):3373-3736 (1993).
Written Opinion for PCT/IB2009/007923, 8 pages (dated Sep. 6, 2010).
Written Opinion for PCT/IB2015/000395, 10 pages (dated Oct. 30, 2015).
Written Opinion for PCT/JP11/55018, 3 pages (dated Mar. 29, 2011).
Written Opinion for PCT/JP11/71559, 6 pages (dated Dec. 20, 2011).
Written Opinion for PCT/JP15/50716 and English Translation, 11 pages (dated Apr. 21, 2015).
Written Opinion for PCT/JP2010/065900, 5 pages (dated Sep. 15, 2010).
Written Opinion for PCT/JP2013/004303, 6 pages (dated Aug. 13, 2013).
Written Opinion for PCT/JP2015/050714, and English Translation, 11 pages (dated Apr. 21, 2015).
Written Opinion for PCT/JP2015/050718 and English Translation, 6 pages ( dated Apr. 21, 2015).
Written Opinion for PCT/US2010/041068, 11 pages, (dated Sep. 1, 2010).
Written Opinion for PCT/US2011/064287, 14 pages (dated Apr. 12, 2012).
Written Opinion for PCT/US2012/046805, 9 pages (dated Sep. 19, 2012).
Written Opinion for PCT/US2013/050407, 12 pages (dated Jan. 9, 2014).
Written Opinion for PCT/US2016/043542, 14 pages (dated Dec. 28, 2016).
Written Opinion for PCT/US2016/043598, 10 pages (dated Nov. 28, 2016).
Written Opinion for PCT/US2016/056123, 15 pages (dated Mar. 17, 2017).
Written Opinion for PCT/US2017/022135, 11 pages (dated Jun. 6, 2017).
Written Opinion for PCT/US2017/030753, 13 pages (dated Sep. 26, 2017).
Written Opinion for PCT/US2017/030777, 10 pages (dated Oct. 2, 2017).
Written Opinion for PCT/US2017/035837, 15 pages (dated Aug. 24, 2017).
Written Opinion for PCT/US2017/043431, ISA/US, 38 pages (dated Dec. 21, 2017).
Written Opinion for PCT/US2017/045218, 11 pages (dated Sep. 27, 2017).
Written Opinion for PCT/US2017/055601, ISR/US, 16 pages (dated Feb. 15, 2018).
Written Opinion for PCT/US2017/062996, 9 pages (dated Mar. 9, 2018).
Wu, X. et al., Synthesis of 5′-C- and 2′-O-(Bromoalkyl)-Substituted Ribonucleoside Phosphoramidites for the Post-synthetic Functionalization of Oligonucleotides on Solid Support, Helvetica Chimica Acta, 83: 1127-1144 (2000).
Xiang, Y. et al., Effects of RNase L mutations associated with prostate cancer on apoptosis induced by 2′,5′-oligoadenylates, Cancer Research, 63(20):6795-6801 (2003).
Xiong, H.Y. et al., The human splicing code reveals new insights into the genetic determinants of disease, Science, 347(6218): 144 1254806-1-1254806-8 (2015).
Xu, D. and Esko, J.D., Demystifying Heparan Sulfate-Protein Interactions, Annu. Rev. Biochem., 83: 129-157 (2014).
Xu, L. et al., Cyclic ADP-ribose analogues containing the methylenebisphosphonate linkage: effect of pyrophosphate modifications on Ca2+ release activity, J. Med. Chem., 48(12): 4177-4181 (2005).
Xu, Y. et al., Functional comparison of single- and double-stranded siRNAs in mammalian cells, Biochemical and Biophysical Research Communications, 316: 680-687 (2004).
Yamada, O. et al., Diastereoselective Synthesis of 3,4-Dimethoxy-7-morphinanone: A Potential Route to Morphine, Organic Letters, 2(18): 2785-2788 (2000).
Yamakage, S-i. et al., 1-(2-Chloroethoxy)Ethyl Group for the Protection of 2′-Hydroxyl Group in the Synthesis of Oligoribonucleotides, Tetrahedron Letters, 30(46): 6361-6364 (1989).
Yamamoto, S. et al., Unique Palindromic Sequences in Synthetic Oligonucleotides are Required to Induce INF and Augment INF-Mediated Natural Killer Activity, J. Immunol., 148(12): 4072-4076 (1992).
Yamato, K. et al., Enhanced specificity of HPV16 E6E7 siRNA by RNA-DNA chimera modification, Cancer Gene Therapy, 18: 587-597 (2011).
Yanai, H. et al., Suppression of immune responses by nonimmunogenic oligodeoxynucleotides with high affinity for high-mobility group box proteins (HMGBs), PNAS Early Edition, 1-6 (2011).
Yasuda, K. et al., CpG motif-independent activation of TLR9 upon endosomal translocation of “natural” phosphodiester DNA, European Journal of Immunology, 431-436 (2006).
Ye, S. et al., An efficient procedure for genotyping single nucleotide polymorphisms, Nucleic Acids Research, 29(17): e88 1-8 (2001).
Yu, D. et al., Accessible 5′-end of CpGcontaining phosphorothioate oligodeoxynucleotides is essential for immunostimulatory activity, Bioorganic & Medicinal Chemistry Letters, 10: 2585-2588 (2000).
Yu, D. et al., Single-Stranded RNAs Use RNAi to Potently and Allele-Selectively Inhibit Mutant Huntingtin Expression, Cell, 150: 895-908 (2012).
Yu, D. et al., Stereo-Enriched Phosphorothioate Oligodeoxynucleotides: Synthesis, Biophysical and Biological Properties, Bioorganic & Medicinal Chemitry, 8: 275-284 (2000).
