CA2110988C - Sequence-specific binding polymers for duplex nucleic acids - Google Patents

Sequence-specific binding polymers for duplex nucleic acids

Info

Publication number
CA2110988C
CA2110988C CA002110988A CA2110988A CA2110988C CA 2110988 C CA2110988 C CA 2110988C CA 002110988 A CA002110988 A CA 002110988A CA 2110988 A CA2110988 A CA 2110988A CA 2110988 C CA2110988 C CA 2110988C
Authority
CA
Canada
Prior art keywords
base
polymer
subunit
sequence
subunits
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002110988A
Other languages
French (fr)
Other versions
CA2110988A1 (en
Inventor
James E. Summerton
Dwight D. Weller
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sarepta Therapeutics Inc
Original Assignee
Antivirals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Antivirals Inc filed Critical Antivirals Inc
Publication of CA2110988A1 publication Critical patent/CA2110988A1/en
Application granted granted Critical
Publication of CA2110988C publication Critical patent/CA2110988C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/548Phosphates or phosphonates, e.g. bone-seeking
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6839Triple helix formation or other higher order conformations in hybridisation assays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds

Abstract

The present invention describes a polymer composition effective to bind in a sequence-specific manner to a target sequence of a duplex polynucleotide at least two different-oriented Watson/Crick basepairs at selected positions in the target sequence. The composition includes an uncharged backbone with 5- or 6-membered cyclic backbone structures and selected bases attached to the backbone structures effective to hydrogen bond specifically with different oriented base-pairs in the target sequence. Also disclosed are subunits useful for the construction of the polymer composition. The present invention also includes methods for (i) coupling a first free or polymer-terminal subunit, and (ii) isolating, from a liquid sample, a target duplex nucleic acid fragment having a selected sequence of base-pairs.

Description

FOR DUPLEX NUChEIC ACIDS
1. Field of the Invention The present invention relai~es to an uncharged polymer capable of binding with sequence specificity to double stranded nucleic acids containing a selected base-pair sequence.
l0 2. References Aboderin, Delpierre, and Fruton, J. Amer. Chem. Soc.
1965, 87, 5469 Aoyama (1987). Bull. Chem. Soc. Jpn. 60 2073.
Arnott & Bond (1973). Science 181 68; Nature New Biol. 244 99.
Arnott & Selsing (1974). J. Molec. Biol. 88 509.
Balgobin, McBride, Kierzek, Beaucage and Caruthers Bassingdale (1986). J. Amer. Claem. Soc. 108 2040.
Barwolff and Langen, in "Nucleic Acid Chemistry,"
Townsend and Tipson, Ed. Wiley, New York, 1978 page 359.
Belikova, Zaratova, & Grincwa (1967). Tet. Letters 37 3557.
Bischofberger, Tetrahedron Letters (1987) 28 2821.
Bredereck, et al, Chemisch~e Berichte (1968) 101 41.
Bunemann et al. (1981). Biochem. 20 2864.
finalize\asc\0450-4.sub WO 93,1Q(13~2 'CT/LS92/05208 Carnelley and Dutt, J. Chem. Soc. 125, 2483.
Chamberlin -& Patterson (1965). J. Molec. Biol. 12 410.
Chelsky et al. (1989). Mol. Cell. Biol. 9 2487.
Cooney et al. (1988). Science 241 456.
-Corey, Gilman, and Ganem (1968). J. Am. Chem. Soc. 90 5616.
Elguero et al. (1976). The Tautomerism of Hetero-cycles, Adv. in Heterocyclic Chew" Supplement I. Academic Press. NY.
Fischer-Fantuzzi & Vesco (19813). Molec. & Cell. Biol.
_8 5495.
Flavell & Van den Berg (1975). FEBS Letters ~ 90.
Gregoriadis & Needunjun (19?5). Biochem. Biophys. Res.
Commun. 65 537.
Himmelsbach and Pfleiderer (1983). Tet. Lett. 24 3583.
Hoffer (1960). Chemische Berichte ~3_ 2777.
~Hoogsteen (1959). Acta Cryst. ~? 822.
Inman (1964). J. Mol. Biol. ~0 137.
Jones (1979). Int. J. Biol. Macromolec. ~, 194.
Jurgens (1907). Chemische Be.richte 40 4409.
Kabanov (1989). FEES Letters X58 343.
Kamimura, Tsuchiya, Urakami, R;oura, Sekine, Shinozaki, Miura and Hata (1984). J. Amer. C:hem. Soc. x,06 4552.
Karpova et al. (1980). FEBS Letters ~2_ 21.
Katritzky and Yates (1976). J. Chem. Soc.., Perkin Trans . ~, 3 09 .
King, McWhirter, and Barton (:1945). J. Am. Chem. Soc.
_67 2089.
Kosturko et al. (1979). Biochem. ~8_ 5751.
Kundu & Heidelberger (1974). Biochem. Biophys. Res.
Comm. 60 561.
Kundu et al. (1975). J. Med. Chem. 38 395 & 399.
Kundu (1980). J. Med. Chem. ;?3 512.
Lemaitre; Bayard & Leblue (1987). PNAS 84 648.
Maeba -et al (1983). J. Org. Chem. 48 2998.
Mahler, Wold, Dervan (1989). Science 245 725.
C

WO 93/00352 PC1'/US92/05208 21 109 8a :3 Miller et al. (1979). Biochemistry ~8_ 5134.
Miller et al. (1980). J. Biol. Chem. 255 9659.
Miller et al. (1985). Biochimie 67 769.
Miura and Hata (1984). J. Amer. Chem. Soc. ~ 4552.
Morgan & Wells (1968). (:f. Molec. Biol. 37 63.
Moser & Dervan (1987). Science ~3$ 645.
Myers and Lee (1984). C:arb. Res. ~ 61.
Ozdowska (1974). Rocz. Chem. ~ 1065.
Pelaprat et al. (1980) .J. Med. Chem. ~ 1330, 1336.
Peltier (1956). Belg. Soc. Science, Bretagne 31 26.
Phillips (1928). J. Chsam. Soc. 2393.
Pickering, Srivastava, Hlitkowski, and Robins, Nucleic Acid Chemistry, Part 1, Ed. by Townsend and Tipson, John Wiley and Sons, New York, p :145.
Pitha & Pitha (1970). B:iopolymers ~ 965.
Poisel and Schmidt (15175). Chemische Berichte 108 2547.
Povich (1989). J. Amer. Chem. Soc. ~ 3059.
Rich & Seeman (1975). Handbook of Biochemistry and Molecular Biology, 3rd Edition, Vol. 2, pages 465-466.
Robins, Naik, and Lee (1974). J. Org. Chem. 39 1891.
Robins, Hansske, Bernie:r (1981). Can. J. Chem. 59 3360.
Sakore et al. (1969). .J. Molec. Biol. 43 385.
Schmitz & Galas (1979). Nucleic Acids Res. 6 111.
Schnneller and Christ (1981). J. Heterocyclic Chem.
~, 654.
Schultz, Taylor, & Derv,an (1982). JACS 104 6861.
Schultz & Dervan (1983). PNAS 80 6834.
Sekine, Peshakova, Hata, Yokoyama and Miyazawa (1987).
J. Org. Chem. 52 5061.
Shioiri, Ninomiya, Yamada (1972). J. Amer. Chem. Soc.
94 6203.
Sluka et al. (1987). Science 238 1129.

WO 93/00352 'CI~/US92/05208 a~ ~ 09 ea Smith, Rammler, Goldberg and Khorana (1961). Amer.
J.

Chem. Soc. 84 430.

Stirchak, Summerton, & Welle:r (1987). J. Chem.
Org.

52 42 02.

Stirchak, Summerton, & Welle:r (1989). NucleicAcids Res. ~7 6129.

Summerton & Bartlett (1978b). J. Molecular Biology 145-162.

Summerton (1979). J. Theor. 13io1. 78 61-76.

~ummerton (1979). J. Theor. l3iol. 78 77-99.

Tamura and Okai (1984). Carb. Res. ,~33 207.

Toulme et al. (1986). PNAS 83 1227.

Trattner, et al (1964). J. Org. Chem. 29 2674.

Trichtinger, Charbula and Pfleiderer (1983). Tet.

Lett. 24 711.

Voet & Rich (1970). Progress in Nucleic Acids Res. &

Molec.
Biol.

- 265.

Youngquist & Dervan (1985). ;7ACS X07 5528.

Zamecnik & Stephenson (1978). PNAS 75 280.

Zuidema, Van den Berg & Flave:ll (1978). NucleicAcids Res. 5 2471.
3. Background of the Invention Oligonucleotides or oligonucleotide analogs designed to inactivate selected single-stranded genetic sequences unique to a target pathogen were first reported in the late 1960's by Belikova, 1967, and subsequently by: Pitha, 1970;
Summerton, 1978 .b, 1979a,b; Zamecnik, 1978; Jones, 1979;
Karpova, 1980; Miller, 1979, 198 0, 1985; Toulme, 1986;
Stirchak, 1987, 1989. Polymer~.c agents of this type achieve their sequence specificity by exploiting Watson/Crick base pairing between the agent and its comple-mentary single-stranded target genetic sequence. Because such polymers only bind single-stranded target genetic se-quences, they are of limited value where the genetic infor-C

~1 109 88 mation one wishes to inactivate exists predominantly in the double-stranded state.
For many pathogens and pathogenic states duplex gene tic sequences offer a more suitable target for blocking 5 genetic activity. One of the: earliest attempts to develop a sequence-specific duplex-directed nucleic acid binding agent was reported by Kundu,, Heidelberger, and coworkers during the period 1974 to 1980 (Kundu 1974; Kundu 1975;
Kundu 1980). This group reported two monomeric agents, each designed to hydrogen-bond to a specific base-pair in duplex nucleic acids. However, these agents were ineffec-tive, probably for two reasons. First, they utilized a nonrigid ambiguous hydrogen-bonding group (an amide) which can act as either a proton donor or acceptor (in the hydrogen-bonding sense). Secondly, they provided an insufficient number of hydrogen bonds (two) for complex stability in aqueous solution. Experimental results from a variety of systems suggest l;.hat hydrogen-bonded complexes are stable in aqueous solution only if there are a substan-tial number (probably at lea:~t 12) of cooperative intermo-lecular hydrogen bonds, or if there are additional stabi-lizing interactions (electrostatic, hydrophobic, etc.).
Another early attempt was reported by Dattagupta and Crothers at Yale and coworkers in Germany (Kosturko 1979;
Bunemann 1981). These workers employed a polymer prepared from a dye known to intercalate into duplex DNA rich in G:C
base-pairs and another dye which preferentially binds to duplex DNA rich in A:T base-pairs, probably via minor-groove sites. Preparation of the polymer involved modification of the two dyes lby adding acrylic moieties and then polymerization of a mixaure of the modified dyes in the presence of duplex DN.A of defined sequence (the template). The expectation was that the resultant polymer would show a specific affinity for duplex DNA having the same sequence as the template: DNA. However, such material 6 ' proved to exhibit only nominal sequence specificity. A
variety of bis-intercalating agents designed to bind to specif is sequences in duplex DNA have also been reported (Pelaprat, 1980), but such agents inherently give only minimal sequence specificity.
More recently, Dervan ha,s taken a natural B-form-spec-ific minor-groove-binding antibiotic (Distamycin) and systematically extended its structure to achieve a signif i-cant level of sequence specificity (Schultz 1982; Schultz 1983; Youngquist 1985). &e has also appended to this oligomer an EDTA/Fe complex which under certain conditions acts to cleave the duplex target sequence near the agent's binding site. However, this particular approach will not lead to the high level of specificity which is needed for therapeutic applications because the inherent symmetry of the H-bonding sites in the minor groove provides too little sequence information.
Still more recently, Dexvan and coworkers reported a binding agent which utilizea the informationally-richer polar major-groove sites of a target genetic duplex for sequence-specific recognition (Sluka 1987). This entailed adapting a synthetic polyp~eptide, comprising the DNA-sequence-recognition portion of a DNA-binding protein, for cleaving DNA at the protein's binding site on duplex DNA.
The cleaving activity was achieved by linking an EDTA/Fe complex to the amino terminu;a of the synthetic peptide and demonstrating that this comp:Lex selectively cleaved duplex DNA at or near the parent protein's natural target sequence.
Another approach to duplex targeting has grown out of studies ffirst reported in the late 1950's that demonstrat-ed, via X-ray diffraction, that under high salt conditions an all-thymine or all-uracil polynucleotide can bind to specific polar major-groove sates on a Watson/Crick genetic duplex having all adenines inione strand and all thymines 2~ 109 as or uracils in the other strand (Hoogsteen 1959). Subse-quently, it was reported that in high salt and at pH values lower than 7, an all-cytosine polynucleotide, having the cytosine moieties protonated, can bind in a similar manner to a Watson/Crick duplex having all guanines in one strand and all cytosines in the other strand.
Thereafter, it was demonstrated that under high salt conditions and at a pH below 7, a polynucleotide containing both cytosines and thymines (or uracils) can bind to a Wat-son/Crick duplex having the appropriate sequence of purines in one strand and pyrimidines in the other strand (Morgan, 1968).
In the 1970's this Hoogsteen binding mechanism was exploited for affinity chromatography purification of duplex genetic fragments containing runs of purines in one strand and pyrimidines in the other strand (Flavell, 1975;
Zuidema, 1978). In 1987 Dervan and coworkers exploited this Hoogsteen binding mechanism to position an all-pyrimidine polynucleotide, carrying an EDTA/Fe cleaving moiety, onto a target genetic duplex having a specific sequence of purines in one strand and pyrimidines in the other strand (Mosey, 1987).
A major-groove binding mode different from the Hoog steen mode was reported in the mid-1960's and involves binding of an all-purine polynucleotide, poly(dI), to a poly(dI)/poly(rC) duplex (Inman 1964) and to a poly(dI)/
poly(dC) duplex (Chamberlin 1965). Similarly, a mostly-purine polynucleotide has been recently used by Hogan and coworkers (Gooney, 1988) for blocking the activity of a selected natural duplex genetic sequence. These workers reported that in the presence of 6 mM Mg++ a mostly-purine polynucleotide (24 purines, 3 pyrimidines) of a specific sequence inhibits transcription of the human C-myc gene in a cell-free system.

To date, reported polynucleotides used for binding to genetic duplexes fail to satisfy one or more important criteria for effective use within living organisms. First, the Hoogsteen-binding polynucleotides (polypyrimidines) containing cytosines require a lower-than-physiological pH
in order to achieve effective binding (due to the necessity of protonating the cytosine moieties), although it has recently been demonstrated by Dervan and coworkers that the use of 5-methylcytosines in place of cytosines allows Hoogsteen binding at a pH somewhat closer to physiological pH (Mahler, 1989), and use of both 5-methylcytosines in place of cytosines and 5-bromouracils in place of thymines (or uracils) improves binding still further (Povsic, 1989).
Secondly, in the case of polypurine polynucleotides, both inosine (hypoxanthine) and adenine moieties lack adequate sequence specificity and adequate binding affinity for effective major-groove binding in intracellular appli cations. The inadequate sequence specificity for inosine (Inman, 1964) and adenine (Gooney, 1988) moieties derives from the fact that inosine can bind with similar affinity to the central polar major-groove sites of both a C:I (or C:G) base-pair (i.e., NH4 of C and 06 of G or I) and an A:T
or A: U base-pair ( i . a . , NH6 of A and 04 of T or U) , and because adenine can bind with similar affinity to the central polar major-groove sites of both a T:A or U:A base-pair (i.e., 04 of T or U and NH6 of A) and a G:C base-pair (i.e., 06 of G and NH4 of C), as discussed further below.
The low binding affinity of inosine for its target base-pairs and of adenine for its target base-pairs is due to the fact that these purines can form only two less-than optimal hydrogen-bonds to the major-groove sites of their respective target base-pairs.
Thirdly, both polypyrimidine and polypurine polynu cleotides fail to achieve effective binding to their target genetic duplexes under physiological conditions, due to the ..._~_ ~_.~...._~. _.__ ~.._~

substantial electrostatic repulsion between the three closely-packed polyanionic backbones of the three-stranded complexes. Although this repulsion can be attenuated by high salt (Morgan, 1968), divalent cations (Gooney, 1988), or polyamines (Mosey, 1987), nonetheless, for applications in living cells, and particularly cells within intact organisms, control of intracellular cation concentrations is generally not feasible.
In addition, for therapeutic applications polynucleot ides are less than optimal because: they are rapidly sequestered by the reticu.loendothelial lining of the capillaries, they do not readily cross biological mem branes, and they are sensitive to degradation by nucleases in the blood and within cells.
Finally, for many in vivo applications of sequence-specific duplex-directed nucleic acid-binding agents, the principal target is DNA, which appears to exist within cells predominantly in a B or B-like conformation. In this context, polynucleotides which .have been used for major-groove binding to genetic duplexes (Mosey, 1987; Gooney, 1988) have a unit backbone length which is shorter than optimal for binding to duple:K genetic sequences existing in a B-type conformation.
4. Summary of the Invention The present invention .includes a polymer composition effective to bind in a sequence-specific manner to a target sequence of a duplex polynucleotide containing at least two different oriented Watson-Crick base-pairs at selected positions in the target sequence. The polymer is formed of a specific sequence of subunits selected from the following forms i.
2110~~$
Y 5, O R ~
Y 5, O R i 4' 1' 3'. . 2' 5 O R~ 3, N
where Y is a 2- or 3-atom length, uncharged intersubunit linkage group; R' is H, OH, or O-alkyl; the 5'-methylene has a ~ stereochemical orientation in the 5-membered ring 10 and a uniform stereochemical orientation in the 6-membered ring; R~ has a ~ stereochemical orientation; and at least about 70% of R~ groups in the polymer are selected from two or more of the following base-pair-specificity groups:
(a) for a T:A or U:A oriented base-pair, R~ is 2,6-diamino purine; (b) for a C:G oriented base-pair R~ is guanine or 6-thioguanine; (c) for a G:C oriented base-pair, R~ is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B indicates the polymer backbone:
B
N
HN / B HN B
H N H N HN N N
H * H * H H
where the * ring position may carry a hydrogen-bond acceptor group; such as a carbonyl oxygen; and (d) for an A:T or A:U oriented base-pair, R~ is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B
indicates the polymer backbone:
* B
H N H
N~ B N B H
H N H N N N N
H * H H

1.1 where the * ring position may carry a hydrogen-bond donat-ing group, such as NHz.
In one embodiment, for u.se in sequence-specific bind ing to a duplex nucleic acid :sequence in an A conformation, the Y linkage group is two .atoms in length. In another embodiment, for use in sequence-specific binding to a B-form DNA-DNA duplex nucleic acid sequence, the Y linkage group is three atoms in length.
In another aspect, the invention includes a method for coupling a first free or po7lymer-terminal subunit having one of the following subunit forms:
HO OH OH
where R; is a planar ring structure having two or more hydrogen-bonding sites, with a second free or polymer-terminal subunit having one of the following subunit forms:
H H HNH HNH HNH
N I I I
Z O Ri Z O Ri Z O Rt Z O Rt HO ''OH HO OH HO N OH
OH
where Z is a 2-atom or 3-atom long moiety. The method includes i) oxidizing the l:irst subunit to generate a dialdehyde intermediate; ii) contacting the dialdehyde intermediate with the second subunit under conditions effective to couple a primary amine to a dialdehyde; and (iii) adding a reducing agent. effective to give a coupled structure selected from the following fonas:
... p R ~ ... O
HO OH

WO 93/00352 PC1'/US92/05208 '2l~pg~g ... O p i ... O p i ... O p i N N
N I I
Z O p~ Z O p~ Z O
HO OOH N
HO OH
OH
In still another aspect, the invention provides a method for isolating, from a liquid sample, a target duplex nucleic acid fragment having a selected sequence of base-pairs. The method includes first contacting the sample with a polymer reagent containing structure which allows isolation of the reagent from solution, and attached to this structure, a polymer composition of the type described above, where the polymer composition has a subunit sequence effective to bind in a sequence-specific manner with the selected sequence of base-pairs. The contacting is carried out under conditions effective for sequence-specific binding of the polymer composition to the selected sequence of base-pairs.
Further, the polymers of the present invention can be used to detect the presence of a target nucleic acid sequence. For example, a support-bound polymer composition can be contacted with a test solution containing the selected duplex genetic sequence under conditions effective for sequence-specific binding of the polymer composition to its target sequence of base-pairs. The support-bound polymer with bound selected duplex genetic sequence is then separated from the test solution. The presence of the polymer/target duplex sequence is then detected. Detecting the selected genetic sequence may, for example, utilize one of the following: fluorescent compounds, such as, ethidium bromide and propidium iodide, effective to intercalate into duplex genetic sequences; or reporter moieties linked to ~1 10!9 88 oligocationic moieties effective to bind to the polyanionic backbone of nucleic acids.
Also forming part of the invention is a subunit compo sition for use in forming a polymer composition effective to bind in a sequence specific manner to a target sequence in a duplex polynucleotide. The composition includes one of the following subunit structures:
(a) (b) (c) (d) Y O Ri y O Ri HNH HNH
I
Z O H~ Z
O Ri O R' HO OH HO OH
OH
where R' is H, OH, or O-alkyl; the 5'-methylene has a stereochemical orientation in subunit forms (a), (c), and (d) and a uniform stereochemical orientation in subunit form (b); X is hydrogen or a protective group or a linking group suitable for joining the subunits in any selected order into a linear polymer; Y is a nucleophilic or electrophilic linking group suitable for joining the subunits in any selected order into a linear polymer; and X and Y together are such that when two subunits of the subunit set are linked the resulting intersubunit linkage is 2 or 3 atoms in length and uncharged; Z is a 2-atom or 3-atom long moiety; and, R~, which may be in the protected state and has a ~ stereochemical orientation, is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B indicates the aliphatic backbone moiety:

N B
HN--~ 8 HN--~ B
H N H N HN N N
H * I~ * H H
where the * ring position may carry a hydrogen-bond acceptor group; or, where Ft~ is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B
indicates the aliphatic backbone moiety:
*
B
HN 'N B HN ' 8 H
H N H IN N N N
H * H H
where the * ring position may carry a hydrogen-bond donating group.
Another embodiment of t:he present invention includes a method for inhibiting the biological activity of a selected duplex genetic sequence. In this method, a suitable target sequence of base-pairs is selected within the selected duplex genetic sequence whose activity is to be inhibited. A polymer composition, as described above, is provided which is effective to bind in a sequence-specific manner to the target sequence. The polymer composition is contacted with the selected duplex genetic sequence under substantially physiological conditions.
This method may further include contacting the polymer composition with the selected genetic sequence where contacting the polymer composition with the selected genetic sequence entails targeting the polymer composition to a tissue or site containing the selected genetic sequence to be inactivated. Some methods of delivery include delivering the polymer composition in the form of an aerosol to the respiratory tract of a patient and/or injecting an aqueous solution of the polymer composition into a patient.
5 These and other objects and features of the present invention will become more fully apparent when the follow-ing detailed description of i~he invention is read in conjunction with the accompanying drawings.
10 Brief Description of the Drawingvs Figures lA-iD illustrate T:A (lA), A:T (iB), C:G (iC) and G:C (1D) oriented Watson-Crick base-pairs, showing the major-groove hydrogen-binding sites of the base-pairs (arrows);
15 Figures 2A and 2B illustrate tautomeric forms of 2-amino pyrimidine (2A), and 2-~pyrimidinone (2B);
Figures 3A and 3B illustrai~e rigid (3A) and non-rigid (3B) hydrogen-bonding arrays;
Figures 4A-4D illustrate :standard positioning for a U:A base-pair in an A conformation and the approximate position of the helical axis for an A-form duplex (4A), the use of this positioning scheme for assessing R,, 8,, and A
values for a subunit base hydrogen bonded to the polar major-groove sites of a U:A basES-pair in an A conformation (4B), the standard positioning for a T:A base-pair in a B
conformation and the approximate position of helical axis for a B-form duplex (4C), and the use of this positioning scheme for assessing Rb, Bb, and A values for a subunit base hydrogen bonded to the polar major-groove sites of a T:A
base-pair in a B conformation (~4D);
Figures 5A-5C show representative 2'-deoxyribose (5A), ribose (5B), and ribose-derived backbone structures (5C) suitable for use in forming the polymer of the invention;

211U9~~
:L 6 Figures 6A-6F show repreasentative morpholino backbone structures suitable for use in forming the polymer of the invention;
Figures 7A-7E show red>resentative acyclic backbone structures suitable for forming the polymer of the invention;
Figure 8A shows a representative coupled acyclic backbone structure with a 4-atom unit backbone length, 8B-8C show coupled acyclic backbone structures with a 5-atom unit backbone length, and 8D-8E show coupled acyclic backbone structures with a 6-atom unit backbone length.
Figures 9A-9D show reapresentative coupled cyclic backbone structures with a 6-atom unit backbone length, and 9E-9F show representative: coupled cyclic backbone structures with a 7-atom unit backbone length;
Figures l0A and lOB illustrate a guanine base and its binding to a C:G oriented Waiaon-Crick base-pair (l0A) and a diaminopurine base and ita binding to a T:A oriented Watson-Crick base-pair (lOB);
Figures 11A and 11B show hydrogen bonding of a cytosine base to a G:C (ilA) and T:A (11B) oriented base-pair;
Figures 12A and 12B show hydrogen bonding of a uracil base to an A:T (12A) and C:G (12B) oriented base-pair;
Figures 13A-13D illustrate the general skeletal ring structure, hydrogen bonding array, and backbone attachment position of a tautomeric base: designed for binding to a G:C
or A:T Watson-Crick base-pair (13A), and three specific embodiments of the 13A structure (13B-13D);
Figures 14A and 148 show the hydrogen bonding of the Figure 13B structure to a G:C: (14A) and A:T (14B) oriented base-pair;
Figures 15A-15D illustrate the general skeletal ring structure, hydrogen bonding array, and backbone attachment position of a base designed for binding to a G:C Watson Crick base-pair (15A), and three specific embodiments of the Figure 15A structure (1-°iB-15D);
Figure 16 shows the hydrogen bonding of the Figure 15D
structure to a G:C oriented base-pair;
Figure 17A illustrate.: the general skeletal ring structure, hydrogen bonding array, and backbone attachment position of a base designed for binding to a G:C Watson Crick base-pair, and Figure 17B shows a specific embodiment of the 17A structure hydrogen bonded to a G:C oriented base-pair;
Figure 18A-18D illustrate the general skeletal ring-structure, hydrogen bonding .array, and backbone attachment position of a base designed for binding to an A: T or A: U
Watson-Crick base-pair (18A), and three specific embodiments of the 18A structure (18B-18D);
Figure 19 shows the hydrogen bonding of the Figure 18D
structure to an A:T oriented base-pair;
Figure 20 illustrates t:he coupling cycle used in an exemplary solid-phase synthesis of one embodiment of this binding polymer;
Figure 21 illustrates a segment of a polymer constructed according to the invention, and designed to bind to a region of an A-form genetic duplex having the sequence of base-pairs: C:G, A:T, T:A, and G:C.
Figure 22 illustrates the coupling cycle in a novel method for assembling nucleic: acid-binding polymers.
Detailed Description of the Invention I. Polvmer Subunit Construction The polymer of the invention is designed for binding with base-pair specificity to a selected sequence (the target sequence) in a strand of duplex nucleic acid. As used herein, duplex sequence refers to a sequence of contiguous oriented Watson/Crick base-pairs, where the four oriented base-pairs are: A:T (or A:U), T:A (or U:A), G:C, 21 109 88 , , .
1.B
and C:G, where A, T, U, G,, and C, refer to adenine, thymine, uracil, guanine, and cytosine nucleic acid bases, respectively.
The polymer is formed of subunits, each of which comprises a cyclic backbone structure and linkage group, which collectively form an uncharged backbone, and a base attached to the cyclic backbone structure, which provides base-pair-specific hydrogen-bonding to the target. The requirements of the backbone .structure, linkage group, and attached base in the polymer subunits are detailed below.
In the context of these duplex binding polymers, the term "base" refers to planar base-pair-specific hydrogen-bonding moieties.
A. Subunit Base Reauirements.
Because of the symmetr;Y of the polar minor-groove sites and the asymmetry of polar major-groove sites in Watson/Crick base-pairs, to achieve a given level of sequence specificity a minor-groove-binding agent would have to recognize twice as many base-pairs as would a corresponding major-groove-binding agent. Accordingly, hydrogen-bonding of the subun.it base is to the polar sites in the major groove of the target duplex.
Figures lA-1D shows T:A, A:T, C:G, and G:C oriented Watson/Crick base-pairs, with the major-groove hydrogen bonding sites indicated by arrows in the figure. For the T:A and A:T oriented base-pairs, the polar major-groove sites include the N7 and a hydrogen on the N6 of adenine and the 04 of thymine (or uracil). For the C:G and G:C
oriented base-pairs, the polar major-groove sites include the 06 and N7 of guanine a.nd a hydrogen on the N4 of cytosine.
In order to make a significant contribution to the free energy of binding and t:o provide adequate base-pair specificity, the subunit base should form at least two hydrogen bonds to its target base-pair. That is, each subunit base in the polymer should contain at physiological pH a hydrogen-bonding array suitable for binding to two or three of the polar major-groove sites on its respective oriented target base-pair. Table 1 shows the hydrogen bonding arrays comprising the: polar major-groove sites for each of the four oriented Watson/Crick base-pairs, and the corresponding hydrogen-bonding array of the subunit base suitable for hydrogen-bonding to said polar major-groove sites.
Tab:Le 1 Oriented hydroaen-bonding Reauired hydrocxen-bonding base-pair array of base-paid array of subunit base A:T ** H ** N X ( N X N
H ** H ** H
T:A ** H ** X N , N X N
** H H ** H
C:G H ** ** N N , X N N
H H ** H H
G:C ** ** H N N , N N X
H H H H **
In the table, X is generally an N, O, or S atom, but can also be F, C1, or Br, having a non-bonded pair of electrons suitable for hydrogen bonding, and ** represents the nonbonded pair of eleci~rons suitable for hydrogen bonding.

2~ ~ o~ 8s As indicated above, the polymer subunit base should contain the specified hydrogen-bonding array at physio-logical pH (in contrast to the case for cytosine moieties used for Hoogsteen-type major-groove binding). This 5 assures that at physiological pH, binding of the subunit base makes a substantial contribution to the free energy of binding between the polymer and its target duplex.
At physiological pH the subunit base should be predo minantly non-ionized. More specifically, basic moieties 10 should have pKb values of at least 7.5 or greater, and acidic moieties should have pKa values of at least 7.7 or greater. This lack of substantial ionic charge provides two advantages. First, for applications in living cells, the lack of ionic groups on the binding polymers facili-15 tates passage of the polymer across biological membranes.
Second, lack of negative charges avoids the problem of charge repulsion between the binding polymer and the negatively charged phosphates of its target duplex.
Major-groove hydrogen-bonding arrays of the four ori-20 ented Watson/Crick base-pairs are illustrated in Table 2.
Table 2 C:G H ** ** G:C ** ** H
A:T(U) ** H ** T(U):A ** H **

In Table 2, H is a hydrogen bound to a nitrogen, and ** is an electron pair of nitrogen or oxygen available for hydrogen bonding.
The respective positioning of the base-pair H-bonding arrays shown in Table 2, which approximates their relative positions in the major-groove of a duplex genetic sequence, illustrates the fact that two of the H-bonding sites of a 21 109 88 ~,~
2.1 C:G base-pair (NH4 and 06) a:re positioned nearly the same as two of the H-bonding sites of an A:T(U) base-pair (NH6 and 04). Likewise, two of the hydrogen-bonding sites of a G:C base-pair (06 and NH4) a:re positioned nearly the same as two of the H-bonding sites of a T(U):A base-pair (04 and NH6). Because of these similarities in positioning between central hydrogen-bonding sitEa of the oriented base-pairs, subunit bases which hydrogen-bond only to the polar sites near the center of the major-groove (underlined in the above table) lack adequate specificity for a given base-pair. Accordingly, in order for a subunit base to achieve high specificity for a single oriented base-pair, the base should hydrogen bond to the N7 of its respective target base-pair.
If a subunit base is to bind to only one of the four oriented Watson/Crick base-pairs, the tautomeric state of that subunit base should be sufficiently fixed under conditions of use so that at least two of the hydro-gen-bonding groups positioned for base-pair binding will not tautomerize to give a structure capable of H-bonding with comparable affinity to~ a base-pair other than the intended one. To illustrate, Figure 2A shows an acceptably fixed structure (2-amino pyrimidine, which exists almost exclusively in the 2-amino tautomeric fona). Figure 2B
shows a second structure which lacks specificity for a single base-pair due to its facile tautomerization under physiological conditions (2-pyrimidinone). Dominant tautomeric forms of a wide assortment of representative heterocyclic structures have been tabulated in a book edited by Elguero, Marzin, Katritzky & Linda (1976).
The subunit bases should have structures which provide a relatively rigid arrangement of at least two of the base-pair H-bonding groups positioned for base-pair bind-ing. Such rigidity is best afforded by a ring structure wherein at least two of the polar hetero atoms to be in-~~ ~~ 09 sa w volved in H-bonding to the target base-pair are either part of the ring or directly attached to the ring. To illus-trate, Figure 3A shows a structure (2-amino-3-cyano pyr-role) which satisfies this rigidity requirement. Figure 3B
shows a structure (2-carboxamide pyrrole) which fails to satisfy the requirement.
The simplest sequence-specific binding polymers are those which bind to a target which is composed of contigu-ous base-pairs in the polynucleotide duplex. This, in turn, requires that the subunit bases of the binding poly-mer be no thicker than the target base-pairs to which they are to bind. Accordingly, each subunit structure should be planar. This is best achieved by using subunit bases having aromatic character and/or having plane trigonal bonding for most or all ring atoms.
B. Subunit-Binding Constraints Considering now the geometric requirements of the polymer subunits, most duplex nucleic acids adopt either of two general conformations. RNA/RNA and RNA/DNA duplexes adopt an A-type conformation. DNA/DNA duplexes adopt a B-type conformation, but can readily convert to an A confor-mation under certain conditions, such as high salt or low polarity solvent.
In duplex nucleic acids the polar major-groove sites on each of the Watson/Crick base-pairs are fairly regularly positioned with respect to corresponding arrays of major-groove sites on neighboring base-pairs, with the relative positions being defined by the helical conformation para-meters of axial position, axial rise, and axial rotation.
In principle, the backbone attachment positions of the different subunit bases, when the bases are hydrogen-bonded to their respective target base-pairs, need not be posi-tioned in any regular way relative to their target base-pairs. However, when there is significant variability in ~1 109 88 the relative backbone attachnnent positions of the different subunit bases relative to their target base-pairs, each of the backbone structures of 'the component subunits in the polymer must be custom tailored with respect to backbone length and position of subun.it base attachment, leading to extremely high development and production costs.
However, if all of the subunit bases of a given sub-unit set have similar backbone attachment positions and angles relative to their respective target base-pairs, then all subunits of the set can have identical backbone struc-tures, greatly simplifying the synthetic effort required for polymer construction. '.~o this end, the polymer sub-units used in the present invention are selected, according to criteria described below, to have similar backbone attachment positions and angles.
To understand what is meant by similar backbone posi-tions and angles, reference is made to Figure 4A, which shows a Watson/Crick base-pair (W/C bp) positioned relative to the helical axis (denoted H~) of an A-form genetic duplex, i.e., (A, 12, 0.326) RNA. The lower horizontal line in the figure connects the two ribose C1' atoms of the Watson-Crick base-pair, and i~he vertical line (denoted PB) is the perpendicular bisector' of the first-mentioned line.
The backbone attachment position and angles of a sub unit base are then determinEad by positioning the subunit base on its corresponding target base-pair in this stan dardized position, with the subunit base being hydrogen bonded to the appropriate polar major-groove sites on the Watson-Crick base-pair, as shown for a 2,6-diaminotriazine subunit base in Figure 4B.
The backbone attachment: position of the subunit base, relative to its A-form target duplex, can then be described by an R) and 8, value, where R~ is the radial distance, in angstroms, from the helical. axis of the A-form target duplex to the center of the backbone atom (denoted B) to 21 109 88 t which the subunit base is attached, and 8, is the angle, in degrees, about this helical axis, measured clockwise from the perpendicular bisector to the center of the afore-mentioned backbone atom. The attachment angle, A, is defined as the angle, in degrees, measured clockwise from the perpendicular bisector, between the perpendicular bisector and a line parallel to the bond between the subunit base and the backbone moiety.
Figure 4B illustrates R~, 9~, and A parameters for a 2,6-diaminotriazine subunit base hydrogen-bonded to a U:A
base-pair in an A conformation. Figure 4C illustrates a correspondingly positioned base-pair of a H-form duplex, and Figure 4D illustrates Rb, 9b, and A parameters for this 2,6-diaminotriazine subunit base hydrogen-bonded to a T:A
base-pair in a B conformation.
In order to unambiguously define the target base-pair for a selected subunit base with a given backbone attach-ment site, two orientations for each Watson/Crick base-pair in the target duplex must be considered. The resultant 4 oriented base-pairs are designated as A:T, T:A, C:G, and G:C (and corresponding base-pairs where U replaces T). The orientations of these base-pairs are defined in Table 3.
Table 3 Oriented Base-pair 8 value for N7 of Purine Designation of Target Base-pair A:T (A; U) > 180°
T:A (U: A) < 180°
C:G < 180°
G: C > 180°
In principle, the backbone attachment position for any given subunit base, in position on its target base-pair, can have a 8 value, X°, in the range of 0° to 180°. By flipping the target base-pair, the 8 value of that same target-bound subunit base is changed to 360° - X°. The convention used in the following discussion is that the 8 value for each subunit base of the binding polymer is less 5 than 180°.
Thus, in the context of selecting a subunit set suitable for assembling the binding polymers disclosed herein, to explicitly define which orientation of a given base-pair constitutes the targE~t for a specified subunit 10 base, it is important to design<~te the orientation of that target base-pair such that the backbone attachment position of the base-pair-bound subunit x>ase has a B value less than 180°. To illustrate, a 2,6-diaminotriazine subunit base having a backbone moiety attached through the C4 of the 15 triazine (Figure 4B) can bind i:o a U:A base-pair in an A
conformation to give a B value of 28°. When this same subunit base is hydrogen-bonded to that same base-pair in the base-pair's opposite orientation (ie., A:U), the B
value for the subunit base is ?;32° (ie. , 360° - 28°) .
The 20 convention used herein dictates that the target base-pair for this subunit base is U:A (where B is < 180°), and not A:U (where 8 is > 180°).
Acceptable values of R, 8, and A for prospective recognition moieties can be readily obtained with CPK
25 molecular models (The Ealing Corp., South Natick, Mass., USA). Slightly more accurate ~ralues can be estimated by optimization of the hydrogen-bonding in the subunit base/
base-pair triplex via a computer molecular mechanics program, such as are available commercially. The subunit bases should be so selected that for a given subunit set (the set of subunits used in assembly of a given polymer) all have R values within about 2 angstroms of each other, B values within about 20° of each other, and A values within about 3 0° of each other .

In order for a subunit base to have a high specificity for only one of the oriented base-pairs, it is important that the subunit base not be able to bind to a given base-pair in both orientations (eg., G:C and C:G) simply by rotation of the subunit base about its linkage to its backbone structure. Therefore, the earlier-described backbone attachment position or angle should be asymmetrical with respect to the C1' positions of the target base-pair. Specifically, 9a for the subunit base should have a value greater than about 10°, or the attachment angle, A, for the: subunit base should have a value greater than about 25°.
C. Backbone Structure Constraints This section considers the backbone structure con-straints for a selected subunit set. Principally, the structure should be joinable in any selected order to other subunit structures via uncharged linkages having the gene-ral properties discussed in Section D below. Further, the subunit backbone structures and linkages must provide pro-per spacing and allow correct: orientation and positioning of their respective subunit bases for effective binding of the subunit bases to their rEapective oriented base-pairs in the target duplex sequence..
A principal requiremeni~ for the subunit backbone structure and linkage is that it provide a means for join-ing the subunits in essentially any specified order. This requirement can be satisfied by structures containing either heterologous or homologous linking groups. Iietero-logous type backbone moieties contain a nucleophilic group (N) on one end and an electro;philic group (E) on the other end, as illustrated below.
N_______E;
The preferred functional groups for Ithe N component include primary and secondary amine, hydrazine, hydroxyl, 2~ 21 109 88 sulfhydryl, and hydroxylamine. The preferred functional groups for the E component include the following acids and derivatives thereof: carboxylic:, thiocarboxylic, phospho-ric, thiophosphoric, esters, thioesters, and amides of phosphoric and thiophosphoric, phosphonic and thiophos-phonic, and sulfonic acid. Other suitable E groups include aldehyde, dialdehyde (or vicinal hydroxyls suitable for conversion to a dialdehyde), alkyl halide, and alkyl tosylate.
Homologous type backbone moieties can be of two types, one type having nucleophilic end groups and the other type having electrophilic end group:; or, a single homologous backbone moiety can be alternated with an appropriate link-er. These alternatives are illustrated below:
N-------N alternated with E-------E
N-------N alternated with E linker N linker alternated with E-------E
Preferred functional group:~ for N and E are as in the heterologous backbone moietie:~. Preferred E linkers include carbonyl; thiocarbonyl; alkyl, ester, thioester, and amide of phosphoryl and thiophosphoryl; phosphonyl and thiophosphonyl; sulfonyl; and, oxalic acid. A preferred N
linker is 1,2-dimethylhydrazine.
The present invention contemplates a variety of both cyclic and acyclic backbone structures, as will be illus trated in Figures 5-9 below. One limitation of acyclic backbone structures is that activation of the electrophilic linking groups preparatory to polymer assembly, can lead to varying amounts of undesired intramolecular attack on sites of the subunit base. By contrast, with properly structured cyclic backbone moieties, the activated electrophile can be effectively isolated from reactive sites on the subunit base, thereby reducing unwanted intramolecular reactions.
However, use of aliphatic cyclic backbone moieties does entail the presence of multiple chiral centers in each backbone structure. With proper selection of cyclic backbone structures, synthetic challenges ajssociated with such multiple chiral centers can be largelyl, circumvented, by utilizing readily available natural products for the backbone moiety or, preferably, for the entilre subunit, or as a proximal precursor thereao.
This preference for backbone structures, or entire subunits, from natural sources reflects the difficulty, and corresponding greater expense, of de novo preparation of aliphatic ring structures having multiple rhiral centers.
Accordingly, preferred categories of dyclic backbone moieties are those comprising, or readilyiderived from, deoxyribose or ribose. In ad~~dition, certain other natural cyclic structures wherein a single enantiome~ is available, or can be readily prepared or isolated, are also preferred.
Figures 5A-5C illustrate exemplary cyclic backbone struc-tures comprising or derived from deoxyribosides or ribo-sides. R' in the figure indicates H or',alkyl, and R
indicates the subunit base, which, as seen, has the same ~i-orientation as natural nucleosides. Figurles 6A-6F illu-strate exemplary cyclic morpholino backbbne structures derivable from ribosides, having either a, /i-orientation (Figures 6A-6C) or an a-orientation (Figures',6D-6F) for the 5'-methylene (numbered as in the parent ribose), again with a ~ orientation of the R~ base. The synthesis of such subunits will be described below and inlExamples 1-5.
Figures 7A-7E show representative types of aøyclic backbone structures.
D. Intersubunit Linkaaes This section considers several types and properties of intersubunit linkages used in linking subun~ts to form the polymer of the invention. First, the backbone must be stable in neutral aqueous conditions. Since the binding polymers are designed for u;ae under physiological condi-~e 21 109 88 ~' tions it is necessary that the intersubunit linkages be stable under said conditions. The linkages must also be stable under those conditions required for polymer assem-bly, deprotection, and purification. To illustrate this stability requirement, an alkyl sulfonate (R- (SOZ) -O-CHZ-R' ) is precluded because the resultant structure is unduly sensitive to nucleophilic attach: on the CH2. Further, while carbonates (R-O-(C=O)-O-R') and esters (R-(C=O)-O-R') can be successfully prepared, their- instability under physio-logical conditions renders them of little practical value.
Secondly, the backbone must be adaptable to a confor mation suitable for target binding. If the intersubunit linkage is such that it exhibits specific rotational con formations (as is the case for amides, thioamides, ureas, thioureas, carbamates, thiocarbamates, carbazates, hydra-zides, thiohydrazides, sulfonamides, sulfamides, and sulfonylhydrazides) then it is important that either the rotomer compatible with target binding be the lowest energy conformation, or that the barrier to rotation between the conformations be relatively low (ie., that the conforma-tions be rapidly interchangeable at physiological tempera-tures). Thus, a secondary amide (N-alkyl amide, which prefers to adopt a traps conformation) would be acceptable if the traps conformation is suitable for pairing to the target duplex. By contrast, tertiary amides and related N,N-dialkyl structures generally have two approximately equal low energy conformations, and so to be useful in a binding polymer, the linkages should have a relatively low energy barrier to interconversion between the two conforma tions.
The barrier to rotation between two conformers can be assessed by NMR as follows: At a temperature where the two conformers are interconverting slowly relative to the NMR
time scale (on the order of 10-8 sec) two distinct signals are often seen, each representing a single conformer. As :-WO 9~,L,p03~2 PCT/L~S92/05208 the NI~t spectra are taken at progressively higher tempera-tures, the two conformer signals coalesce - indicating rapid interconversion. The coalescence temperature (Tc) thus provides a useful measure of the rotational freedom of 5 various linkage types. For example, N,N-dimethylformamide exhibits a Tc of about 114°C (Bassindale, 1984) and con-formers of analogous tertiary amides have been found to interconvert slowly in biological macromolecules. By contrast, an N,N-dialkyl carbamate-containing structure 10 exhibits a Tc just under 44°C (unpu!blished results obtained in support of the present invention), indicating reasonable conformational freedom at physiological temperature.
An N,N-dialkylsulfinamide (wh.ich should have a rota tional energy barrier similar to 'that of sulfonamide and 15 related substances) has been reported to have a Tc lower than minus 60°C. Based on these considerations, backbone linkages containing N,N-dialkyl-type carbamate, thiocarbamate, c:arbazate, and various amidates of phosphorous and sulfur are preferred, while 20 N,N-dialkyl-type amide, thioam:ide, urea, thiourea, hydrazide, and thiohydrazide linkages are generally unacceptable.
Third, the backbone should be uncharged. For thera peutic applications it is desirable. to design these binding 25 polymers so that they i) are not se~3uestered by the reticu loendothelial lining of the capillaries; ii) readily cross cell membranes; iii) are resistant to degradation by nucleases; and, iv) are not repelled by the high density of negative charge on the backbones of the target duplex.
30 These design objectives are best achieved by using both intersubunit linkages and backbone moieties which are largely uncharged (non-ionic) at physiological pH.
When the subunit bases are positioned on contiguous base-pairs of their target sequence via hydrogen-bonding, and if all recognition moieties o~f the subunit set have :31 well matched R, B, and A values, then the distance from the subunit base attachment position of one backbone moiety to the attachment position of the next backbone moiety is the square root of:
(R sine(rot))Z + (R cosine(rot) - R)z + (rise)z where R is the distance from the helical axis to the center of the atom of the backbone: moiety to which the subunit base is attached, rot is the: axial rotation value for the target duplex (typically about 30° to 33° for an A-form l0 duplex and 36° for a B-form duplex), and rise is the axial rise value for the target duplex (typically about 2.8 to 3.3 A for an A-form duplex and 3.4 A for a B-form duplex) .
It is this distance which must be spanned by the unit backbone length of the binding polymer, i.e., the length of one backbone structure plus the intersubunit linkage between backbone structures. However, it should be emphasized that both A-form (RNA/RNA and RNA/DNA duplexes) and B-form (DNA/DNA) target duplexes are somewhat flexible and so can generally accommodate binding polymers which have unit backbone lengths which are a fraction of an angstrom shorter or longer than the calculated length requirement. Further, it should be appreciated that DNA/DNA in a B conformation can be converted to an A
conformation under certain conditions.
In selecting a particvular backbone structure, the following factors bear on the: required length and so should be taken into consideration: first, any conformational restrictions imposed by hinds:red rotations about bonds such as amides and carbamates; second, when the subunit bases are in position on their target base-pairs, any steric interactions between these bases and the target duplex, and between the bases and the polymer backbone; third, steric interactions between different components of the backbone structure; and fourth, fo:r cyclic backbone moieties, ~11.0~~8 favored conformations of the component ring structure of the subunit backbone structures.
A generally satisfactory way to determine whether or not a prospective polymer backbone is likely to be acceptable for use against a particular target conformation (e. g., A-form or B-form) is to assemble with CPK molecular models a representative target genetic duplex in the desired conformation, with subunit bases H-bonded thereto, and then add the prospective polymer backbone. If the prospective polymer backbone can be easily attached without having to adopt an energetically unfavorable conformation, and if the attachment of the polymer backbone does not cause significant perturbation of the target structure, and if there are no unacceptable steric interactions, then the backbone should be operable. Additional support for the suitability of a prospective backbone structure can be obtained by modeling the polymer/target triplex on a computer using a molecular mechanics program to obtain an optimized bonding structure via an energy minimization procedure. Such modeling can, on occasion, identify significant unfavorable interactions (eg., dipole-dipole repulsions) which might be overlooked in the initial CPK
modeling.
As noted above, such factors as R, B, and A values for the subunit bases of a given subunit set, and steric and rotational constraints of particular subunit structures and intersubunit linkages, bear on how long a unit backbone must be in order to provide the correct spacing of subunit bases for binding to a target duplex in a given conformation. However, as a rule, subunit sets wherein the subunit bases of the set have R~ values less than about 7 angstrox~s and B values clustered within about 12° of each other, and A values clustered within about 20° of each other, generally require a 4-atom or 5-atom unit-length acyclic-type backbone, such as shown in Figures 8A-8C, or :33 a 6-atom unit-length cyclic-type backbone, such as shown in Figure 9A-9D, for binding to target duplexes in an A-type conformation.
Subunit base sets having Rb values less than about 11.5 Angstroms, 66 values within about 9° of each other, and A
values clustered within about 20° of each other generally require a 6-atom unit-length acyclic-type backbone, such as shown in Figure 8D-8E, or a ;~-atom unit-length cyclic-type backbone, such as shown in Figure 9E-9F, for binding to target duplexes in a B-type conformation.
However, it should be noted that DNA/DNA duplexes, which generally exist in a B conformation, can readily convert to an A conformation. Two such conditions which cause this B to A transition are high salt and low polarity solvent. It also appears ,that a B to A conformational transition of the target duplex can be induced by duplex-directed binding polymers having backbone unit-lengths shorter than optimal for lbinding to a H-form duplex.
However, such conformation transitions incur a cost in free energy of binding, and so, to~ compensate, the binding poly-mer's affinity for its target must be increased accordingly. Because of the feasibility of this B to A
conformational transition of target duplexes, for some ap-plications the shorter unit-length backbones suitable for A-form target duplexes can also be used for targeting genetic sequences which exisi~ normally in a B conformation.
E. Subunit Sets When the subunit basea of a set have acceptably matched R, B, and A values, and when subunit backbone structures which are identical or very similar in length and subunit base attachment position and orientation are used for all subunits of the set, the subunits of that set can be assembled in any desired order for targeting a selected duplex sequence.