Yu, R.Z. et al., Cross-species comparison of in vivo PK/PD relationships for second-generation antisense oligonucleotides targeting apolipoprotein B-100, Biochem. Pharmacol., 77: 910-919 (2009).
Yu, S. et al., A One-Pot Formal [4+2] Cycloaddition Approach to Substituted Piperidines, Indolizidines, and Quinolizidines. Total Synthesis of Indolizidine (-)-209I, Journal of Organic Chemicals, 70:7364-7370 (2005).
Zhang, J. et al., Optimization of Exon Skipping Therapies for Duchenne Muscular Dystrophy, WAVE Life Sciences, PPMD: Parent Project Muscular Dystrophy Meeting, Orlando, FL, Poster, 1 page (Jul. 25, 2016).
Zhang, L. et al., A simple glycol nucleic acid, Journal of the American Chemical Society,127(12):4174-4175 (2005).
Zhang, R.S. et al., Synthesis of two mirror image 4-helix junctions derived from glycerol nucleic acid, Journal of the American Chemical Society, 130(18):5846-5847 (2008).
Zhang, Y. et al., Structural Isosteres of Phosphate Groups in the Protein Data Bank, J. Chem. Inf. Model, 1-18 (2017).
Zhang, Y., Investigating phosphate structural replacements through computational and experimental approaches, Academic Dissertain, University of Helsinki, 119 pages (2014).
Zhao, J. et al., Genome-wide Identification of Polycomb-Associated RNAs by RIP-seq, Molecular Cell, 40: 939-953 (2010).
Zhong, Z. et al., WAVE Life Sciences: Developing Stereopure Nucleic Acid Therapies for the Treatment of Genetic Neurological Diseases, World CNS Summit 2017, Boston, MA, WAVE Life Sciences, Poster, 1 page (Feb. 20-22, 2017).
Zlatev et al., Phosphoramidate dinucleosides as hepatitis C virus polymerase inhibitors, J Med Chem., 51(18): 5745-57 (2008).
Zlatev, I. et al., 5′-C-Malonyl RNA: Small Interfering RNAs Modified with 5′-Monophosphate Bioisostere Demonstrate Gene Silencing Activity, ACS Chem. Biol., 8 pages (2015).
Zon, Automated synthesis of phosphorus-sulfur analogs of nucleic acids-25 years on: potential therapeutic agents and proven utility in biotechnology, New J. Chem., 34(5): 795-804 (2010).
Zon, G and Stec, W.J., Phosphorothioate oligonucleotides, Oligonucleotides and Analogues: A Practical Approach, 87-108 (1991).
ALS Association, The ALS Association and the Packard Center Partner to Develop Animal Model Systems for Most Common Cause of Familial Als, 4 pages (Mar. 1, 2012). URL: http://www.alsa.org/news/archive/new-animal-model-systems.html [Retrieved Dec. 14, 2017].
CAS Registry File RN 121563-98-2; Chemical Abstracts Accession No. 1989:450484, 2 pages (2018).
CAS Registry No. 1223431-57-9, Chemical Abstracts Accession No. 2000:10625, 2 pages (2018).
Donnelly, C.J. et al., M1415. Development of C9orf72 ALS Biomarkers and Therapeutics, Annals of Neurology, 72 (suppl 16): S67-S68 (2012).
Krishna, H. et al., Alkynyl Phosphonate DNA: A Versatile “Click”able Backbone for DNA-Based Biological Applications, J. Am. Chem. Soc., 134: 11618?11631 (2012).
Lee, K.-W. et al., CG sequence- and phosphorothioate backbone modification-dependent activation of the NF-κB-responsive gene expression by CpG-oligodeoxynucleotides in human RPMI 8226 B cells, Molecular Immunology, 41: 955-964 (2004).
Liu, S. et al., Evaluation of protective effect of multi-epitope DNA vaccine encoding six antigen segments of Toxoplasma gondii in mice Parasitol Res, 105:267-274 (2009).
Madsen, A., Antisense Against C90RF72, MDA/ALS News Magazine, 2 pages (Jul. 1, 2012). URL: http://alsn.mda.org/article/antisense-against-c90rf72 [Retrieved Dec. 14, 2017].
Martinez, J.M.L. et al, NMR Characterization of Hydrate and Aldehyde Forms of Imidazole-2-carboxaldehyde and Derivatives, Journal of Organic Chemistry, 75: 3208-3213 (2010).
Nishina, K. et al., DNA/RNA heteroduplex oligonucleotide for highly efficient gene silencing, Nature Communications, 6:7969, pp. 1-13 (2015).
Renton, A.E. et al., A Hexanucleotide Repeat Expansion in C9ORF72 Is the Cause of Chromosome 9p21-Linked ALS-FTD, Neuron 72, 257-268 (Oct. 20, 2011).
Sha, S.J. and Boxer, A., Treatment implications of C9ORF72, Alzheimer's Research & Therapy, 4(46): 7 pages (2012).
Simon-Sanchez, J. et al., The clinical and pathological phenotype of C9ORF72 hexanucleotide repeat expansions, Brain, 135: 723-735 (2012).
Tulic, M.K. et al Amb a 1-immunostimulatory oligodeoxynucleotide conjugate immunotherapy decreases the nasal inflammatory response, J. Allergy Clin. Immunol., 235-241 (2004).
Related Publications (1)
Number Date Country
20180222936 A1 Aug 2018 US
Provisional Applications (1)
Number Date Country
61509526 Jul 2011 US
Continuations (2)
Number Date Country
Parent 15222910 Jul 2016 US
Child 15941494 US
Parent 14233579 US
Child 15222910 US