zii~~~~

Each subunit of such a matched set consists of a subunit base linked at a standard position to a standard-length backbone structure. The subunit base of each subunit of the set has an R, B, and A value closely matched to the R, 8, and A values of the subunit bases of the other subunits of that set.
According to an important feature of the invention, the polymer subunits in a set must contain at least two different subunit types, each specific for a different oriented base-pair. Specifically, the base of each of at least two different subunits of the set is effective to form at least two hydrogen bonds with the major-groove sites of its respective target base-pair, where one of those hydrogen bonds is to the purine N7 nitrogen of the target base-pair, as discussed above.
The other subunit or subunits in the set may, but do not necessarily bind with high specificity to oriented base-pairs in the target sequence. Thus, another subunit of the set may bind satisfactorily to two different oriented base-pairs, as will be seen below. Such low-specificity or non-specific subunits serve to provide (a) required spacing between high-specificity subunits in the polymer and (b) contribute to stacking interactions between the planar bases in the polymer/duplex complex.
In addition, and according to an important feature of the invention, the subunits in the polymer must provide high-specificity base binding to at least about 70% of the oriented base-pairs in the target sequence. Thus, where a subunit set includes only two high-specificity bases, the target duplex sequence must contain at least 70% oriented base-pairs which are specifically bound by those two high-specificity bases.

E1. Basic Subunit Set for C:G and T:A or U:A Oriented base-pairs The most basic subunit set is suitable for targeting duplex genetic sequences coni:aining only C:G and T:A or U:A
5 oriented base-pairs.
The first member of this basic subunit set is a high-specificity guanine subunit containing a guanine or 6-thioguanine subunit base effective to hydrogen bond specifically to a C:G orient:ed base-pair. As illustrated 10 in Figure 10A, guanine (or 6-thioguanine) forms three hydrogen bonds to the polar' major-groove sites of a C:G
oriented base-pair, including the guanine N7 of that target base-pair. The subunit may be formed with any of a variety of deoxyribose, ribose or morpholino backbone structures, 15 with the base attached to the backbone structure in the ~-stereochemical orientation, as illustrated in Example 2.
The second member of the basic set is a high-specificity diaminopurine subunit containing a 2,6 diaminopurine subunit base effective to hydrogen bond 20 specifically to a T:A or U:A oriented base-pair. As illustrated in Figure lOB, the 2, 6-diaminopurine base forms three hydrogen bonds to the polar major-groove sites of a T:A or U:A oriented base-pair, including the adenine N7 of that target base-pair. As with the guanine subunits, a 25 variety of diaminopurine subunits with deoxyribose, ribose and morpholino backbone structures, and having the desired S-stereochemical attachment of the base to the backbone structure, can be prepared by modifications of commercially available nucleosides, also as illustrated in Example 2.
30 CPK molecular modeling showed that the guanine and diaminopurine moieties should effectively and specifically bind their target base-pairs. Additional support for this major-groove hydrogen-bonding mode was obtained from a best fit analysis carried out for these two trimolecular 35 complexes, C:G:G and U:A:D. An exhaustive review by Voet WO 93/00352 PCT/US92/0520ii ~1109~$

and Rich (1970) tabulates the lengths and angles of hydrogen-bonds from x-ray diffraction studies of crystalline complexes of purines and pyrimidines. In those tabulations NH:N bonds range in length from 2.75 A to 3.15 A and their angles range from 115° to 145°. NH:O bonds range in length from 2.60 A to 3.20 A and their angles range from 110° to 145°.
In the best fit calculations, structural parameters used for the purines and pyrimidines in the Watson-Crick base-pairs are those given by Rich and Seeman (1975).
Those parameters were obtained from x-ray diffraction of ApU and GpC crystals (right handed anti-parallel Watson-Crick) which were solved at atomic resolution. The guanine structural parameters referenced above were also used for the subunit base in Figure 10A. The 2,6-diaminopurine subunit base of Figure lOB was assumed to have structural parameters essentially identical to those of 9-ethyl-2,6-diaminopurine obtained from x-ray diffraction studies of crystalline trimolecular complexes of 9-ethyl-2,6-diaminopurine hydrogen-bonded to two 1-methylthymines (one thymine bonded in the Watson-Crick mode and the other thymine bonded in the reverse-Hoogsteen mode) as reported by Sakore et al. (1969).
To simplify the analysis, the approximation was made that all atoms are in the same plane. Table 4 gives the results of this analysis. In this table the standard purine and pyrimidine numbering system is used throughout, subunit base-G stands for the subunit base of Figure l0A
(guanine) and subunit base-D for the subunit base of Figure lOB (2,6-diaminopurine). Angles are measured as in Voet and Rich referenced above.

Table 4 Guanine subunit base H-bonded to a C:G base-pair WIC hvdroqen-bonds anale en th 02 (C) :NH2 (G) 125° 3 .17 A
N3(C):NH1(G) 119° 2.95 A
NH4(C):06(G) 129° 2.63 A
Major-Groove hvdroaen-bonds angle length NH2(subunit base-G):N7(G) 140° 3.12 A
NH1(subunit base-G):06(G) 115° 2.74 A
06(subunit base-G):NH4(C) 143° 2.63 A
Diaminopurine subunit base H-bonded to a U:A base-pair W/C hydrogen-bonds an a length NH3(U):Nl(A) 119° 2.98 A
04(U):NH6(A) 126° 2.71 A
Maior-Groove hydro eq n-bonds angle length NH2(subunit base-D):N'7(A) 137° 2.85 A
N1(subunit base-D):NH6(A) 139° 2.95 A
NH6(subunit base-D):04(U) 132° 3.00 A
As can be seen from tlhis table, all hydrogen-bond angles and lengths in the subunit base/base-pair complexes fall within established angle and length limits for hydrogen-bonds.
E2. Spacer Subunits for A:T and G:C Oriented Base-hairs The basic guanine plus ~diaminopurine subunit set can be easily prepared from readily available guanosine or deoxyguanosine. However, binding polymers assembled from only these two subunits, and targeted against sequences of WO 93/00352 PCT/US92/0520~
"2 ~.-1:4:9:8 8 at least 16 contiguous base-pairs, are expected to have targets in only quite large viruses having genome sizes on the order of 65,000 base-pairs or greater.
However, it is desirable to have binding polymers which can be targeted against a much broader range of viruses, including even quite small viruses such as Hepatitis B, which has a genome size of only 3, 200 base pairs. One effective approach to extending the targeting range of these binding polymers, without substantially increasing their cost of production, is to target sequences composed predominantly (at least about 70%) of target base-pairs for the guanine and diaminopurine high-specificity subunit bases (ie., oriented base-pairs C:G and T:A or U:A). The remaining base-pairs in the target sequence ( i . e. , no more than about 30% G: C and/or A: T or A: U) can then be accommodated by low-specificity "spacer" bases in the binding polymer, which serve primarily to provide continuity of stacking interactions between the contiguous subunit bases of the binding polymer when that polymer is in position on its target duplex.
Thus, in one embodiment, a polymer assembled from the basic subunit set described in Section E1 additionally includes one or more low-specificity spacer subunit bases.
When the binding polymer is in position on its target duplex, with the subunit bases stacked, the spacer subunit bases (which are not necessarily hydrogen-bonded to their respective base-pairs) should have R, B, and A values which can closely match the R, B, and A values of the high specificity subunit bases. Specifically, for the full subunit set, the R values should all be within about 2 B values should all be Within about 20°, and A values should all be within about 30°. Preferably, the spacer subunit bases should also provide modest hydrogen-bonding to their respective target base-pairs so as to make some contribu-tion to target binding specificity and affinity.

r Where the target sequence contains a G:C oriented base-pair, one preferred spacer subunit in the subunit set contains a cytosine base, which can hydrogen-bond weakly to G:C and to T:A oriented base-pairs. Figure 11A shows cyto-sine hydrogen bonded to the. major-groove sites of a G:C
base-pair, and Figure i1B shows cytosine hydrogen bonded to a T:A base-pair. In neither case does this include a hydrogen bond to the N7 of the purine of a target base-pair.
Where the target sequence contains an A:T or A:U
oriented base-pair, one prs:ferred spacer subunit in the subunit set contains a uraci.l (or thymine) base, which can hydrogen-bond weakly to A:T <ind to C:G oriented base-pairs.
Figure 12A shows uracil hydrogen bonded to the major-groove sites of an A:T base-pair, and Figure 12B shows uracil hydrogen bonded to a C:G ba:ae-pair. As with the cytosine spacer, neither of these hydrogen bonding interactions involve the N7 of the purine: of a target base-pair.
Although these two subunit spacer bases provide only low-specificity and low affinity binding to their target base-pairs, nonetheless: i) they effectively provide for continuity of subunit base sacking in the target-bound binding polymer; ii) they have R, B, and A values which are acceptably matched with the lR, B, and A values of the high specificity guanine and diaminopurine subunit bases of the subunit set; and iii) the spacer subunits, or close pre-cursors thereto, are commercially available and relatively inexpensive.
Syntheses of subunit sets containing the four subunit bases guanine, diaminopurine, cytosine, and uracil (or thymine), and having various deoxyribose, ribose and mor pholino backbone structures,, are described in Example 2.
The sets described in the example have the following back bone structures:
(a) 2~-deoxyribose, seen in Figure 5A1 Example 2A;

(b) 2'-O-methylribose, seen in Figure 5B (R - methyl), Example 2B;
(c) morpholino, seen in Figure 6A, Example 2C;
(d) N-carboxymethylmorpholino-5'-amino, seen in Figure 6C, 5 Example 2D;
(e) N-carboxymethylmorpholino-(alpha)5'-amino, seen gene-rally in Figure 6F, Example 2E;
(f) ribose with 5'carbazate, seen in Figure 5C, Example 2F;
(g) ribose with 5'sulfonylhydrazide, seen in Figure 5C, but 10 where the carbonyl group is replaced by a sulfonyl group, Example 2G;
(h) ribose with 5'glycinamide, seen in Figure 5C, but where the OCONHNHZ group is replaced by NHCOCHZNH2, Example 2H;
and, 15 (i) ribose with 5'(aminomethyl)(ethyl)phosphate, seen in Figure 5C, but where the OCONHNHZ group is replaced by OP02EtCH2NH2, Example 2I.
Table 5 shows the base-pair specificities and approxi mate R, B, and A values for the subunit bases of this 20 guanine, diaminopurine, cytosine, and uracil (or thymine) subunit set.
Table 5 25 Subunit Base Base-pair Specificity Ra 6~ A

G C: G 5. A 33 60 D T:A 5.6 A 32 60 C G: C & T:A 4. A 38 50 U A:T & C:G 4.8 A 38 50 It will be appreciated that binding polymers prepared with the above G, D, C and U or T subunit set also have the potential to bind to single-stranded genetic sequences.
Specifically, the polymer will be able to bind in a Watson-~1 109 88 Crick pairing mode to a single-stranded polynucleotide of the appropriate base sequenc:e.
Since the spacer subuni.ts, C and U or T, in the poly mer are degenerate in binding specificity, at least two of these low-specificity spacer subunits are required to provide a level of target specificity equivalent to that provided by one high-specificity subunit. Thus, a binding polymer containing 16 high-:specificity subunit bases pro-vides about the same level. of target specificity as a binding polymer containing 12 high-specificity subunit bases and 8 low-specificity spacer subunit bases.
E3. Subunit Set with a Tautomeric Subunit Specific for A:T and G:C Oriented Base-oa~irs In another embodiment, the guanine plus diaminopurine subunit set described in Section E1 includes an additional subunit having a tautomeric subunit base capable of hydrogen bonding to either G:C or A:T oriented base-pairs.
A generalized skeletal ring structure and hydrogen bonding array of one preferred base type is shown in Figure 13A, where X~ is H or NHZ; XZ is H, F, or Cl; and B indicates the polymer backbone. Figures 13H-13D show three preferred embodiments of this tautomeric base, as discussed further below.
The hydrogen bonding to target base-pairs by different tautomeric forms of the base from Figure 13D is shown in Figures 14A and 14B for G:C and A:T oriented base-pairs, respectively. As seen from Figure 14, X2 can be hydrogen-bond acceptor when the tautomer is hydrogen bonded to a G:C
base-pair, to provide three: hydrogen bonds to the base-pair. Similarly, X1 can be .a hydrogen-bond donor when the tautomer is hydrogen bonded i~o an A:T base-pair, to provide three hydrogen bonds to the base-pair.

Table 6 shows the base-pair specificities and approxi-mate R, 8, and A values for thEa subunit bases of the gua-nine, diaminopurine, and the subunit base of Figure 14:
Table 6 Subunit Base Base-pair Specificity R, B, A
G C: G 5. 8 A 33° 60°
D T:A 5.6 A 32° 60°
Tautomeric Base G:C & A:T 6.3 A 36° 55°
of Figure 13B
The syntheses of a number of specific embodiments of a tautomeric subunit are described in Example 3. The synthesis of the structures seen in Figure 13B and 13C are described in Example 3A for t;he 2'deoxyribose backbone structure; in Example 3B for the 2'O-methylribose backbone;
and in Example 3C for the morph~olino backbone.
E4. Subunit Set with High-Specificity Subunits for A:T and G:C oriented Base-pairs In still another embodiment., the guanine plus diamino purine subunit set described .in Section E1 includes an additional subunit whose base is specific for hydrogen bonding to a G:C oriented base-pair, or an additional subunit whose base is specific for hydrogen bonding to an A:T (or A:U) oriented base-pair-, or the set includes two additional subunits whose bases are specific for hydrogen bonding to a G:C oriented base--pair and to an A:T or A:U
oriented base-pair, respectively.
Figure 15A shows the ring structure and hydrogen bond-ing array of a general type of base effective to bind a G:C
oriented base-pair. Three prei=erred embodiments of this structure type are shown Figures 15B-15D. Figure 16 shows the structure in Figure 15D hydrogen-bonded to its 21 109 88 ,. , ~E 3 G:C target base-pair. As seen from Figure 15A and Figure 16, the X2 position in the Figure 15A structure may be a hydrogen bond acceptor, e.g., O) for forming three hydrogen bonds between the base and its target G:C base-pair.
Syntheses for subunits having a morpholino backbone structure and the G:C-specifi.c bases of Figures 15B and 15C
are described in Example 4D.
Figure 17A shows the skeletal ring structure and hydrogen bonding array of another general type of base effective to bind a G:C oriented base-pair. A preferred embodiment of this structurE: type hydrogen-bonded to its G:C target base-pair is shown in Figure 17B.
Synthesis of a subunit having a morpholino backbone structure and the G:C-spec:ific base of Figures 17B is described in Example 4E.
Figure 18A shows the skeletal ring structure and hydrogen bonding array of a general type of base effective to bind an A:T or A:U oriented base-pair. Three preferred embodiments of this structure type are shown in Figures 18B-18D. Figure 19 shows the structure in Figure 18D
hydrogen-bonded to its A:T target base-pair.
Syntheses for subunits having a morpholino backbone structure and the A:T or A:U-specific bases of Figures 18B
and 18C are described in Example 4C.
The subunits described in this section whose bases are specific for G:C, A:T and A:U oriented base-pairs, with the guanine and diaminopurine subunits described in Section Elr provide a complete set of sulbunits providing high-specifi-city hydrogen bonding for each of the four possible orien-ted base-pairs in duplex nucleic acids. A subunit set formed in accordance with one aspect of the invention may include any three of these high-specificity subunits effec-tive to bind to three different oriented base-pairs in a duplex target sequence. For exa~ple, in a target sequence containing T:A, C:G, and G:C base-pairs, the selected sub-21~.0~8~

unit set would include three different subunits containing a common or similar backbone structure and diaminopurine, guanine (or thioguanine), and one of the above G:C-specific bases. A subunit set suitable for a target sequence con-s taining all four oriented base-pairs would additionally include a subunit whose base is one of the above high-specificity bases for an A:T oriented base-pair.
Table 7 shows the base-pair specificities and appro ximate R, B, and A values for the subunit bases comprising guanine, diaminopurine, and the high-specificity bases of Figures 15, 17, and 18.
Table 7 Subunit Base Base-pair Specificity R, B, A

G C:G 5.8 A 33 60 D T:A 5.6 A 32 60 Base of Fig. G:C 6.3 A 36 55 Base of Fig. G:C 4.8 A 38 50 Base of Fig. A:T 6.4 A 36 55 The table illustrates the general suitability of this set of bases in regard to R, B, and A values.
II. Polymer Preparation This section describes assembly of the subunits com-prising a subunit set described above, to give a sequence-specific duplex-binding polymer.
A. Polymer Seauence and Length The polymer of the invention is designed to bind to and inactivate a target duplex sequence, such as a sequence essential for a given pathogen, without inactivating normal host genetic sequences. Thus, the sequence information recognized by the polymer should be sufficient to rigorous-~~ 5 ly distinguish the pathogen sequence from all normal host sequences.
A reasonable estimation of the amount of sequence information which a duplex :nucleic acids-binding polymer should recognize in a disease-specif is sequence in order to avoid concomitant attack on normal cellular sequences can be calculated as follows. The human genome contains roughly 3 billion base-pairs of unique-sequence DNA. For a gene-inactivating agent to have an expectation of having l0 no fortuitous target sequences in a cellular pool of 3 billion base-pairs of unique sequence genetic material, it should recognize at least n base-pairs in its target, where n is calculated as 4° - 3 x 109, giving a minimal target recognition requirement of approximately 16 base-pairs.
This suggests that a gene-inactivating polymer recognizing in excess of 16 base-pairs in its target sequence will likely have no targets in i~he cellular pool of inherent DNA. Obviously as the numbs:r of base-pairs recognized in the target sequence increases over this value the probability that the polymer will attack inherent cellular sequences continues to decrease. It is noteworthy that as the number of base-pairs recognized by the agent increases linearly, this "safety factor" increases exponentially.
To illustrate, Table 8 tabulates the number of base pairs recognized in a target sequence and the corresponding expected number of fortuitous targets in a pool of 3 billion base-pairs of unique:-sequence genetic material.

Table 8 Number of base-pairs Hxpected number of fortuitous recognized in target duplex targets in human genome 8 45,776 2,861 14 11.2 16 0.7 10 18 0.044 0.0027 The numbers in Table 8 indicate that in order to achieve adequate specificity for the pathogen or pathogenic 15 state, a binding agent for duplex nucleic acids should recognize at least 16, and preferably 18 or more base-pairs of the target sequence.
In addition to target sequence length, it is important to consider how many of the four possible oriented base 20 pairs in duplex nucleic acids (ie., A:T, C:G, G:C, and T:A) must be specifically recognized by the polymer bases in order to allow practical targeting of various viral patho-gens. Table 9 shows the approximate number of targets expected in a relatively small viral genome (about the size of the HIV provirus) as a function of the number of dif-ferent base-pair-binding specificities in a 16-subunit polymer. The values in the table were calculated on the assumption that the purine to pyrimidine ratio in a given strand of the pathogen's genome is approximately 1.0 and that the bases are effectively in a random order.

.. 2~ ~ pg 88 ~4 7 Table 9 Number o base-hair-binding ~pected number of continuous ~~pr;firities in subunit set i6-base-pair targets in a ~ 0 000 base pair viral aenome 1 0.000002 2 0.15 10,000 The tabulated values demonstrate that, in general, homopolymers (i.e., polymers assembled from subunits having specificity for just one oriented base-pair) are unlikely to have any practical targeas in natural duplex genetic sequences. Further, copolymers of just two subunit types with specificities for only two of the four oriented base-pairs are expected to have contiguous 16-base-pair targets in only quite large viruses: (eg. Herpes). In contrast, binding polymers assembled from subunit sets having specificities for three or four of the oriented base-pairs have a quite adequate number of targets in even the smallest DNA viruses (eg., hepatitis B with a genome size of only 3200 base-pairs).
As described in Section I, the basic two-subunit set formed in accordance with t;he present invention includes two subunits which are specific for two different oriented base-pairs, C:G and T:A or U:A. To increase targeting ver satility, another embodiment includes an expanded subunit set which includes one or two spacer subunits. Still another embodiment comprises. the basic two-subunit set plus an additional semi-specific subunit whose base is capable of hydrogen bonding to either of two different oriented base-pairs. As noted above, this semi-specific subunit base recognizes only half the sequence information recog-nized by a high-specificity subunit base, and thus its use 21 109 88 ~' will require a correspondingly longer polymer in order to achieve adequate specificity for its target. Yet another embodiment comprises the basic t:wo-subunit set plus one or two additional subunits whose high-specificity bases are each capable of hydrogen bonding to just one of the four oriented base-pairs. Such a subunit set containing sub-units for all four of the oriented base-pairs allows tar-geting of essentially any desired duplex genetic sequence.
B. Subunit Activation and Polvmer Assembly The subunits, prepared as in Examples 1 - 5, can be activated and then coupled in a controlled sequential man-ner to give the desired binding polymer. Representative polymer assembly procedures for deoxyribose-containing and 2~-O-methylribose-containing subunits are described in Example 6. Representative activation procedures for mor-pholino-containing subunits are described in Example 7;
Example 8 describes an exemplary procedure for assembling these activated subunits via solid-phase stepwise addition to give the desired binding polymers; and, Example 9 describes their purification. Figure 20 illustrates one subunit addition cycle of this stepwise assembly procedure using a representative morpholino subunit prepared as in Example 2C and activated as in Example 7A. Figure 21 illustrates a four-subunit-long segment of a representative polymer assembled from the subunit set prepared as in Example 4A-4D, and activated as. in Example 7A.
C. Novel Polymer Assembly Comprising: Oxidation,/Rinq, Closure~Reduction In addition to the above, a novel coupling procedure can also be used for assembling the desired nucleic acid binding polymers, of which one Eambodiment is illustrated in Figure 22. This procedure involves:

i) providing a subuni.t, or block of linked subunits, which contains vicinyl aliphatic hydroxyls, but no free primary amine (e. g., structure 1 of Figure 22);
ii) oxidizing those vicinyl hydroxyls to give a dialdehyde component (eg., structure 2 of Figure 22);
iii) providing a subunit, or block of subunits, which contains a free primary a7.iphatic amine (eg., structure 3 of Figure 22, and subunit.s prepared as in Examples 2F -2I);
iv) contacting the dialdehyde component with the primary amine component to effect coupling of the two components via formation of a cyclic morpholino structure having hydroxyls on the carbons adjacent to the morpholino nitrogen (eg., structure 4 of Figure 22); and, v) during or after t:he coupling reaction, or after completion of polymer assembly, adding a reducing agent to remove the hydroxyls on thEa carbons adjacent to the morpho-lino nitrogen, to give the: desired morpholino ring struc-ture (e. g., structure 5 of Figure 22).
The vicinyl-hydroxyl--containing moiety can be other than ribose, such as galac;tose or glucose. Further, this coupling method can be used in either a solution-phase or a solid-phase mode for polymer assembly. Also, the oxida-tion step and the subsequE:nt coupling step are preferably carried out in alcohol or water or a mixture thereof, and at a pH near neutrality. Although the reduction can be carried out during or after the coupling, best results are obtained when reducing agent, e.g., NaCNBH4, is present during the coupling step. Complete reduction and disrup-tion of borate complexes (generated when NaCNBH4 is used for the reduction) is best achieved by a final acidic wash having a pH in the range of 3 to 5 - which can be carried out after each coupling, or after all couplings are completed.

21 109 88 v Example 10 describes a representative application of this "oxidation/ring closure/reduction" coupling method for stepwise solid-phase assembly of a binding polymer.
5 D. Polymer Modifications Some of the polymer t;rpes of the invention have relatively poor solubilities for polymer sizes above about 15-20 subunits, e.g., in the ?Low-micromolar range. It may thus be desirable to enhance t:he solubility of the polymer 10 by addition of a hydrophilic moiety, such as a polyethylene glycol (PEG) chain. This can :be accomplished, according to one approach, by deprotectin~g the polymer terminus, and reacting the polymer with an Excess of activated hydrophi-lic compound, e. g. , PEG activated by bis (p-nitrophenyl) car-15 bonate. Thereafter the binding polymer is cleaved from the synthesis support and treated with ammonium hydroxide to remove the base-protecting groups, and then purified, preferably by ion exchange chromatography at pH 10.5. One preferred hydrophilic molecu7_e is PEG having an average 20 molecular weight of about 1000 daltons (commercially available from Polysciences, :Cnc. and Aldrich Chem. Co.).
For some applications it: may be desirable to modify the polymer to favor its cellular uptake via endocytosis.
This may be done, for example, by derivatizing the polymer 25 with a polycationic molecule, such as polylysine. Coupling of such a molecule containing one or more primary amine moieties may be by reaction of the base-protected polymer with a bifunctional coupling agent, such as disuccinimidyl suberate, or other commercially available agent (e. g., 30 Pierce Chemical Company) and then adding the amine-containing polycationic molecule.
Where the polymer molecules are to be attached to a solid support, for use in a diagnostic system, the terminal N- protective group can be cleaved (leaving the bases still 21 109 ~8 in the protected state), and reacted with a suitable cross-linking agent, such as disu.ccinimidyl suberate. This pre-paration is then added to the support material, such as latex microparticles containing suitable linker arms termi-nating in primary amine moieties.
Alternatively, if it i.s desired to purify the binding polymer prior to attachment to a support, a methoxytritryl-protected 6-aminocaproic acid can be linked to the unpro-tected N-terminus of the binding polymer using DCC. The binding polymer is then treated with ammonium hydroxide to deprotect the bases, purified by standard methods, and the terminal methoxytrityl is cleaved from the aminocaproic acid moiety. Finally, the purified polymer is mixed with support material having suitable linker arms terminating in p-nitrophenylester moietie:~, to give covalent coupling of the polymer molecules to the support.
Binding polymers constructed from subunits having cyclic backbone moieties have a strand polarity analogous to the 5' to 3' strand polarity exhibited by standard phosphodiester-linked polynucleotides. As a consequence, for a given heteromeric target sequence of base pairs, two binding polymers can be constructed, one having the proper sequence of bases ordered from 5' to 3', and the other having the same sequence o~f bases, but ordered 3' to 5' .
The preferred polymer for .any selected target sequence of base pairs is readily determined by assembling the two binding polymers containing the appropriate sequence of bases in both possible oris:ntations, and testing these two polymers for their respective binding affinities for the selected duplex target sequence. Similar approaches for determining proper binding orientations for standard polynucleotides are well-known in the art. It should be appreciated that these binding polymers have the potential to bind their target duplex in either or both of the two orientations.

2110g$~

III. Utilitv A. Diagnostics: Detection of Sequences in Duplex form In one application, the polymer of the invention is used in a diagnostic method for detecting a duplex target nucleic acid sequence in an analyte. The target sequence is typically a pathogen-specific sequence, such as a virus or bacterial genome sequence, which is to be detected in a biological sample, such as a blood sample.
The target sequence is preferably 15 to 25 subunits in length, to provide the requisite sequence specificity, as discussed above. In one assay format, the diagnostic reagent is a solid support, such as a micro-bead, coated by covalently-bound polymers effective to specifically bind to the duplex target sequence. After sample treatment to release the analyte duplex from bacterium or virus in free form, if necessary, the sample is contacted with the solid support under conditions sufficient to effect base-pair-specif is binding of the analyte duplex to the support-bound polymer. Typically, the binding reaction is performed at 20°-37°C for 10 minutes to 2 hours. After washing the solid support to remove unbound material, the support is contacted with a reporter reagent effective to bind to the captured target duplex, to allow detection of said duplex.
The reporter may be a soluble duplex-binding polymer, formed in accordance with the present invention, which is base-pair-specific for a second analyte-specific target sequence in the analyte duplex, and which is labeled with a suitable signal group, such as a fluorescent moiety, for signal detection. The signal group is coupled to the poly-mer by standard coupling methods, such as described in Section II.
After washing the support, it is examined for bound reporter, which will be proportional to the amount of analyte bound to the support via the sequence-specific binding polymer.

2~ ~ oa 8a Alternatively, the washed support containing bound analyte duplex may be reacted with a fluorescent interca-lating agent specific for nucleic acids, such as ethidium bromide, and then the polymer-bound analyte is assessed by its fluorescence. Another alternative is to react the washed support containing bound analyte duplex with a reporter-labeled polycationic molecule, such as a fluores-cent-labeled oligo-cation, as described in co-owned pub-lished PCT Application No. PCT/US86/00545 (WO 86/05519).
The reporter molecule bind:: by electrostatic interactions with the negatively charged analyte duplex backbone, but does not bind the substantially uncharged polymer molecules on the solid support. After washing the support to remove unbound material, the reporter bound to the solid support, via the sequence-specific analyte/polymer complex, is measured.
B. In situ Hvbridization In many applications, the in situ hybridization is directed toward a target sequence in a double-stranded duplex nucleic acid, typically a DNA duplex associated with a pathogen or with a selected sequence in chromosomal DNA.
In the method, as it has been practiced heretofore, a labeled nucleic acid probe is added to the permeabilized structure, the structure is heated to a temperature suf-f icient to denature the target duplex nucleic acid, and the probe and denatured nucleic acid are allowed to react under suitable hybridization conditions. After removing unbound (non-hybridized) probe, the structure is examined for the presence of reporter label, allowing the sites) of probe binding to target nucleic acid to be localized in the bio-logical structure.
The method has been widely applied to chromosomal DNA, for mapping the location of specific gene sequences and determining distances between known gene sequences, for WO 93/00352 ?CT/US92/05208 21 '109 8a studying chromosomal distribution of satellite or repeated DNA, for examining nuclear organization, for analyzing chromosomal aberrations, and for localizing DNA damage in single cells or tissue. Several studies have reported on the localization of viral sequences integrated into host-cell chromosomes. The method h,as also been used to study the position of chromosomes, by three-dimensional recon-struction of sectioned nuclei, and by double in situ hybridization With mercurated arid biotinylated probes, using digital image analysis to study interphase chromosome topography. Another general application of the in situ hybridization method is for detecting the presence of virus in host cells, as a diagnostic tool.
In the present application, the polymer of the inven tion is designed for targeting a specific duplex genetic sequence associated with a cel7.ular or subcellular struc ture of interest, such as a chromosomal preparation. The polymer is derivatized with a suitable label such as a fluorescent tag. The polymer is preferably added directly to cells or tissue containing the structure being studied, without first permeabilizing t:he material. Because the polymer is uncharged it can more readily penetrate into living cells without the need for a: permeabilization treat ment. It further offers the advan~age of being resistant to nuclease degradation.
Once in contact with the duplex target material of interest, base-pair-specific binding can occur at nonaal physiological temperatures, again allowing detection of duplex targets under conditions of normal cell activity, and without heat disruption of the material being studied.
After a time sufficient for binding to the target duplex, and washout of unbound polymer, the structure being studied may be examined directly, e.g., by fluorescence microscopy, to observe site-specific localization of the duplex target sequence and possible movement thereof. Alternatively, to C

21 109 88 ~w reduce fluorescence background, the material may be fixed, e.g., by ethanol treatment, washed to remove unbound repor-ter, and viewed in fixed form by microscopy.
5 C. Isolation of Duplexes Containing Tarctet Sequence Another general application of the polymer invention is for isolating duplex nuc:Leic acid structures from a nucleic acid mixture, such a:a a mixture of genomic frag-ments, a blood sample containing a selected viral duplex, 10 or a mixture of plasmids with. different duplex inserts in different orientations.
The binding polymer used in the method is (a) designed for base-pair-specific binding to a selected target duplex sequence and (b) capable of :being isolated from a liquid 15 sample after capture of the itarget duplex. To this end, the polymer may be bound to a~ solid support, as described above, or may be derivatized with a ligand moiety, such as biotin, which permits capture on a solid support, or immunoprecipitation, after binding to the target duplex.
20 The polymer is added to ithe sample material and incu-bated under conditions which allow binding of the polymer to its target sequence, typically for 10 minutes to 2 hours at 20°-37°C. After binding has occurred, the polymer and bound material are isolated from the sample. The isolated 25 material may be released from the polymer by heating, or by chaotropic agents, and further amplified, if necessary by polymerase chain reaction methods, and/or clonal propaga-tion in a suitable cloning vector.
30 D. Site Specific DNA Modification The polymer of the invention is also useful for selected site-specific modii:ications of duplex DNA in vitro. These may include cutting a duplex species at a selected site, or protecting a selected region against 35 restriction or methylating enzymes. The latter application is useful particularly in recombinant DNA technology, where it is often advantageous to be able to protect a vector or heterologous DNA sequence against cutting by a selected restriction endonuclease, or where it is desired to selec-tively prevent methylation at a given restriction site.
To produce site-specific cleavage in a selected base sequence, the polymer is derivatized with a cleaving moiety, such as a chelated iron group, capable of cleaving duplex DNA in a polymer bound state. The polymer sequence is selected to place the cleaving group, which is typically coupled at one polymer end, adjacent to the site to be cleaved. To protect a selected region of duplex target sequence against restriction or methylase enzymes, the polymer includes a sequence for binding to the 4-8 base-pair sequence which specifies a selected restriction enzyme sequence - plus any additional proximal bases effective to give increased specificity f or a unique target sequence .
After addition of the polymer to the duplex material, the material is treated with the selected restriction or methylating enzyme. After enzyme treatment, the treated duplex is "deprotected" by heating.
E. ~heraoeutic Application_ The polymers of the invention, by their ability to bind to duplex target sequences, have the potential to inactivate or inhibit pathogens or selected genomic sequences, such as oncogenes, associated with disease.
Origins of replication and enhancer and promoter sequences are particularly sensitive to inactivation by duplex-directed binding agents, because the agent can occupy a target site required for initiation of replication or transcription of the targeted gene. Such gene-control sequences are known for many pathogenic genes, and also for a variety of oncogenes which have been characterized in humans.

21 'I 09 88 For some therapeutic applications, it may be desirable to modify the binding polymer to favor its delivery to cer-tain cells or tissues, or t:o favor its delivery to certain subcellular organelles, such as the nucleus (Chelsky).
This can be accomplished, for example, by linking the bind-ing polymer to a suitable signal structure, such as desialylated galactosyl-containing proteins (Gregoriadis, 1975) or a cluster of galactose moieties, which favors uptake by liver cells; or such as D-mannose or L-fucose, which favor uptake by Kupffer cells and macrophages; or such as insulin or related peptides, which may then be actively transported across the blood/brain barrier.
Additionally, the binding polymers can be incorporated into surfactant micelles, with or without brain-specific antibodies, to enhance delivery across the blood/brain barrier (Kabanov).
For the reasons discussed above, the polymer should generally contain at least 16 base-pair-specific subunits, to minimize the possibility of undesired binding to sequen-ces other than the intended target sequence. Candidate target structures can be determined from analysis of geno-mic sequences, such as are available in a variety of sequence databases. Preferred target structures are those which are (a) well conserved across strains, and (b) have a base-pair sequence which is compatible with the set of subunits available for forming the polymer. For example, if the subunit set includea a guanine, diaminopurine, and one or two spacer subunit~;, as detailed in Section I, the target sequence preferably contains at least about 70% C:G
and T:A oriented base-pair's, and the remainder G:C and/or T:A.
As an example, a search was made of the HIV-I genome, in the duplex proviral stage, for sequences which are both well conserved across strains and suitable targets for binding polymers assembled predominantly from guanine and ~.~~pgg8 2,6-diaminopurine-containing subunits. Table 10 shows several such selected target sequences, and positioned thereon, binding polymers assembled from the "two subunits plus spacers" set of the type described in Section I.E2.
Table 10 Position Gene Polymer/Target Complex in Genome DDDDDUGDUDGGGGGDD Polymer _____*__*________ 2431 Pol 5'-AAAAATGATAGGGGGAA Target TTTTTACTATCCCCCTT-5' Duplex DUDDDGDDDDDDGDCDG Polymer _*____________*__ 2735 Pol 5'-ATAAAGAAAAAAGACAG Target TATTTCTTTTTTCTGTC-5' Duplex GGDDDGGUGDDGGGGCDGUDGUDD Polymer _______*_______*__*__*__ 4956 Pol 5'-GGAAAGGTGAAGGGGCAGTAGTAA Target CCTTTCCACTTCCCCGTCATCATT-5' Duplex In the table, "-" represents a high-specificity base-pair binding, and "*" represents a low-specificity base-pair binding.
The following examples detail synthetic methods for preparing a variety of subunits, subunit sets, and poly-mers, in accordance with the invention. The examples are intended to illustrate but not limit the invention.

5a 21 109 88 Example 1 Subunit Protecaion Methods A. General procedure for the: protection of primary amino groups on bases of subunits.
Unless otherwise indicated, chemicals are purchased from Aldrich Chemical Co., Milwaukee, WI.
The subunit, generally a nucleoside or nucleoside analog, (10 mmol, which has been dried by coevaporation with pyridine several times) is dissolved or suspended in pyridine (50-100 mL), and treated with chlorotrimethyl-silane (2-3 equivalents of sil.ane per hydroxyl group in the substrate). The solution is stirred one hour, or until solution is complete (sonication may be employed with dif-ficultly soluble substrates). An alkyl chloroformate, acid chloride, or anhydride, or other suitable activated car-boxylic acid derivative is added (1.05-4.0 equivalents per amino group in the substrate:). After stirring for 1-24 hours at room temperature, tree reaction is cooled to 0°C, and treated slowly with a 1:1 mixture of pyridine/water (20 mL). After 10 minutes concentrated ammonium hydroxide (20 mL) is added and stirring continued for 15 minutes. The solution is concentrated under vacuum and dissolved in ethyl acetate (or ether or chloroform) and shaken with water. The organic phase is removed and the product allowed to crystallize. If no crystallization occurs, the solvent is removed and the residue chromatographed on silica to yield the N-acylated species. Typical chloroformates which are useful include 9-fluore:nylmethoxycarbonyl chloride, 2-(p-nitrophenyl)ethoxycarbonyl chloride (Himmelsbach), and 2-(phenylsulfonyl)ethoxycarbonyl chloride (Balgobin).
Typical acid chlorides include benzoyl, isobutyryl, and trichloroacetyl. Typical anhydrides include acetic, isobu-tyric, and trifluroacetic.

21 109 88 a Other acid derivatives include acyl hydroxybenzotriazolides (prepared from the acid chloride and dry hydroxybenzotri-azole in acetonitrile). The latter are advantageously used to introduce the phenylacetyl group. Alternatively, pri-5 mary amino groups may be protected as amidines by the procedure of McBride, et al.
B. Procedure for the differential protection of primary diamines on base-pair recognition moieties.
10 2,6-Diaminopurineriboside (Pfaltz and Bauer, Inc.) is converted by the general proc=edure in Example lA into the N-2 , N-6 bis- (phenylacetyl ) amide . The acyl group at the N-6 position is selectively cleavE:d by treatment of the nucleo-side with 1N LiOH in pyridine/ethanol at 0°C. The reaction 15 mixture is neutralized with act. HC1 and the solvents evapo-rated. The residue may be: recrystallized from ethyl acetate/ethanol or purified by silica gel chromatography.
The crude product, or the purified nucleoside, is resubjec-ted to acylation by the gene=ral procedure using benzoyl 20 chloride to introduce the N--6 benzoyl group. For this second acylation only a slight excess of the acylating agent (1.05-1.2 equivalents) is employed.
C. Procedure for the protection of oxo groups in the 25 recognition moieties.
2',3',5'-Tri-0-isobutyryl N2-isobutyrl deoxyguanosine is converted by the procedure: of Trichtinger, et al, into the o6 2-(p-nitrophenyl)ethyl. derivative. Alternatively, guanosine may be converted into the 06 diphenylcarbamoyl 30 derivative by the method of Kamimura, et al. Following treatment with ammonia (1:1 c:onc. ammonium hydroxide/DMF) or iN LiOH in pyridine/ethanol at 0°C, the N2-propionyl 06-diphenylcarbamoyl guanosine is produced. These proce-dures are applicable to the preparation of N-2 acylated O-4 35 protected 2-amino-4(3H)-quinazolinone derivatives and N-7 acylated O-9 protected 7-amino-9(8H)-imidazo[4,5-:E]quinazolinone derivatives.
D. General procedure for the introduction of a dimeth-oxytrityl substituent at a primary alcohol.
The alcohol bearing substrate (10 mmol) is dissolved or suspended in pyridine (50-100 mL) and treated with 4,4'-dimethoxytrityl chloride, triethylamine (20 mmol) and 4-dimethylaminopyridine (0,.5 mmol). After several hours at room temperature the mixture is treated with water (5 mL) then poured into cold, satd. aq. sodium bicarbonate solution. The mixture is s:xtracted with ethyl acetate (or chloroform) and the combined organic layers are dried (sodium sulfate) and evaporated. The residue is chromatographed on silica t:o give pure dimethoxytritylated compound.
Example 2 Preparation of "2-Subunits plus Spacers" Set.
A. Subunits containing 2'~-Deoxyribose moiety.
The 5'-O-dimethoxytrityl protected derivatives of the following are available from Sigma (St. Louis, MO, USA) .
N-4 benzoyldeoxycytidine, N-2 isobutyryldeoxyguanosine, 25~'~ thymidine. 2,6-Diaminopurine-2'-deoxyriboside is available from Sigma and is protected. at the primary amino groups and the primary hydroxy group :by the methods in Example 1.
B. Subunits containing 2'-0-Methylribose moiety.
The 2'-O-methylribonu.cleosides of uracil, cytosine, guanine, adenine, and 7-dea~zaadenine may be obtained by the method of Robins, et al (1974) or Sekine, et al. The guanosine and 2-aminoadenoaine 2'-0-methyl ethers are also advantageously prepared by the method of Robins, et al, (1981). They may be converted into their base protected a .

analogues by the general methods in Example 1 (for example, N-2 isobutyryl for the guanosine derivative, N-2 phenylacetyl, N-6 benzoyl for the 2-aminoadenosine derivative, N-4 benzoyl for the cytidine derivative). The primary hydroxy is protected as in Example 1.
C. Subunits containing Morpholino moiety.
A ribose-containing subunit, having the base in the protected form, is oxidized with periodate to a 2'-3' dialdehyde. The dialdehyde is closed on ammonia or primary amine and the 2' and 3' hydroxyls (numbered as in the parent ribose) are removed by reduction with cyanoborohydride.
An example of this general synthetic scheme is described below with reference to the synthesis of a base protected cytosine (R~*) morpholino subunit. To 1.6 L of methanol is added, with stirring, 0.1 mole of N4 benzoylcytidine and 0.105 mole sodium periodate dissolved in 100 ml of water. After 5 minutes, 0.12 mole of ammonium biborate is added, and the mixture is stirred 1 hour at room temperature, chilled and filtered. To the filtrate is added 0.12 mole of sodium cyanoborohydride. After 10 minutes, 0.2 mole of toluenesulfonic acid is added. After another 30 minutes, another 0.2 mole of toluenesulfonic acid is added and the mixture is chilled and filtered. The solid precipitate is dried under vacuum to give the tosylate salt of the free amine. The use of a moderately strong (pKa < 3) aromatic acid, such as toluenesulfonic acid or 2-naphthalenesulfonic acid, provides ease of handling, significantly improved yields, and a high level of product purity.
Filtration of the tosylate salt of the 2,6-diaminopurine-containing morpholino subunit also works well. However, the tosylate salts of the guanine-containing and uracil-containing subunits are generally ?1 109 88 more soluble in methanol. Thus, for G and U subunits the methanol is removed under reduced pressure and the residue partitioned between brine: and isopropanol - with the desired product going into the organic phase.
The base-protected m,orpholino subunit can then be protected at the annular nitrogen of the morpholino ring using trityl chloride.
As an example of the ~tritylation step, to 2 liters of acetonitrile is added, with stirring, 0.1 mole of the tosylate salt from above, followed by 0.26 mole of triethylamine and 0.15 mole of trityl chloride. The mixture is covered and stirred for 1 hour at room temperature, after which 100 ml of methanol is added, followed by stirring for 15 minutes. The solvent is removed under reduced pressure and then 400 ml of methanol is added. After the solid is thoroughly suspended as a slurry, 5 liters of water is added, the mixture is stirred for 30 minutes, and filtered. The solid is washed with 1 liter of water, filtered and dried under vacuum. The solid is resuspended in 500 ml of dichloromethane, filtered, and rotovaped until precipitai=ion just begins, after which 1 liter of hexane is added and stirred for 15 minutes. The solid is removed by filtering, and dried under vacuum.
The above procedure yields the base-protected morpholino subunit tritylated on the morpholino nitrogen and having a free 5' hydroxyl (numbered as in the parent ribose) .
D. Subunits containing N-~Carboxymethylmorpholino-5'-amino moiety.
A ribose-containing subunit, having the base-pair recognition moiety in the protected form, is converted to the 5'amine and that 5' amine tritylated, as per Stirchak, Summerton, and Weller (1987), or by the method described in Example 2E below. Following the general procedures of 21 109 88 ~' Example 2C above, the viciny:L 2' and 3' hydroxyls of the ribose are then oxidized with periodate to give a 2'-3' dialdehyde. The dialdehyde is closed on glycine in the presence of triethylamine. The 2' and 3' hydroxyls (num-bered as in the parent ribose) are subsequently removed by reduction with cyanoborohydride.
Alternatively, the dialdE:hyde can be closed on ammonia and reduced as in Example ~>.C, and then the morpholino nitrogen alkylated with bromoacetic acid buffered with N, N
diethylaniline.
These procedures yield t:he base-protected morpholino subunit having a tritylated 5' amine and a carboxymethyl group on the morpholino nitrogen.
E. Subunits containing N-Carboxymethylmorpholino-alpha(5'-amino) moiety.
Examples 2C and 2D illustrate the preparation of morpholino-containing subunit;s wherein the 5' methylene is in the beta orientation -- that is, the same orientation as in the parent ribose. Analogous morpholino-containing subunits wherein the 5' methy:lene is in the alpha orienta-tion can be prepared by the following general approach.
The 5' hydroxyl of a ribose-containing subunit, having the base-pair recognition moiety in the protected form, is converted to a secondary amine by established methods (see Example 2D above). Thereafter, following the general procedures of Example 2C above, the vicinyl 2' and 3' hydroxyls of the ribose are oxidized with periodate to give a 2'-3' dialdehyde. The 2' a:ldehyde rapidly closes on the secondary amine at the 5' position (numbered as in the parent ribose). Reduction with cyanoborohydride then gene-rates a structure containing a morpholino ring wherein the annular morpholino nitrogen is tertiary, and containing a 5'aldehyde in the alpha orientation. Subsequent addition 21 109 88 ~..
6:5 of ammonia or a primary amine in the presence of excess cyanoborohydride, generates a 5' amine (primary or secondary, respectively) in the alpha orientation.
The above general strategy can be applied to prepare subunits containing an N-carboxymethylmorpholino-alpha(5' amino) moiety, as well as a number of other useful varia tions. One method to introduce the desired secondary amine at the 5' position of the ribose moiety entails: a) conver sion of the 2',3' hydroxyls to an acetal as per the method of Smith, Rammler, Goldberg a.nd Khorana (1961) ; b) oxida-tion of the 5'hydroxyl to an <~ldehyde using DMSO/pyridine/
trifluoroacetic acid/diisoproylycarbodiimide (the Moffat oxidation); c) reacting this 5' aldehyde with glycine (or the tert-Butyl ester of glycine) in the presence of cyano-borohydride; and, regeneration of the 2',3' hydroxyls by acid cleavage of the acetal.
F. Subunits containing Ribose with 5'-Carbazate.
A ribose-containing subunit can be converted to the 5'carbazate as follows. To 10 mmol of ribose-containing subunit, having exocyclic amines of the base-pair recogni tion moiety in the protected) state, is added 100 ml of anisylaldehyde and 0.5 g of tosic acid. The reaction mixture is stirred at room temperature for 48 hours. The reaction mixture is added to 500 ml hexane and the pre-cipitate is collected. The product is then purified by silica gel chromatography f,eveloped with ether. The resulting product is reacted with 2 equivalents of bis(p-nitrophenyl)carbonate plus 2 equivalents of triethylamine in acetonitrile for 8 hours at 30 deg. C. The product is purified by silica gel chromatography developed with a 5%
to 15% acetone/chloroform mixture. The product is reacted with 4 equivalents of t-butylcarbazate in DMF for 4 hrs at 50 deg. C. The reaction mixture is added to water and the precipitate collected and suspended in DMF/conc. NH40H, 1:1 by vol overnight at 30 deg. C. The ammonium solution is added to brine and the insoluble product collected and dried under vacuum. The d.ry product is dissolved in trifluoroacetic acid and, after 5 minutes, ether is added to precipitate the product, which is triturated twice with ether. The product is dissolved in methanol containing sufficient N-ethylmorpholine to neutralize all residual trifluoroacetic acid and the ~groduct again precipitated by addition of ether, and the product dried under vacuum. The desired 5'carbazate product can generally be purified by silica gel chromatography dEaveloped with N-ethylmorpho-line/methanol/chloroform, 1:4:6 by volume, or preferably, purified by recrystalization from a suitable aqueous/
organic mixture.
G. Subunits containing Ribose with 5'-Sulfonylhydrazide.
A ribose-containing subunit can be converted to the 5'-sulfonylhydrazide as follows. Ten mmol of ribose-con-taining subunit, having exocyclic amines of the base-pair recognition moiety in the protected state, is converted to the anisylacetal derivative as described in Example 2F
above.
To 10 mmol of sulfonyl chloride in dichloromethane chilled on dry ice is added 1~5 mmol of N,N-diethylaniline.
Next is slowly added, with rapid stirring, a dilute solu-tion of 10 mmol of N-aminophthalimide in dichloromethane.
After 20 minutes, the anisylacetal subunit derivative is added to this chlorosulfo:nylhydrazide solution. With rapid stirring is slowly added 30 mmol of diisopropyl-ethylamine in 30 ml of dichloromethane. After stirring 1 hour at room temperature, the solvent is removed under reduced pressure and the product is purified by silica gel chromatography developed with an acetone/ chloroform mixture.
The product is then treai~ed with hydrazine acetate in methanol, the solvent removed under reduced pressure, and ~~ ~ os 8s DMF/con NH40H, 1:1 by vol i;s added and the preparation incubated at 30 deg. C overnight. Lastly, the product is treated with trifluoroacetic acid and worked up as in Example 2F.
H. Subunits containing Ribose with 5'-glycinamide A primary amine is introduced into the 5' position of a ribose-containing subunit following the oxidation/reduc-tive alkylation procedure des~~ribed in Example 2E, except-ing ammonia is used instead of glycine. This 5' primary amine is then acylated with N--tert-butoxycarbonyl glycine, p-nitrophenyl ester. After purification, the protective groups are removed by treatment with DMF/con NH40H, and then with trifluoroacetic acid, a.nd the final 5'-glycinamide derivative worked up as in Example 2F.
I. Subunits containing Ribose with an aminomethylethyl-phosphate group linked to the 5'oxygen.
Aminomethylphosphonic acid (Aldrich Chem. Co.) is re acted with trityl chloride in the presence of triethyl amine. The di-anionic pho:aphonate product, where the counter ions are triethylammonium, is suspended in ethanol and then a carbodiimide, such as dicyclohexylcarbodiimide (DCC), is added. The resultant mono-anionic product is shaken with a mixture of water and chloroform containing pyridinium hydrochloride. This procedure gives a mono-ionic phosphonic acid having a pyridinium counter ion.
This product is added to chloroform, followed by addition of the ribose-containing subunit wherein exocyclic amines of the base is in the protected form and the 2'and 3' hydroxyls are protected as thEa anisylacetal. DCC is added to couple the phosphonate to the 5'oxygen of the subunit.
The product is dried and chromatographed on silica using methanol/chloroform 2~1 109 88 mixtures. The pure product ~'.s next base-deprotected with DMF/conc. NH40H, 1:1 by vol. and then suspended in trifluoroacetic acid to remove the trityl and the anisyl protective group.
Examx~le 3 Preparation of Subunit;s With Tautomeric Base A. Subunit containing 2'-Deoxyribose moiety.
1. Preparation of N-glycosyl isoindoles 4-Acetylamino-2-methylbenzoic acid (Peltier) is converted into the 5-nitro compound by treatment with cold fuming nitric acid. The reaction mix is poured into crushed ice and the solid product collected by filtration and purified by recrystallization from DMF/water or by silica chromatography. The a.cetamide is removed by alka-line hydrolysis with 1-10% NaOH solution in 90% ethanol.
The reaction mixture is added to excess dilute HC1 and the solvent evaporated. The crude acid is esterified with satd. methanolic HC1 at room i~emperature for several days.
After removal of solvent the product is partitioned between ethyl acetate and satd. sodium bicarbonate. After washing with water the organic phase :is evaporated and the residue purified by silica chromatography. The nitro group is reduced to the amino using hydrogen and palladium on carbon in ethanol or DMF. After filtration through celite and evaporation, the crude diamine is converted to the methyl 2-amino-6-methylbenzimidazole-5-carboxylate using cyanogen bromide in methanol at reflux. The mixture is cooled and poured into satd. aq. sodium bicarbonate and the solid product filtered and purified by recrystallization. The exocyclic amino group is acylated by refluxing with phthaloyl dichloride in pyridine followed by reaction of the diazepine with pyrazole in refluxing acetonitrile according to the method of 6 !3 Katritzky. The compound is reacted with either bromine or N-bromosuccinimide or 1,3-dibromo-5,5-dimethyl-hydantoin either neat or in carbon tetrachloride or chloroform or 1,1,1-trichloroethane with the aid of a high-intensity sun lamp and/or benzoyl peroxide, to provide the benzylic bromide. It is possible to a.cylate the diazepine further with isobutryl chloride in pyridine to produce a triply acylated benzimidazole species. This is normally done prior to the bromination.
The crude benzylic bromide is reacted with sodium azide in dry DMF and reduced with hydrogen over platinum or palladium to produce the lactam. This lactam is O-silylated with one equivalent of trimethylsilyl trifluoromethane-sulfonate or tert-butyldime.thylsilyl trifluoromethane-sulfonate to produce the O-sil:yl lactim ether/benzimidazole trifluoromethanesulfonate saT~~t. This is reacted with 3,5-di-O-toluyl-alpha-D-erythropentofuanosyl chloride (Hoffer) in THF or acetonitrile in the presence of p-nitro-phenol by the method of Aoyama to give the protected nucle-oside which is purified by silica chromatography. The acyl groups are all removed by a 'two step procedure requiring first, hydrazineolysis with hydrazine/ethanol at room temperature, then evaporation of solvent and heating the crude residue in refluxing s;thanol to fully cleave the phthaloyl residue. The aminobenzimidazole is protected by reaction with 4-(dimethoxymethyl)-morpholine (prepared from 4-formyl morpholine by the general procedure of Bredereck et. al.) in methanol to form the amidine. The remaining reactive site of the benzimidazole is protected by reaction with pivaloyl chloride under the conditions of Example 1.
Alternatively, the final acylation may be done with (di-methylamino) benzoyl chloride. An alternative amino pro-tecting group is formed by reaction of the unprotected benzimidazole with 4-(dimethylamino)benzaldehyde in methanol in the presence of piperidine (10 mole) and methanesulfonic acid (5 mole). The resulting imine is acylated as for the amidine. The primary hydroxyl group is protected with the dimethoxyt:rityl group as per Example 1.
2. Preparation of 2-glycosyl benoxazoles 3-Acetamidophenol (Aldrich Chemical Co.) is nitrated to give the 2-nitro-!5-acetamidophenol. Reduction with hydrogen and palladium/carbon and reaction with tri-fluoroacetic anhydride or trichloroacetic anhydride yields the 2-trihaloacetamido derivative. This is nitrated to give the 4-nitro species and the trihaloacetyl group removed by brief ammonolysis to give 5-acetamido-2-amino-4-nitrophenol.
2,5-Anhydro-3,4,6-tri-O-benzoyl-D-allonothioamide (Dickering) is treated with methyl iodide and sodium hydride to give the corresponding methyl thioimidate.
Alternatively the thioamide is reacted with di-tert-butyl dicarbonate (Aldrich) and 4-dimethylaminopyridine in di-chloromethane to produce thca imide. Alternatively, the imide is treated with methyl iodide or methyl triflate in the presence of diisopropylethylamine to give the N-tert-butoxycarbonyl methyl thioimidate. Any of these are suitable for reaction with aromatic 1,2-diamines or ortho aminophenols to produce be.nzimidazole or benzoxazole derivatives of deoxyribosides, respectively.
The aminophenol is reactead with the appropriate acti-vated thioamide from the previous paragraph to produce the 2-(tri-O-benzoyl-beta-deoxyrihosyl)benzoxazole. The N-acetyl and O-benzoyl groups are removed by ammonolysis or hydrazinolysis and the nitro group reduced with hydrogen and palladium/carbon. The aromatic diamine is reacted with cyanogen bromide in refluxin~g methanol, and the product 6-amino-2-(tri-O-benzoyl-beta-deoxyribosyl)imidazo[4,5-f]
benzoxazole derivative protected as in Example 3A1, and the primary hydroxy protected as per Example 1.

~1 109 88 B. Subunit containing Ribose moiety 1. N-glycosyl isoindoles The ribose nucleoside is prepared as for the deoxyribonucleoside in Example 3A1 except that the O-sily lated lactam is reacted in the: presence of mercuric bromide or silver trifluoromethane~~ulfonate with the ribosyl bromide prepared by treatment of 1-O-acetyl-2,3,5-tri-O-benzoyl-D-ribofuranose with HBr in benzene as per the procedure of Maeba et al.
2. 2-glycosyl benzoxazoles 2,5-Anhydro-3-deoxy-4,6-di-O-toluoyl-D-ribo-hexanothioamide (Pickering) p.s converted into the methyl thioimidate, the imide, or the N-tert- butoxycarbonyl methyl thioimidate as in Example 3A2. Any of these are suitable for reaction with aromatic 1,2-diamines or ortho aminophenols to produce be:nzimidazole or benzoxazole derivatives of ribosides, respectively.
By the same procedures in. Example 3A2, the aminophenol is reacted with the activated. thioamide from the previous paragraph to produce the be~nzoxazole which is further converted into the protected nucleoside by the procedures in Example 3A2.
C. Subunit containing Morpholino moiety.
1. N-glycosyl isoindol~es The morpholine nucls~oside is prepared by reaction of the O-silylated lactam from. Example 3A1 with tetraacetyl alpha-D-glucopyranosyl bromide, (Sigma) (with or without the presence of mercuric bromide or silver trifluoromethane-sulfonate). The glycoside is converted into the morpholino nucleoside in the usual way except that twice the normal amount of sodium periodate is employed. Following N-tritylation (Example 2C) and lhydrazinolysis of the base ~1~.A98g protecting groups, the base is reprotected as in Example 3A1.
Alternatively, the morpholine nucleoside is prepared by reaction of the benzylic bromide from Example 3A1 with beta-D-glucopyranosylamine (Tamura) to give the glycosyl lactam directly. This is converted into the morpholino nucleoside by the usual procedure except that twice the amount of sodium periodate must be employed in the oxida-tion step. Following N-tritylation (Example 2C) and hydrazinolysis of the base, reprotection is accomplished as in Example 3A1.
Alternatively, the methyl 4-acetamido-2-methyl-5-ni-trobenzoate from Example 3A1 is brominated as in Example 3A1 and reacted with beta-D-glucopyranosylamine. The N-acetyl is removed with 1-10% NaOH in 90% ethanol, the nitro is reduced with palladium/carbon and hydrogen, and the aminobenzimidazole is formed by reaction with cyanogen bromide in refluxing ethanol. The aminobenzimidazole is protected as in Example 3A1.
Alternatively, the riboside prepared in Example 3B1 is converted into a morpholine-containing subunit following the procedure in Example 2C. This procedure is accom-plished prior to deacylation of the phthaloyl group from the aminobenzimidalole. After morpholine formation and protection as the N-trityl species, the phthaloyl group is removed as in Example 3A1.
The morpholine nitrogen is protected as the N-trityl by reaction of the free amine or the tosylate salt with trityl chloride in acetonitrile containing triethyamine.
The reaction mix is poured into water and the solid product isolated by filtration and purified by silica gel chromatography.

21 109 88 ~~

2. 2-glycosyl benzoxazoles By the procedures described in Myers , 2 , 3 , 4 , 6-te-tra-O-acetyl-alpha-D-galactopyranosyl bromide is converted into 2,3,4,6-tetra-0-acetyl-alpha-D-galactopyranosyl cyanide and then into the corresponding thioamide by the method of Dickering, et al, and then into its activated thioamide derivatives as in Example 3A2. These are suit-able for reacting with 1,2-di.amines or ortho aminophenols to produce benzimidazoles or benzoxazole derivatives of galactosides, resp. A simil;~r procedure may be employed beginning with other hexose nitriles (Myers).
By the same procedures in Example 3B2, the aminophenol is reacted with the activated thioamide from the paragraph above to produce the benzoxazole which is further converted into the N-protected galactoside by the procedures in Example 3B2. This is converted into the morpholine nucleo-side by the usual procedure .except that twice the normal amount of periodate must be employed in the oxidation step.
The N-trityl group is introduced by the method in Example 3C1.
Example 4 Preparation of 4-Membered High-Specificity Subunit Set Containing Morpholino Backbone Moieties A. CG-specific subunit.
Guanosine is converted into its 2-phenylacetyl deri-vative by the method in Example 1. This is converted into the morpholine nucleoside tosylate salt by the methods in Example 2C. It may be trit~~lated by reaction with tri-phenylmethyl chloride in acetonitrile containing triethyl-amine. The reaction mixture is poured into water and the product filtered. It is purified by recrystallization from acetonitrile.
?r, 21~098g B. TA-specific subunit.
2,6-Diaminopurineriboside is converted into its N2-phenylacetyl N6-benzoyl derivative by the method in Example 1. This is converted into the morpholine nucleoside by the methods in Example 2C. It is tritylated by the procedure in Example 5A
C. AT-specific recognition moiety.
1. 2-glycosylbenzoxazoles 5-Hydroxy-2(3H)-benzoxazolone (Ozdowska) is acetylated with acetic anhydride and then nitrated with cold fuming nitric acid to the 6-vitro-5-acetoxy species. This is dissolved in ethanol and treated with potassium carbonate, than hydrogenated over palladium to reduce the vitro group to an amino group. The isolated aminophenol is reacted with an active thioamide derivative from Example 3C to give the 6-(2,3,4,6-tetra-O-acetyl-galactosyl)-oxazolo[4,5-f]--2(3H)-benzoxazolone. Reaction with phosphoryl chloride followed by ammonolysis gives the 2-aminobenzoxazole. This is N-protected by the usual procedure to prepare the benzoyl, isobutyryl, acetyl, methoxyacetyl, phenoxyacetyl or trichloroacetyl amides.
The morpholine nucleoside is prepared from the galactosyl species above by the procedures in Example 2C
except with double the usual amount of sodium periodate in the oxidation step in order to form the dialdehyde required for reductive amination. The latter step is performed by the usual methods. The morpholine is tritylated as in Example 5A and purified by silica gel chromatography.
2. 2-glycosylisoindoles 2-Methyl-4-hydroxybenzoic acid (King) is nitrated with cold fuming nitric acid to give the 5-vitro derivative which is reduced using palladium catalyst in a hydrogen atomosphere to the 5-amino species. This is converted into 21 10~ 88 the methyl ester by the procedure in Example 3A1. This is converted to the 2-aminobe:nzoxazole using cyanogen bromide and the exocyclic amino group acylated by the methods in Example 1 with acetyl, :methoxyacetyl, trichloroacetyl, 5 isbutyryl or benzoyl. The: compound is converted into the benzylic bromide by the methods in Example 3A1.
The morpholine nucleoside is prepared first by reac-tion of the benzylic bromide with beta-D-glucopyrano-sylamine as in Example 3C. Then, methanolic periodate l0 cleavage using twice the usual amount of sodium periodate and reductive amination give the morpholine nucleoside.
This is tritylated by the procedure in Example 5A and purified by silica gel chromatography.
Alternatively, the bs:nzylic bromide is reacted with 15 ammonia to produce the lactam which is O-silylated with trimethylsilyl trifluorome,thanesulfonate or tert-butyldi methylsilyl trifluorometha.nesulfonate and 2,6-di-tert-bu tylpyridine. The O-silylat:ed lactam is reacted with tetra acetyl alpha-D-glucopyranosyl bromide (with our without the 20 presence of silver trifluoromethanesulfonate or mercuric bromide), followed by ammonolysis and reprotection of the primary amino group as in Example 5C1. The glycoside is converted into the morpholine nucleoside in the usual way except that twice the normal amount of sodium periodate is 25 employed. The morpholine is tritylated as in Example 5A
and purified by silica gel chromatography.
D. GC-specific subunit.
1. 2-glycosylbenzoxazoles 30 5-Chloro-2,4-dinitrop:henol (Carnelley) is treated with chloromethyl benzyl ether and diisopropylethyl amine, and the ether is treated with i:.he sodium salt of methyl cyano-acetate (or malononitrile) followed by reduction with iron in acetic acid. Cleavage of the acetal (hydrogen/palladium 35 on carbon) and reaction with an activated thioimide deriva-21'loaaa tive from Example 3C produces the pyrrolobenzoxazole which, after ammonolysis, may be base protected by the procedure in Example 1 to prepare the benzoyl, isobutyryl, acetyl, methoxyacetyl, phenoxyacetyl or trichloroacetyl amides.
The morpholine nucleoside is prepared by reaction of the galactoside with double the usual amount of sodium periodate in order to form the dialdehyde required for reductive amination. The latter step is performed by the usual methods. The molecule is tritylated by the method in Example 5A and purified by silica gel chromatography.
2. 2-glycosylisoindoles 4-Chloro-2-methylbenzoic acid (Pfaltz and Bauer Chemi cal Co) is converted into its methyl ester (HC1/methanol) and further converted into the benzylic bromide by the pro cedure in Example 3C. Reaction with two equivalents of ammonia provides the lactam which is nitrated in fuming nitric acid to give the 4-vitro-5-chloro-2-oxoisoindole.
The lactam from above is O-silylated as in Example 5C2. The lactim ether is reacted with tetraacetyl alpha-D-glucopyranosyl bromide (with or without the presence of silver trifluoromethanesulfonate or mercuric bromide). This is reacted the sodium salt of methyl cyanoacetate (or malononitrile) followed by reduction with iron in acetic acid. The acyl groups are all removed by ammonolysis and the base reprotected by the usual procedure as the benzoyl, isobutyryl, acetyl, methoxyacetyl, phen-oxyacetyl or trichloroacetyl amides.
Alternatively, 4-chloro-2-methylbenzoic acid is nitrated with fuming nitric acid in concentrated sulfuric acid to give the 5-vitro derivative. Following esteri fication by the method in Example 3A, this is reacted with the sodium salt of methyl cyanoacetate (or malononitrile) followed by reduction with iron in acetic acid. The amine is protected by reaction with trichloroacetic anhydride, ~1 109 88 methoxyacetic anhydride, acetic anhydride, isobutyryl chloride or benzoyl chloride. This is converted into the benzylic bromide by the methods in Example 3C. The benzylic bromide is converted into the lactam glucoside by treatment with beta-D-glucopyranosylamine.
The glucoside above is reacted with methanolic perio-date using twice the usual amount of sodium periodate followed by reductive ami.nation to give the morpholino nucleoside. This is tritylated by the procedure in Example l0 5A and purified by silica gel chromatography.
E. Synthesis of pyrimidopy;ridine.
5-Formyl-2'-deoxyurid:ine (Barwolff and Langen) is dissolved in methanol and treated with manganese dioxide in the presence of sodium cyanide and acetic acid according to the general procedure of Corey to provide the methyl ester.
The ester is reacted with t:ert-butyldimethylsilyl triflate in dichloromethane in the: presence of diisopropylethyl amine to protect the alcohols. The heterocycle is acti vated by the method of Bi:~chofberger (NaH, triisopropyl-benzenesulfonyl chloride, THF). The 4-O-sulfonated hetero-cycle is treated with the tosylate salt of benzhydryl alanine (Aboderin) in the: presence of diisopropylethyl amine in DMF to give the cytosine derivative. The cyto-sinyl alanine derivative i.s oxidized to the dehydroamino acid by the general procedure of Poisel and Schmidt (tert-butyl hypochlorite in THF, followed by one equivalent of potassium tert-butoxide in THF). The product is treated with a catalytic amount of potassium tert-butoxide in hot THF to provide the pyrimidopyridine. The benzhydryl ester is removed by hydrogenolysis using hydrogen over palladium/carbon. The acid is treated with diphenylphos-phoryl azide in benzyl alcohol (or benzyl alcohol/dioxane) containing triethylamine according to Shioiri, et al.
Following hydrogenolysis to cleave the carbamate, and HF-21 109 88 ~' pyridine to remove the silyl groups, the molecule is N-protected as the trichloroacsaamide or phenylacetamide by the usual procedure.
In a similar manner, 5'--formyluridine, prepared from 5-methyluridine by the procedures in Barwolff and Langen, is converted into the corresponding pyrimidopyridine riboside. The riboside is converted into the morpholine nucleoside by the usual procedure, and protected as the N
trityl derivative.
l0 Examx~le 5 Preparation of 4-Membered High-Specificity Subunit Set Containina N-Carboxvmethylmorpholino-5~-amino Backbone Subunits containing rik>ose, galactose, or glucose moieties are prepared as in Example 4, and their respective sugar moieties are converted i~o the N-carboxymethylmorpho-lino-5'-tritylated amine form by the method described in Example 2D.
Example 6 Representative Polymer Assembly Procedures for 2'-O-Methylribose and 2'-Deo:~cyribose-containing subunits The protected 2'-deoxyriboside-containing subunits and the protected 2'-0-methylriboside-containing subunits are converted into their corresponding 3'-H-phosphonate salts by the methods given in Sakataume, Yamane, Takaku, Yamamoto, Nucleic Acids Res. 1990, 18, 3327 and polymerized on solid support by the method in this source. When the assembly of the polymer chain is completE~, the supported molecule is treated with a primary or secondary amine in the presence of either iodine or carbon tei:rachloride as per the method of Froehler, Tetrahedron Lett. 1986, 27, 5575. The phos phoramidate-linked polymer is removed from the support and deprotected by the usual methods involving ammonolysis (See second ref.) iri'i 7 .9 Examx~le 7 Representative Activation Procedures for Morpholino-Containing subunits A. Activation of 5'-Hydroxyl of Morpholino Subunit.
Dimethylaminodichlorophosphate is prepared as follows:
a suspension containing 0.1 mole of dimethylamine hydro chloride in 0.2 mole of phosphorous oxychloride is refluxed for 12 hours and then distil:Led (boiling point is 36°C at 0.5 mm Hg).
Activation of the 5'-hydroxyl of a morpholino-contain-ing subunit prepared as in E~;:ample 2C entails dissolving one mmole of 5'hydroxyl subun:it, base-protected and trity-lated on the morpholino nitrogen, in 20 ml of dichloro-methane. To this solution 4 mmole of N,N-diethylaniline and 1 mmole of 4-methoxypyrid:ine-N-oxide are added. After dissolution, 2 mmole of dimet:hylaminodichlorophosphate is added. After two hours the product is isolated by chroma-tography on silica gel devealoped with 10% acetone/90%
chloroform. The same procedure, except substituting ethyl-dichlorothiophosphate instead of dimethylaminodichlorophos-phate, gives an activated subunit with similar utility.
B. Activation of 5'-Amine of lKorpholino-containing Subunit The 5'hydroxyl of a morpholino-containing subunit, having exocyclic amino groups of the base-pair recognition moiety in the protected form, prepared as in Example 2C can be converted to the amine as follows. To 500 ml of DMSO is added 1.0 mole of pyridine (:Pyr), 0.5 mole of triflouro-acetic acid (TFA), and 0.1 mole of the morpholino subunit.
The mixture is stirred until dissolved, and then 0.5 mole of diisopropylcarbodiimide (17IC) or dicyclohexylcarbodi-imide (DCC) is added. After 2 hours the reaction mixture is added to 8 liters of rapidly stirred brine, which is .~.

WO 93/00352 PCf/US92/05208 ~1109~g stirred for 30 minutes and filtered. The solid is dried briefly, washed with 1 liter of ice cold hexanes, filtered, and the solid is added to 0.2 mole of sodium cyanoboro-hydride in 1 liter of methanol, stirred for 10 minutes, 0.4 5 mole of benzotriazole or p-nitrophenol is added, followed by 0.2 mole of methylamine (40% in HZO) and the preparation is stirred four hours at room temperature [Note: the benzotriazole or p-nitrophenol buffers the reaction mixture to prevent racemization at the 4' carbon of the subunit at 10 the iminium stage of the reductive alkylation]. Finally, the reaction mixture is poured into 5 liters of water, stirred until a good precipitate forms, and the solid is collected and dried. This dried product is next suspended in DMF and 4 equivalents of So3/pyridine complex is added.
15 Over a period of several hours, 8 equivalents of triethyl amine is added dropwise with stirring. After an additional two hours the preparation is dumped into a large volume of brine and the solid collected by filtration and dried.
This sulfamic acid preparation is then purified by silica 20 gel chromatography.
Ten mmole of the triethylamine salt of sulfated sub-unit protected on the recognition moiety and on the nitro-gen of the morpholino ring is dissolved in 10 ml of dichlo-romethane and then 40 mmole of pyridine is added. This 25 solution is chilled for 15 minutes on a bed of dry ice and then 1.1 mmole of phosgene (20% in Toluene) is slowly added while the solution is rapidly stirred. After addition, the solution is allowed to come to room temperature and then washed with aqueous NaHC03, dried, and chromatographed on 30 silica gel eluted with a mixture of chloroform and acetone to give the desired sulfamoyl chloride.
C. Activation of Annular Morpholino Nitrogen This example describes the preparation of a morpholino 35 subunit protected on its 5' oxygen and sulfated on its mor-2t 109 sa pholino ring nitrogen. Morpholino-containing subunit pre-pared as in Example 2C, but not carried through the last tritylation step, is silylate.d on its 5' hydroxyl with t-butyldimethlsilyl chloride. This product is then treated with S03/pyridine complex (with excess pyridine) in di-methylformamide (DMF) to give a sulfamic acid on the annular morpholino nitrogen.
It should be mentioned that the salts of sulfamic acids can be chromatographed on silica gel using triethyl amine/methanol/chloroform mixtures if the silica is first pre-eluted with 2~ triethylamine in chloroform.
This sulfamic acid on the morpholino nitrogen is con-verted to the sulfamoyl chloride and purified as in Example 7B above.
D. Activation of N-Carboxym~ethyl of Morpholino Carboxylate-containing subunits, such as prepared in Examples 2D and 2E, are activ<~ted as follows. Ten mmol of the subunit is dissolved in 17MF containing 20 mmol of p nitrophenol and 15 mmol of dic.yclohexylcarbodiimide. After 1 hour the product is rotovaped and then purified by silica gel chromatography developed with a mixture of acetone and chloroform.
Example 8 Representative Solid-Phase Polymer Assembly of Morpholino-containing Subunits This example describes a method which is generally applicable for assembly of aci:.ivated subunits, prepared as in Examples 7A and 7B, to give phosphorodiamidate-linked, ethylthiophosphoramidate-linked, and sulfamate-linked binding polymers. A similar scheme wherein the coupling step includes the addition of silver trifluoromethanesul-fonate, and use of N,N-diisopropyl-2-methoxyethylamine instead of diisopropylethanolamine, is suitable for assembly of subunits prepared as in Example 7C
to give sulfamate-linked polymers. A similar scheme, wherein the coupling step is carried out in dimethylform-amide instead of dichloromethane, is suitable for assembly of subunits activated as in Example 7D to give amide-linked polymers.
A. Linker l0 Aminomethyl polystyrene resin (Catalog no. A1160, from Sigma Chemical Co.) 1% divinylbenzene crosslinked, 200 to 400 mesh, 1.1 mmol of N per gram, is suspended in dichloro-methane and transferred to a :L cm diameter column having a frit on the bottom, to give a resin bed volume of 2.5 ml.
One mmol of bis[2-(succ:inimidooxycarbonyloxy)ethyl]
sulfone (Pierce Chemical Co. of Rockford, Illinois, USA) is added to a dichloromethane solution containing 1 mmol of N-tritylated piperazine. After 2 hours the reaction mixture is chromatographed on silica gel developed with an acetone/
chloroform mixture to give a mono-activated beta-elimina-tion-cleavable linker.
134 micromole of the above linker is dissolved in 1 ml of dichloromethane and added i:o the resin in the synthesis column and the resin suspension agitated for 3 hours at 30 deg. C. Next, 1 mmol of d_Lisopropylaminoethanol and 1 mmole of acetic anhydride is added and agitation continued for 10 minutes, followed by addition of 2 mmole of benzyl-methylamine and agitation for' 20 minutes. The column is washed with 30 ml dichloromethane. Based on release of trityl, the above procedure typically gives on the order of 100 to 110 micromoles of bound linker.
B. Coupling cycle (Detritylation/Coupling/Capping) L

21 109 88 ~w 8 .3 The coupling cycle described below is used for adding each subunit in an order appropriate to give a polymer having the desired sequence of subunits.
i) Detritylation. To the column is added a solution con-taining 53 ml of dichlorometha.ne, 6 ml of trifluoroethanol, and 1 gram of cyanoacetic acid. After this solution has passed through, the column is washed with 40 ml of dichloromethane, followed by 20 ml of dichloromethane containing 4 mmol of diisopropylaminoethanol. The column is then washed with 10 ml of ~dichloromethane.
ii) Coupling. To the column is added 1 ml of dichloro-methane containing 120 microliter of diisopropylamino-ethanol to 0.25 mmol of activated subunit (prepared as in Example 7A or 7B) followed by agitation at 37 deg. C for 1 hr. The column is washed with 30 ml dichloromethane.
Note: excess unreacted activated subunit can be convenient ly recovered simply by adding 4 volumes of hexane to this eluant and filtering.
iii) Capping. To the column is added 2 ml of dichloro-methane containing 1 mmol of diisopropylaminoethanol and 1 mmol of acetic anhydride, followed by agitation at 37 deg.
C for 10 minutes. Added to the column is 10 ml of dichlo-romethane containing 1 mmol o~f benzylmethylamine, and the resin bed is agitated at 37 d<ag. C for 20 min. The column is then washed with 30 ml dicl'zloromethane.
C. Cleavage from support and deprotection After all the subunits have been added by the above coupling procedure, the full length polymer is cleaved from the support by eluting the co:Lumn with a solution consist-ing of 2.5 ml of diethylmalonate, 5 ml of 1,8-diazabi-cyclo[5.4.0] undec-7-ene, and 43 ml dichloromethane. The polymer is then precipitated from this eluant by adding ether.
If it is desirable to add a moiety to enhance aqueous solubility, or to enhance target binding affinity, or to facilitate uptake by specific: cell or tissue types, then the secondary aliphatic amine generated upon cleavage from the polystyrene support provides an excellent site for attachment of said moieties <~t this stage of the polymer preparation.
The polymer product is next dissolved in DMF and an equal volume of conc. NH40H added, the preparation capped tightly, and incubated 18 hrs at 37 deg. C. Subsequently, the preparation is dried under reduced pressure to give a polymer preparation wherein the base-pair recognition moieties are deprotected and at one end of the polymer is a trityl moiety, and at the other end is a secondary aliphatic amine - which, as noted above, may be derivatized prior to the ammonia treatment.
Example 9 Polymer Purification Methods The full-length polymer having a terminal trityl moiety (typically greater than 50~ of the total mass of the preparation for a 24-subunit long polymer) can be separated from the capped failure sequences by low pressure chromato-graphy on a column of chromatographic grade polypropylene (Catalog No. 4342 from PolySciences Inc.) developed with an acetonitrile/water gradient, with the eluant monitored photometrically at 254 nm. Pu:rifications generally proceed mare favorably when the polymer is suspended in water and then the solution adjusted to pH 11 with dimethylamine and the eluting solvents also adjusted to pH 11 with dimethyl-amine. In this system, the tritylated full-length polymer elutes appreciably later than the non-trityl-containing capped failure sequences.

The fractions containing full-length polymer are col-lected and dried down under reduced pressure. The polymer preparation is then detrity7Lated by suspending in tri-fluoroethanol (1 g polymer i.n 25 ml TFE) and 1.5 ml of 5 mercaptoacetic acid added. After 10 minutes, 100 ml of ether is added and the final pure product collected by centrifugation or filtration.
Examp.Le 10 10 Polymer Assembly Via Novel Oxidation~Rina Closure/Reduction Method A. Synthesis support The solid support used in this synthesis should be 15 hydrophilic, but should not contain vicinyl hydroxyls. An aqueous slurry of Macro-Prep 50 CM (Catalog No. 156-0070 from Bio-Rad Laboratories, Richmond, Calif., USA) is added to a fritted column to give a 5 ml packed bed volume (con taining approximately 1 mmol of carboxylate). The synthe 20 sis support is then washed with 100 ml of 0.1 N HC1 and then 50 ml water. 50 ml of: DMF (dimethylformamide) is passed through the column and drained. 5 ml of DMF
containing 5 mmol of diisopropylcarbodiimide and 5 mmol of p-nitrophenol is added, followed by incubation with 25 agitation at 30 deg. C for 3 hours. The column is washed with 100 ml of DMF, followed by addition of 20 mmol of piperazine in 10 ml of DMF, a.nd agitation for 15 minutes.
The column is then washed with 50 ml of DMF and drained.
30 B. Addition of linker and first subunit To 1 mmol of a ribose-containing subunit having a carbazate moiety at the 5' of the ribose (prepared as in Example 2F) in 5 ml of DMF, is added 3 mmol of bis[2-(suc-cinimidooxycarbonyloxy)-ethyl]sulfone (Pierce Chemical Co.
35 of Rockford, Illinois, USA), followed by incubating at 30 deg. C for 3 hours. To the reaction mixture is added ether and the ;, ~.D ,.

8,~
precipitate is collected. ThE~ precipitated linker-subunit is washed with ether, resuspended in 5 ml of DMF, added to the synthesis support, and incubated with agitation for 3 hrs at 30 deg. C. The support. is washed with 50 ml of DMF, and then with 100 ml of water.
C. Coupling cycle i) Oxidation of vicinyl hydroxyls 5 mmol of sodium periodate is dissolved in 10 ml of water, added to column, and agitated for 10 minutes. The column is washed with 50 ml of water and then drained.
ii) Morpholino ring closure/reduction 2 mmol of sodium cyanoborohydride is dissolved in 5 ml of water, the pH adjusted to between 7 and 8 with trimethyl acetic acid, to which is addedl 1.5 mmol of the next ribose containing 5'-carbazate subunit, followed by addition to the column containing the synthesis support. The column is incubated with agitation for 30 min at 30 deg. C. Formic acid is added to reduce the pH: to between 3 and 4, followed by incubation at 30 deg. C for 10 minutes. The wash column is washed with 100 ml of water.
This coupling cycle is repeated until all subunits have been added to give the desired full-length polymer.
Addition of terminal moieties If it is desirable to a.dd to the binding polymer a moiety to enhance aqueous solubility, or to enhance target binding affinity, or to facilitate uptake by specific cell or tissue types, this can be conveniently achieved at this stage by oxidizing the vicin.yl hydroxyls of the terminal subunit of the polymer and, by the morpholino ring closure/reduction procedure described above, adding said moieties containing a primary aliphatic amine.

2~ 109 8a 8~
Cleavaqe from the support After all the subunits of the polymer, and any desired additional groups, have been added by the above coupling procedure, the polymer i.s c,leaved from the support by washing the column with 50 ml of DMF, and then eluting the column with a solution consisting of 2.5 ml of diethyl-malonate, 5 ml of 1,8-diazabicyclo[5.4.0]undec-7-ene, and 43 ml of DMF. The polymer is then precipitated from this eluant by adding ether.
The full-length po7.ymer can be purified by low pressure chromatography on a column of chromatographic grade polypropylene (Catalog No. 4342 from PolySciences Inc.) developed with an acetonitrile/water gradient, with the eluant monitored photometrically at 254 nm. Purifica-tions generally go better when the polymer is suspended in water and then the solution adjusted to pH il with di-methylamine and the eluting solvents also adjusted to pH 11 with dimethyl amine.
E~,:ample 11 Polymer Structural Characterization.
NMR, and even two-dimensional NMR, appears to provide little useful structural information for these heteropoly-mers when they are of any significant length. Likewise, elemental analysis has not. been found to be of value.
Polymers prepared as in Example 8 and cleaved from the solid support, but not yet treated with ammonium hydroxide, generally show relatively clean parent ions for polymers up to about 16 to 18 subuni.ts in length, when assessed by positive fast atom bombardment mass spectrometry. For longer polymers, and for polymers lacking protective groups on the bases ( such as prepared in Example 10 ) , ef f ective mass analysis requires procedures such as laser desorption or electro spray.

$$
Although the invention has been described with respect to particular polymer subunits, methods of preparing the subunits, and polymer assembly, it will be appreciated that various modifications and changes may be made without departing from the invention.

Claims (20)

CLAIMS:
1. A polymer composition effective to bind in a sequence-specific manner to a target sequence of a duplex polynucleotide containing at least two different-oriented Watson/Crick base-pairs at selected position in the target sequence, comprising a specific sequence of subunits selected from the forms:
where Y is a 2- or 3-atom length, uncharged subunit linkage group; R' is H, OH, or O-alkyl; the 5'-methylene has a .beta.
stereochemical orientation in the 5-membered ring and a uniform stereochemical orientation in the 6-membered ring;
R i has a .beta. stereochemical orientation; and at least about 70% of R i groups in the polymer are selected from two or more of the following base-pair-specificity groups and at least one R i is selected from group (c) or group (d):
(a) for a T:A or U:A oriented base-pair, R i is 2,6 diaminopurine:
(b) for a C:G oriented base-pair, R i is guanine or 6-thioguanine;
(c) for a G:C oriented base-pair, R i is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B indicates the polymer backbone:
where the * ring position may carry as part of the ring or directly attached thereto a hydrogen-bond acceptor group containing an atom selected from the group consisting of N, O, S, F, C1 and Br; and, (d) for an A:T or A:U oriented base-pair, R i is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B indicates the polymer backbone:
where the * ring position may carry directly attached thereto a hydrogen-bond donating group selected from either H or NH2 and wherein up to about 30% of said ~ groups comprise low or non-specificity bases selected from the group consisting of cytosine, uracil, thymine and adenine.
2. The polymer composition of claim 1, containing one or more subunits of the form:
3. The polymer composition of claim 1, containing one or more subunits of the form:
4. The polymer composition of claim 3, containing one or more subunits of the form:
5. The polymer composition of claim 1, for use in sequence-specific binding to a B-form DNA-DNA duplex nucleic acid, wherein the Y in each of the subunits is three atoms in length.
6. The polymer composition of claim 5, wherein one or more subunits of the polymer are selected from the group consisting of:
7. The polymer composition of claim 6, wherein one or more subunits of the polymer are selected from the group consisting of:
8. The polymer composition of claim 1, for use in sequence-specific binding to an A-form duplex nucleic acid, wherein the Y in each of the subunits is two atoms in length.
9. The polymer composition of claim 8, wherein one or more subunits of the polymer are: selected from the group consisting of:

~~
10. The polymer composition of claim 8, wherein one or more subunits of the polymer are selected from the group consisting of:

~~
11. The polymer composition of claim 1, wherein the R i structure is selected from the group consisting of:

~ ~~ ~ ~~ ~ ~~
12. The polymer of claim 1, wherein the R i structure specific for a G:C target orientation is selected from the group consisting of the following bases:

13. The polymer of claim 1, wherein the R i structure specific for a A:T target orientation is selected from the group consisting of the following bases:

14. The polymer composition of claim 1, wherein up to about 30% of the R i groups in the polymer are cytosine, at polymer subunits corresponding to a G:C base-pair orientation in the target sequence, and thymine, at polymer subunits corresponding to A:T or A:U base-pair orientations in the target sequence.
15. The composition of claim 1, wherein the polymer contains one or more attached moieties effective to enhance the solubility of the polymer in aqueous medium.
16. A method for coupling a first free or polymer-terminal subunit having one of the following subunit forms:

where R i is a planar ring structure having two or more hydrogen-bonding sites, with a second free or polymer-terminal subunit having one of the following subunit forms:

where Z is a 2-atom or 3-atom long moiety, said method comprising:
i) oxidizing the first subunit to generate a dialdehyde intermediate;
ii) contacting the dialdehyde intermediate with the second subunit under conditions effective to couple a primary amine to a dialdehyde; and (iii) adding a reducing agent effective to give a coupled structure selected from the following forms:

~~ ~~
17. A method for isolating, from a liquid sample, a target duplex nucleic acid fragment having a selected sequence of base-pairs, comprising:
i) contacting the sample with a polymer reagent composed of the polymer composition of claim 1 adapted to permit isolation of said reagent from solution and having a subunit sequence effective to bind in a sequence-specific manner with the selected sequence of base-pairs, under conditions effective for sequence-specific binding of the polymer composition to the selected sequence of base-pairs;
and ii) separating the polymer reagent from the fluid sample.
18. The method of claim 17, for use in detecting the presence of such target fragment in a liquid sample, which further includes testing the separated polymer reagent for the presence of bound duplex nucleic acid.
19. The method of claim 18, wherein said polymer reagent includes said polymer composition bound to a solid support, and said testing includes adding to the duplex nucleic acid, a fluorescent compound effective to intercalate into duplex DNA.
20. A subunit composition for use in forming a polymer composition effective to bind in a sequence specific manner to a target sequence in a duplex polynucleotide, comprising one of the following subunit structures:

where R' is H, OH, or O-alkyl; the 5'-methylene has a .beta.
stereochemical orientation in subunit forms (a), (c), and (d) and a uniform stereochemical orientation in subunit form (b); X is hydrogen or a protective group or a linking group suitable for joining the subunits in any selected order into a linear polymer; Y is a nucleophilic or electrophilic linking group suitable for joining the subunits in any selected order into a linear polymer; and X and Y together are such that when two subunits of the subunit set are linked the resulting intersubunit linkage is 2 or 3 atoms in length and uncharged; Z is a 2-atom or 3-atom long moiety; and, R i, which may be in the protected state and has a .beta. stereochemical orientation, is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B indicates the aliphatic backbone moiety:

where the * ring position may carry as part of the ring or directly attached thereto a hydrogen-bond acceptor group containing an atom selected from the group consisting of N, O, S, F, Cl and Br; or, R i is selected from the group consisting of planar bases having the following skeletal ring structures and hydrogen bonding arrays, where B indicates the aliphatic backbone moiety:
where the * ring position may carry directly attached thereto a hydrogen-bond donating group selected from either H or NH2.
CA002110988A 1991-06-20 1992-06-18 Sequence-specific binding polymers for duplex nucleic acids Expired - Fee Related CA2110988C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US07/719,732 US5166315A (en) 1989-12-20 1991-06-20 Sequence-specific binding polymers for duplex nucleic acids
US07/719,732 1991-06-20
PCT/US1992/005208 WO1993000352A1 (en) 1991-06-20 1992-06-18 Sequence-specific binding polymers for duplex nucleic acids

Publications (2)

Publication Number Publication Date
CA2110988A1 CA2110988A1 (en) 1993-01-07
CA2110988C true CA2110988C (en) 1999-08-31

Family

ID=24891147

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002110988A Expired - Fee Related CA2110988C (en) 1991-06-20 1992-06-18 Sequence-specific binding polymers for duplex nucleic acids

Country Status (10)

Country Link
US (1) US5166315A (en)
EP (1) EP0592511B1 (en)
JP (1) JPH06508526A (en)
KR (1) KR100225090B1 (en)
AT (1) ATE157098T1 (en)
AU (1) AU665560B2 (en)
CA (1) CA2110988C (en)
DE (1) DE69221730T2 (en)
TW (1) TW466243B (en)
WO (1) WO1993000352A1 (en)

Families Citing this family (989)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5405938A (en) * 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5506337A (en) * 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
US5719262A (en) * 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US6414112B1 (en) 1991-05-24 2002-07-02 Ole Buchardt Peptide nucleic acids having 2,6-diaminopurine nucleobases
US6713602B1 (en) * 1991-05-24 2004-03-30 Ole Buchardt Synthetic procedures for peptide nucleic acids
US5766855A (en) * 1991-05-24 1998-06-16 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity and sequence specificity
US5714331A (en) * 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US7223833B1 (en) * 1991-05-24 2007-05-29 Isis Pharmaceuticals, Inc. Peptide nucleic acid conjugates
DK51092D0 (en) * 1991-05-24 1992-04-15 Ole Buchardt OLIGONUCLEOTIDE ANALOGUE DESCRIBED BY PEN, MONOMERIC SYNTHONES AND PROCEDURES FOR PREPARING THEREOF, AND APPLICATIONS THEREOF
US6451968B1 (en) 1991-05-24 2002-09-17 Isis Pharmaceuticals, Inc. Peptide nucleic acids
US6441130B1 (en) 1991-05-24 2002-08-27 Isis Pharmaceuticals, Inc. Linked peptide nucleic acids
US5726014A (en) * 1991-06-27 1998-03-10 Genelabs Technologies, Inc. Screening assay for the detection of DNA-binding molecules
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US8153602B1 (en) 1991-11-19 2012-04-10 Isis Pharmaceuticals, Inc. Composition and methods for the pulmonary delivery of nucleic acids
US5473060A (en) * 1993-07-02 1995-12-05 Lynx Therapeutics, Inc. Oligonucleotide clamps having diagnostic applications
JP3484197B2 (en) 1993-09-03 2004-01-06 アイシス・ファーマシューティカルス・インコーポレーテッド Amine derivatized nucleosides and oligonucleosides
WO1998003542A1 (en) * 1996-07-24 1998-01-29 Buchardt, Dorte Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US6710164B1 (en) 1993-11-22 2004-03-23 Peter E. Nielsen Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
GB2284208A (en) * 1993-11-25 1995-05-31 Pna Diagnostics As Nucleic acid analogues with a chelating functionality for metal ions
WO1995018971A1 (en) * 1994-01-11 1995-07-13 Affymax Technologies N.V. Methods for the solid phase synthesis of glycoconjugates
US7038026B2 (en) 2000-05-26 2006-05-02 Centelion Purification of a triple heli formation with an immobilized oligonucleotide
FR2728264B1 (en) * 1994-12-16 1997-01-31 Rhone Poulenc Rorer Sa DNA PURIFICATION BY TRIPLE PROPELLER FORMATION WITH A IMMOBILIZED OLIGONUCLEOTIDE
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
JPH11506605A (en) * 1995-06-07 1999-06-15 アボツト・ラボラトリーズ Method of masking probes to reduce background in amplification reactions
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20030044941A1 (en) 1996-06-06 2003-03-06 Crooke Stanley T. Human RNase III and compositions and uses thereof
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US6046004A (en) * 1997-02-27 2000-04-04 Lorne Park Research, Inc. Solution hybridization of nucleic acids with antisense probes having modified backbones
US6251591B1 (en) 1997-02-27 2001-06-26 Lorne Park Research, Inc. Quantitative method for detecting nucleotide concentration
JP2002510319A (en) 1997-07-01 2002-04-02 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for delivery of oligonucleotides through the gastrointestinal tract
US6383808B1 (en) 2000-09-11 2002-05-07 Isis Pharmaceuticals, Inc. Antisense inhibition of clusterin expression
US7321828B2 (en) 1998-04-13 2008-01-22 Isis Pharmaceuticals, Inc. System of components for preparing oligonucleotides
US20040186071A1 (en) 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
EP1469009A2 (en) * 1998-05-21 2004-10-20 Isis Parmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
WO1999060167A1 (en) 1998-05-21 1999-11-25 Isis Pharmaceuticals, Inc. Compositions and methods for topical delivery of oligonucleotides
US6255050B1 (en) 1998-05-22 2001-07-03 Lorne Park Research, Inc. Dynamic hybridization system
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6867294B1 (en) * 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6225293B1 (en) 1998-09-02 2001-05-01 Isis Pharmaceuticals, Inc. Methods and compounds for tracking the biodistribution of macromolecule-carrier combinations
US6335194B1 (en) 1998-09-29 2002-01-01 Isis Pharmaceuticals, Inc. Antisense modulation of survivin expression
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
EP1117762A4 (en) 1998-09-29 2004-02-25 Gamida Cell Ltd Methods of controlling proliferation and differentiation of stem and progenitor cells
US6300320B1 (en) 1999-01-05 2001-10-09 Isis Pharmaceuticals, Inc. Modulation of c-jun using inhibitors of protein kinase C
US6127124A (en) * 1999-01-20 2000-10-03 Isis Pharmaceuticals, Inc. Fluorescence based nuclease assay
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US7534605B2 (en) 1999-06-08 2009-05-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem CD44 polypeptides, polynucleotides encoding same, antibodies directed thereagainst and method of using same for diagnosing and treating inflammatory diseases
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US7332275B2 (en) 1999-10-13 2008-02-19 Sequenom, Inc. Methods for detecting methylated nucleotides
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
EP1290217A2 (en) * 2000-02-04 2003-03-12 Aeomica, Inc. Methods and apparatus for predicting, confirming, and displaying functional information derived from genomic sequence
AU2001253408C1 (en) * 2000-04-13 2008-12-04 Mervyn Merrilees Therapeutic compounds and methods for formulating V3, A Versican Isoform
WO2001083740A2 (en) 2000-05-04 2001-11-08 Avi Biopharma, Inc. Splice-region antisense composition and method
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
US6686188B2 (en) * 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
AU2001271873A1 (en) 2000-07-06 2002-01-21 Avi Biopharma, Inc. Transforming growth factor beta (TGF-beta) blocking agent-treated stem cell composition and method
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
US8568766B2 (en) 2000-08-24 2013-10-29 Gattadahalli M. Anantharamaiah Peptides and peptide mimetics to treat pathologies associated with eye disease
JP5305553B2 (en) 2000-10-12 2013-10-02 ユニバーシティー オブ ロチェスター Composition for inhibiting the growth of cancer cells
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US6573051B2 (en) * 2001-03-09 2003-06-03 Molecular Staging, Inc. Open circle probes with intramolecular stem structures
CA2633171C (en) 2001-06-20 2012-11-20 Genentech, Inc. Antibodies against tumor-associated antigenic target (tat) polypeptides
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
CA2790034A1 (en) 2001-06-21 2003-01-03 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US20040096880A1 (en) * 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
EP1423537A4 (en) * 2001-08-07 2006-11-29 Univ Delaware Compositions and methods for the prevention and treatment of huntington's disease
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US20060068405A1 (en) * 2004-01-27 2006-03-30 Alex Diber Methods and systems for annotating biomolecular sequences
NZ573831A (en) 2001-09-18 2010-07-30 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor, particularly breast tumor - TAT193
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
NZ566396A (en) 2001-10-09 2009-07-31 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expressions
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
NZ533933A (en) 2002-01-02 2008-06-30 Genentech Inc Compositions and methods for the diagnosis and treatment of glioma tumor
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US7553619B2 (en) 2002-02-08 2009-06-30 Qiagen Gmbh Detection method using dissociated rolling circle amplification
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
CA2481507A1 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2484676A1 (en) 2002-05-03 2003-11-13 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof, and related methods
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
AU2003231912A1 (en) * 2002-06-12 2003-12-31 Tel Aviv Medical Center Research Development Fund Methods of detecting and treating prostate cancer
AU2003257181A1 (en) 2002-08-05 2004-02-23 University Of Rochester Protein transducing domain/deaminase chimeric proteins, related compounds, and uses thereof
CN1694959B (en) 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
JP2006500030A (en) * 2002-09-20 2006-01-05 イェール ユニバーシティ Riboswitch, method of using the same, and composition for use with riboswitch
US7229976B2 (en) 2002-09-26 2007-06-12 Isis Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
WO2004044132A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
US9150606B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US9150605B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
CA2504720C (en) 2002-11-05 2013-12-24 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
EP1569695B1 (en) 2002-11-13 2013-05-15 Genzyme Corporation Antisense modulation of apolipoprotein b expression
DK2336318T3 (en) 2002-11-13 2013-07-15 Genzyme Corp ANTISENSE MODULATION OF APOLIPOPROTEIN B EXPRESSION
US20060009378A1 (en) 2002-11-14 2006-01-12 Itshak Golan Novel galectin sequences and compositions and methods utilizing same for treating or diagnosing arthritis and other chronic inflammatory diseases
US8007804B2 (en) 2002-11-15 2011-08-30 Musc Foundation For Research Development Complement receptor 2 targeted complement modulators
JP4555089B2 (en) 2002-11-15 2010-09-29 モーフオテク・インコーポレーテツド Method for producing high production amount of antibody from hybridoma created by in vitro immunization
AU2003294462C1 (en) 2002-11-21 2011-06-30 University Of Utah Research Foundation Purinergic modulation of smell
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
WO2004058987A2 (en) 2002-12-20 2004-07-15 Qiagen Gmbh Nucleic acid amplification
US9487823B2 (en) 2002-12-20 2016-11-08 Qiagen Gmbh Nucleic acid amplification
US6977153B2 (en) * 2002-12-31 2005-12-20 Qiagen Gmbh Rolling circle amplification of RNA
DK1597366T3 (en) 2003-02-11 2013-02-25 Antisense Therapeutics Ltd Modulation of expression of insulin-like growth factor receptor I
US7002006B2 (en) * 2003-02-12 2006-02-21 Isis Pharmaceuticals, Inc. Protection of nucleosides
KR101130181B1 (en) 2003-02-27 2012-03-28 예다 리서치 앤드 디벨럽먼트 캄파니 리미티드 Nucleic acid molecules, polypeptides, antibodies and compositions containing same useful for treating and detecting influenza virus infection
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US8043834B2 (en) 2003-03-31 2011-10-25 Qiagen Gmbh Universal reagents for rolling circle amplification and methods of use
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US7468418B2 (en) 2003-04-29 2008-12-23 Avi Biopharma., Inc. Compositions for enhancing transport of molecules into cells
WO2004108081A2 (en) 2003-06-02 2004-12-16 Isis Pharmaceuticals, Inc. Oligonucleotide synthesis with alternative solvents
AU2004253455B2 (en) 2003-06-03 2011-03-03 Eli Lilly And Company Modulation of survivin expression
EP3604537B1 (en) 2003-06-13 2021-12-08 Alnylam Europe AG Double-stranded ribonucleic acid with increased effectiveness in an organism
US7683036B2 (en) 2003-07-31 2010-03-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
WO2005027833A2 (en) * 2003-09-12 2005-03-31 Avi Biopharma, Inc. Compound and method for treating androgen-independent prostate cancer
AU2004274021B2 (en) 2003-09-18 2009-08-13 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
AR045937A1 (en) 2003-09-18 2005-11-16 Lilly Co Eli MODULATION OF THE EXPRESSION OF THE EIF4E EUCARY INITIATION FACTOR
US20050222068A1 (en) * 2003-10-23 2005-10-06 Mourich Dan V Method and antisense composition for selective inhibition of HIV infection in hematopoietic cells
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
PT2161283E (en) 2003-11-17 2014-08-29 Genentech Inc Compositions comprising antibodies against cd79b conjugated to a growth inhibitory agent or cytotoxic agent and methods for the treatment of tumor of hematopoietic origin
WO2006054296A2 (en) 2004-11-17 2006-05-26 Spectrum Dynamics Llc Methods of detecting prostate cancer
EP1711606A2 (en) 2004-01-20 2006-10-18 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US8778900B2 (en) 2004-01-22 2014-07-15 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP1 expression
WO2005072527A2 (en) * 2004-01-23 2005-08-11 Avi Biopharma, Inc. Antisense oligomers and methods for inducing immune tolerance and immunosuppression
US7842459B2 (en) * 2004-01-27 2010-11-30 Compugen Ltd. Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
EP2700720A3 (en) 2004-03-15 2015-01-28 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNASE H
WO2005097817A2 (en) 2004-04-05 2005-10-20 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
JP4584987B2 (en) 2004-04-30 2010-11-24 アルニラム ファーマスーティカルズ インコーポレイテッド Oligonucleotides containing C5-modified pyrimidines
AU2005326817B2 (en) 2004-05-21 2012-04-19 Benaroya Research Institute Variable lymphocyte receptors, related polypeptides and nucleic acids, and uses thereof
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US20090048192A1 (en) * 2004-06-03 2009-02-19 Isis Pharmaceuticals, Inc. Double Strand Compositions Comprising Differentially Modified Strands for Use in Gene Modulation
USRE48960E1 (en) 2004-06-28 2022-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US7807816B2 (en) 2004-06-28 2010-10-05 University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
PT1766012E (en) 2004-07-02 2011-09-05 Avi Biopharma Inc Antisense antibacterial method and compound
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US8129352B2 (en) 2004-09-16 2012-03-06 Avi Biopharma, Inc. Antisense antiviral compound and method for treating ssRNA viral infection
EP1799812A4 (en) 2004-09-16 2009-09-09 Gamida Cell Ltd Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
DK1809303T3 (en) 2004-09-23 2019-06-11 Arc Medical Devices Inc PHARMACEUTICAL COMPOSITION AND METHODS IN RELATION TO FIBROSE ADHESION OR INFLAMMATORIC DISEASE USING LOW SULPHATE FUCAN
US7524829B2 (en) * 2004-11-01 2009-04-28 Avi Biopharma, Inc. Antisense antiviral compounds and methods for treating a filovirus infection
EP1843819A2 (en) * 2004-11-15 2007-10-17 Obe Therapy Biotechnology S.A.S. Methods of reducing body fat
CA2596506C (en) 2005-02-09 2021-04-06 Avi Biopharma, Inc. Antisense composition and method for treating muscle atrophy
AU2006223498A1 (en) 2005-03-10 2006-09-21 Genentech, Inc. Methods and compositions for modulating vascular integrity
US7476733B2 (en) 2005-03-25 2009-01-13 The United States Of America As Represented By The Department Of Health And Human Services Development of a real-time PCR assay for detection of pneumococcal DNA and diagnosis of pneumococccal disease
US20060240032A1 (en) * 2005-03-31 2006-10-26 Hinrichs David J Immunomodulating compositions and methods for use in the treatment of human autoimmune diseases
EP1863908B1 (en) 2005-04-01 2010-11-17 Qiagen GmbH Reverse transcription and amplification of rna with simultaneous degradation of dna
US9505867B2 (en) 2005-05-31 2016-11-29 Ecole Polytechmique Fédérale De Lausanne Triblock copolymers for cytoplasmic delivery of gene-based drugs
WO2006133022A2 (en) 2005-06-03 2006-12-14 The Johns Hopkins University Compositions and methods for decreasing microrna expression for the treatment of neoplasia
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
EP1919930B1 (en) * 2005-07-01 2016-09-14 Dako Denmark A/S New nucleic acid base pairs
WO2007007317A1 (en) * 2005-07-07 2007-01-18 Yissum Research Development Company Of The Hebrew University Of Jerusalem Nucleic acid agents for downregulating h19, and methods of using same
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US7790694B2 (en) * 2005-07-13 2010-09-07 Avi Biopharma Inc. Antisense antibacterial method and compound
EP1915161B1 (en) 2005-07-13 2017-04-26 Sarepta Therapeutics, Inc. Antisense antibacterial method and compound
US7776532B2 (en) 2005-08-11 2010-08-17 Synthetic Genomics, Inc. Method for in vitro recombination
CA2617693A1 (en) 2005-08-17 2007-02-22 Medexis S.A. Composition and method for determination of ck19 expression
JP5523705B2 (en) 2005-08-29 2014-06-18 レグルス・セラピューティクス・インコーポレイテッド Method of using to modulate MIR-122A
WO2007030691A2 (en) * 2005-09-08 2007-03-15 Avi Biopharma, Inc. Antisense antiviral compound and method for treating picornavirus infection
EP1937278B1 (en) * 2005-09-08 2012-07-25 AVI BioPharma, Inc. Antisense antiviral compound and method for treating picornavirus infection
EP1762627A1 (en) 2005-09-09 2007-03-14 Qiagen GmbH Method for the activation of a nucleic acid for performing a polymerase reaction
US8906609B1 (en) 2005-09-26 2014-12-09 Arrowhead Center, Inc. Label-free biomolecule sensor based on surface charge modulated ionic conductance
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for diagnosis of preeclamsia
EP2402435A3 (en) 2005-10-14 2012-03-28 MUSC Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
US8080534B2 (en) 2005-10-14 2011-12-20 Phigenix, Inc Targeting PAX2 for the treatment of breast cancer
EP2325315B1 (en) 2005-10-28 2014-05-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of huntingtin gene
US8501704B2 (en) 2005-11-08 2013-08-06 Sarepta Therapeutics, Inc. Immunosuppression compound and treatment method
CA2626690A1 (en) 2005-11-09 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor v leiden mutant gene
US7807652B2 (en) 2005-11-21 2010-10-05 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
US8313901B2 (en) * 2005-12-21 2012-11-20 Yale University Methods and compositions related to the modulation of riboswitches
EP1976567B1 (en) 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
EP1973945A4 (en) 2006-01-16 2009-11-18 Compugen Ltd Novel nucleotide and amino acid sequences, and methods of use thereof for diagnosis
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
CA2640058C (en) 2006-01-27 2018-04-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for the use in modulation of micrornas
DK2314594T3 (en) 2006-01-27 2014-10-27 Isis Pharmaceuticals Inc 6-modified bicyclic nucleic acid analogues
WO2007103529A2 (en) * 2006-03-07 2007-09-13 Avi Biopharma, Inc. Antisense antiviral compound and method for treating arenavirus infection
ES2544861T3 (en) 2006-03-31 2015-09-04 Alnylam Pharmaceuticals Inc. Compositions and methods to inhibit the expression of the Eg5 gene
MX2008014005A (en) 2006-05-03 2009-01-27 Baltic Technology Dev Ltd Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease.
EP2023939B1 (en) 2006-05-05 2012-06-27 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of pcsk9
US8785407B2 (en) * 2006-05-10 2014-07-22 Sarepta Therapeutics, Inc. Antisense antiviral agent and method for treating ssRNA viral infection
PT2735568T (en) * 2006-05-10 2017-11-14 Avi Biopharma Inc Oligonucleotide analogs having cationic intersubunit linkages
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
US20070265215A1 (en) * 2006-05-11 2007-11-15 Iversen Patrick L Antisense restenosis composition and method
WO2007134181A2 (en) 2006-05-11 2007-11-22 Isis Pharmaceuticals, Inc. 5'-modified bicyclic nucleic acid analogs
NZ572666A (en) 2006-05-11 2010-11-26 Alnylam Pharmaceuticals Inc Compositions comprising double stranded rna and methods for inhibiting expression of the pcsk9 gene
WO2007137156A2 (en) 2006-05-19 2007-11-29 Alnylam Pharmaceuticals, Inc. Rnai modulation of aha and therapeutic uses thereof
US7888498B2 (en) 2006-05-22 2011-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
WO2008011473A2 (en) 2006-07-19 2008-01-24 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hbxip
US20100137440A1 (en) * 2006-09-11 2010-06-03 Yale University Lysine riboswitches, structure-based compound design with lysine riboswitches, and methods and compositions for use of and with lysine riboswitches
WO2008036933A2 (en) 2006-09-21 2008-03-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the hamp gene
MX363224B (en) 2006-10-03 2019-03-15 Alnylam Pharmaceuticals Inc Lipid containing formulations.
WO2008136852A2 (en) 2006-11-01 2008-11-13 University Of Rochester Methods and compositions related to the structure and function of apobec3g
CA2672297A1 (en) 2006-12-11 2008-06-19 University Of Utah Research Foundation Compositions and methods for treating pathologic angiogenesis and vascular permeability
CA2675967A1 (en) * 2007-01-16 2008-07-24 Yissum Research Development Company Of The Hebrew University Of Jerusale M Nucleic acid constructs and methods for specific silencing of h19
US20100196403A1 (en) * 2007-01-29 2010-08-05 Jacob Hochman Antibody conjugates for circumventing multi-drug resistance
CN103966345A (en) 2007-02-09 2014-08-06 西北大学 Particles for detecting intracellular targets
CN101801185A (en) 2007-03-22 2010-08-11 耶鲁大学 Methods and compositions related to riboswitches that control alternative splicing
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
EP2905336A1 (en) 2007-03-29 2015-08-12 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of a gene from the ebola
CA2690281A1 (en) 2007-05-11 2008-11-20 The Johns Hopkins University Biomarkers for melanoma
EP2426219A1 (en) 2007-05-29 2012-03-07 Yale University Riboswitches and methods and compositions for use of and with riboswitches
EP2164994A4 (en) * 2007-05-29 2010-07-21 Univ Yale Inc Methods and compositions related to riboswitches that control alternative splicing and rna processing
US8278425B2 (en) 2007-05-30 2012-10-02 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
CA2689923A1 (en) 2007-05-30 2008-12-11 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
US7807372B2 (en) * 2007-06-04 2010-10-05 Northwestern University Screening sequence selectivity of oligonucleotide-binding molecules using nanoparticle based colorimetric assay
AU2008262391A1 (en) * 2007-06-06 2008-12-18 Avi Biopharma, Inc. Soluble HER2 and HER3 splice variant proteins, splice-switching oligonucleotides, and their use in the treatment of disease
US8278426B2 (en) 2007-06-08 2012-10-02 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
US20090099066A1 (en) 2007-06-29 2009-04-16 Avi Biopharma, Inc. Tissue specific peptide conjugates and methods
CN101688206B (en) 2007-07-05 2013-05-15 诺瓦提斯公司 DsRNA for treating viral infection
US8278283B2 (en) 2007-07-05 2012-10-02 Isis Pharmaceuticals, Inc. 6-disubstituted or unsaturated bicyclic nucleic acid analogs
US8088904B2 (en) 2007-08-15 2012-01-03 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
US8557767B2 (en) 2007-08-28 2013-10-15 Uab Research Foundation Synthetic apolipoprotein E mimicking polypeptides and methods of use
AU2008296478B9 (en) 2007-08-28 2015-03-19 The Uab Research Foundation Synthetic apolipoprotein E mimicking polypeptides and methods of use
US8415455B2 (en) 2007-09-04 2013-04-09 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2009039466A1 (en) 2007-09-20 2009-03-26 Vanderbilt University Free solution measurement of molecular interactions by backscattering interferometry
WO2009039442A1 (en) 2007-09-21 2009-03-26 California Institute Of Technology Nfia in glial fate determination, glioma therapy and astrocytoma treatment
CA2700953A1 (en) 2007-10-02 2009-04-09 Amgen Inc. Increasing erythropoietin using nucleic acids hybridizable to micro-rna and precursors thereof
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
US8916531B2 (en) 2007-11-20 2014-12-23 Isis Pharmaceuticals, Inc. Modulation of CD40 expression
WO2009067647A1 (en) * 2007-11-21 2009-05-28 Isis Pharmaceuticals, Inc. Carbocyclic alpha-l-bicyclic nucleic acid analogs
EP2617828B1 (en) 2007-12-10 2014-09-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
US7989608B2 (en) * 2007-12-28 2011-08-02 Avi Biopharma Inc. Immunomodulatory agents and methods of use
US20110117125A1 (en) 2008-01-02 2011-05-19 Tekmira Pharmaceuticals Corporation Compositions and methods for the delivery of nucleic acids
WO2009100320A2 (en) 2008-02-07 2009-08-13 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
AU2009221775B2 (en) 2008-03-05 2015-05-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 and VEGF genes
EP2282744B1 (en) 2008-03-21 2018-01-17 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucleosides and methods for their use
WO2009124238A1 (en) 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
ES2554168T3 (en) 2008-04-18 2015-12-16 Baxter International Inc. Microsphere-based composition to prevent and / or reverse newly occurring autoimmune diabetes
AU2009275387B2 (en) 2008-08-25 2010-07-08 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
EP3208337A1 (en) 2008-09-02 2017-08-23 Alnylam Pharmaceuticals, Inc. Compositions for combined inhibition of mutant egfr and il-6 expression
EP2356129B1 (en) 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Substituted alpha-l-bicyclic nucleosides
DK2361256T3 (en) 2008-09-24 2013-07-01 Isis Pharmaceuticals Inc Cyclohexenyl-nucleic acid analogues
EP3584320A1 (en) 2008-09-25 2019-12-25 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
CN104119242B (en) 2008-10-09 2017-07-07 泰米拉制药公司 The amino lipids of improvement and the method for delivering nucleic acid
US8334372B2 (en) 2008-10-15 2012-12-18 Isis Pharmaceuticals, Inc. Modulation of factor 11 expression
CN106834291B (en) 2008-10-20 2020-09-29 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of transthyretin
EP2358397B1 (en) 2008-10-24 2020-01-01 Ionis Pharmaceuticals, Inc. 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
EP2447274B1 (en) 2008-10-24 2017-10-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds and methods
KR20230025924A (en) 2008-10-24 2023-02-23 사렙타 쎄러퓨틱스 인코퍼레이티드 Multiple exon skipping compositions for dmd
KR101692880B1 (en) 2008-11-24 2017-01-04 노오쓰웨스턴 유니버시티 Polyvalent rna-nanoparticle compositions
WO2010061393A1 (en) 2008-11-30 2010-06-03 Compugen Ltd. He4 variant nucleotide and amino acid sequences, and methods of use thereof
EP2370582B1 (en) 2008-12-04 2017-05-10 CuRNA, Inc. Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
EP2370580B1 (en) 2008-12-04 2019-09-11 CuRNA, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
ES2629630T3 (en) 2008-12-04 2017-08-11 Curna, Inc. Treatment of diseases related to erythropoietin (EPO) by inhibiting the natural antisense transcript to EPO
WO2010064248A2 (en) 2008-12-05 2010-06-10 Yeda Research And Development Co. Ltd. Methods of diagnosing and treating motor neuron diseases
AU2009324534B2 (en) 2008-12-10 2015-07-30 Alnylam Pharmaceuticals, Inc. GNAQ targeted dsRNA compositions and methods for inhibiting expression
US8592386B2 (en) * 2008-12-17 2013-11-26 Sarepta Therapeutics, Inc. Antisense compositions and methods for modulating contact hypersensitivity or contact dermatitis
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
US20100294952A1 (en) * 2009-01-15 2010-11-25 Northwestern University Controlled agent release and sequestration
US20120101148A1 (en) 2009-01-29 2012-04-26 Alnylam Pharmaceuticals, Inc. lipid formulation
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8536320B2 (en) 2009-02-06 2013-09-17 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
WO2010093906A2 (en) 2009-02-12 2010-08-19 Curna, Inc. Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
ES2560107T3 (en) 2009-02-12 2016-02-17 Curna, Inc. Treatment of diseases related to brain-derived neurotrophic factor (BDNF) by inhibition of natural antisense transcript for BDNF
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
WO2010102058A2 (en) 2009-03-04 2010-09-10 Curna, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt 1
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
CA2755409C (en) 2009-03-16 2019-04-30 Joseph Collard Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases by inhibition of natural antisense transcript to nrf2
JP5904935B2 (en) 2009-03-17 2016-04-20 クルナ・インコーポレーテッド Treatment of DLK1-related diseases by suppression of natural antisense transcripts against Delta-like 1 homolog (DLK1)
AU2010237001B2 (en) 2009-04-15 2016-07-07 Northwestern University Delivery of oligonucleotide-functionalized nanoparticles
EP3248618A1 (en) 2009-04-22 2017-11-29 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
CA2760589C (en) 2009-05-01 2019-08-20 Joseph Collard Treatment of hemoglobin (hbf/hbg) related diseases by inhibition of natural antisense transcript to hbf/hbg
CA3045126A1 (en) 2009-05-05 2010-11-11 Arbutus Biopharma Corporation Methods of delivering oligonucleotides to immune cells
KR20180094137A (en) 2009-05-05 2018-08-22 알닐람 파마슈티칼스 인코포레이티드 Lipid compositions
KR101722541B1 (en) 2009-05-06 2017-04-04 큐알엔에이, 인크. Treatment of tristetraproline(ttp) related diseases by inhibition of natural antisense transcript to ttp
CN102459596B (en) 2009-05-06 2016-09-07 库尔纳公司 By suppression therapy lipid transfer and the metabolic gene relevant disease of the natural antisense transcript for lipid transfer and metabolic gene
WO2010132665A1 (en) 2009-05-15 2010-11-18 Yale University Gemm riboswitches, structure-based compound design with gemm riboswitches, and methods and compositions for use of and with gemm riboswitches
ES2664590T3 (en) 2009-05-18 2018-04-20 Curna, Inc. Treatment of diseases related to reprogramming factors by inhibition of the natural antisense transcript to a reprogramming factor
US8895527B2 (en) 2009-05-22 2014-11-25 Curna, Inc. Treatment of transcription factor E3 (TFE3) and insulin receptor substrate 2(IRS2) related diseases by inhibition of natural antisense transcript to TFE3
WO2010138806A2 (en) 2009-05-28 2010-12-02 Curna, Inc. Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
US8158601B2 (en) 2009-06-10 2012-04-17 Alnylam Pharmaceuticals, Inc. Lipid formulation
US8951981B2 (en) 2009-06-16 2015-02-10 Curna, Inc. Treatment of paraoxonase 1 (PON1) related diseases by inhibition of natural antisense transcript to PON1
JP5944311B2 (en) 2009-06-16 2016-07-05 クルナ・インコーポレーテッド Treatment of collagen gene-related diseases by suppression of natural antisense transcripts against collagen genes
JP6073133B2 (en) 2009-06-24 2017-02-01 クルナ・インコーポレーテッド Treatment of TNFR2-related diseases by suppression of natural antisense transcripts against tumor necrosis factor receptor 2 (TNFR2)
EP2446037B1 (en) 2009-06-26 2016-04-20 CuRNA, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
US8927513B2 (en) 2009-07-07 2015-01-06 Alnylam Pharmaceuticals, Inc. 5′ phosphate mimics
US9512164B2 (en) 2009-07-07 2016-12-06 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
CN102762731B (en) 2009-08-05 2018-06-22 库尔纳公司 By inhibiting to treat insulin gene (INS) relevant disease for the natural antisense transcript of insulin gene (INS)
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
WO2011020023A2 (en) 2009-08-14 2011-02-17 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
WO2011022420A1 (en) 2009-08-17 2011-02-24 Yale University Methylation biomarkers and methods of use
WO2011031482A2 (en) 2009-08-25 2011-03-17 Curna, Inc. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
ES2599076T3 (en) 2009-09-02 2017-01-31 Genentech, Inc. Smoothened mutant and methods of use thereof
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
WO2011045796A1 (en) 2009-10-14 2011-04-21 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Compositions for controlling varroa mites in bees
KR20120105446A (en) 2009-10-22 2012-09-25 제넨테크, 인크. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
JP6147502B2 (en) 2009-10-27 2017-06-14 スウィフト バイオサイエンシーズ, インコーポレイテッド Polynucleotide primers and probes
KR20120136345A (en) 2009-10-30 2012-12-18 노오쓰웨스턴 유니버시티 Templated nanoconjugates
WO2011056215A1 (en) 2009-11-03 2011-05-12 Landers James P Versatile, visible method for detecting polymeric analytes
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
EP2499249B1 (en) 2009-11-12 2018-08-08 The University Of Western Australia Antisense molecules and methods for treating pathologies
US8697858B2 (en) 2009-11-13 2014-04-15 Sarepta Therapeutics, Inc. Antisense antiviral compound and method for treating influenza viral infection
JP2013511285A (en) 2009-11-23 2013-04-04 スイフト・バイオサイエンシズ・インコーポレイテツド Device for extending single-stranded target molecules
AU2010324686B2 (en) 2009-11-30 2016-05-19 Genentech, Inc. Antibodies for treating and diagnosing tumors expressing SLC34A2 (TAT211 = SEQID2 )
ES2661813T3 (en) 2009-12-16 2018-04-04 Curna, Inc. Treatment of diseases related to membrane transcription factor peptidase, site 1 (mbtps1) by inhibition of the natural antisense transcript to the mbtps1 gene
CA2782375C (en) 2009-12-23 2023-10-31 Opko Curna, Llc Treatment of uncoupling protein 2 (ucp2) related diseases by inhibition of natural antisense transcript to ucp2
JP5934106B2 (en) 2009-12-23 2016-06-15 カッパーアールエヌエー,インコーポレイテッド Treatment of HGF-related diseases by inhibition of natural antisense transcripts against hepatocyte growth factor (HGF)
EP2519634B1 (en) 2009-12-29 2016-06-01 CuRNA, Inc. TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
US8921334B2 (en) 2009-12-29 2014-12-30 Curna, Inc. Treatment of nuclear respiratory factor 1 (NRF1) related diseases by inhibition of natural antisense transcript to NRF1
KR101878501B1 (en) 2010-01-04 2018-08-07 큐알엔에이, 인크. Treatment of interferon regulatory factor 8 (irf8) related diseases by inhibition of natural antisense transcript to irf8
KR101853509B1 (en) 2010-01-06 2018-04-30 큐알엔에이, 인크. Treatment of Pancreatic Developmental Gene Related Diseases By Inhibition of Natural Antisense Transcript to A Pancreatic Developmental Gene
ES2664866T3 (en) 2010-01-11 2018-04-23 Curna, Inc. Treatment of diseases related to sex hormone binding globulin (shbg) by inhibition of the natural antisense transcript to shbg
WO2011085102A1 (en) 2010-01-11 2011-07-14 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
SG182365A1 (en) 2010-01-12 2012-08-30 Univ Yale Structured rna motifs and compounds and methods for their use
CN102782135A (en) 2010-01-25 2012-11-14 库尔纳公司 Treatment of RNase H1 related diseases by inhibition of natural antisense transcript to RNase H1
US20130028889A1 (en) 2010-02-04 2013-01-31 Ico Therapeutics Inc. Dosing regimens for treating and preventing ocular disorders using c-raf antisense
US8962586B2 (en) 2010-02-22 2015-02-24 Curna, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcript to PYCR1
WO2011105900A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-alpha (c8-alpha) and uses thereof
US8877897B2 (en) 2010-02-23 2014-11-04 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2011105902A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-beta (c8-beta) and uses thereof
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
WO2011112516A1 (en) 2010-03-08 2011-09-15 Ico Therapeutics Inc. Treating and preventing hepatitis c virus infection using c-raf kinase antisense oligonucleotides
KR101982126B1 (en) 2010-03-08 2019-05-27 몬산토 테크놀로지 엘엘씨 Polynucleotide Molecules for Gene Regulation in Plants
WO2011113015A2 (en) 2010-03-12 2011-09-15 Avi Biopharma, Inc. Antisense modulation of nuclear hormone receptors
US20130101512A1 (en) 2010-03-12 2013-04-25 Chad A. Mirkin Crosslinked polynucleotide structure
ES2743600T3 (en) 2010-03-12 2020-02-20 Brigham & Womens Hospital Inc Methods of treatment of vascular inflammatory disorders
WO2011115818A1 (en) 2010-03-17 2011-09-22 Isis Pharmaceuticals, Inc. 5'-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
US8889350B2 (en) 2010-03-26 2014-11-18 Swift Biosciences, Inc. Methods and compositions for isolating polynucleotides
JP5860029B2 (en) 2010-03-29 2016-02-16 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. SiRNA therapy for transthyretin (TTR) related ocular amyloidosis
WO2011123621A2 (en) 2010-04-01 2011-10-06 Alnylam Pharmaceuticals Inc. 2' and 5' modified monomers and oligonucleotides
CN102858979B (en) 2010-04-09 2018-01-26 库尔纳公司 FGF21 relevant diseases are treated by suppressing the natural antisense transcript of FGF2 1 (FGF21)
WO2011133695A2 (en) 2010-04-20 2011-10-27 Swift Biosciences, Inc. Materials and methods for nucleic acid fractionation by solid phase entrapment and enzyme-mediated detachment
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
JP6066900B2 (en) 2010-04-26 2017-01-25 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of cysteinyl tRNA synthetase
AU2011248614B2 (en) 2010-04-27 2017-02-16 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
CN103154014B (en) 2010-04-28 2015-03-25 Isis制药公司 Modified nucleosides, modified nucleosides-like and oligomeric compounds prepared therefrom
CA2797271C (en) 2010-04-28 2021-05-25 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of alanyl trna synthetases
US9127033B2 (en) 2010-04-28 2015-09-08 Isis Pharmaceuticals, Inc. 5′ modified nucleosides and oligomeric compounds prepared therefrom
EP2563383B1 (en) 2010-04-29 2017-03-01 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl trna synthetases
SI2563920T1 (en) 2010-04-29 2017-05-31 Ionis Pharmaceuticals, Inc. Modulation of transthyretin expression
WO2011139854A2 (en) 2010-04-29 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl trna synthetases
CN103140233B (en) 2010-05-03 2017-04-05 Atyr 医药公司 Treatment, diagnosis and the discovery of antibody compositions related to the protein fragments of methionyl-tRNA synthetase
WO2011139387A1 (en) 2010-05-03 2011-11-10 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
BR112012028010A2 (en) 2010-05-03 2017-09-26 Genentech Inc isolated antibody, cell, isolated nucleic acid, method of identifying a first antibody that binds to a tat425 antigenic epitope attached to an antibody, methods of inhibiting cell growth, therapeutic treatment of determining the presence of a tat425 protein and diagnosing the presence of a tumor in a mammal
CA2797277C (en) 2010-05-03 2021-02-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases
US9034321B2 (en) 2010-05-03 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
CN102985103A (en) 2010-05-04 2013-03-20 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
CA3090304A1 (en) 2010-05-13 2011-11-17 Sarepta Therapeutics, Inc. Antisense modulation of interleukins 17 and 23 signaling
US9050373B2 (en) 2010-05-13 2015-06-09 The Charlotte-Mecklenburg Hospital Authority Pharmaceutical compositions comprising antisense oligonucleotides and methods of using same
JP6396656B2 (en) 2010-05-14 2018-09-26 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of phenylalanyl βtRNA synthetase
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
US9034598B2 (en) 2010-05-17 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
WO2011150226A1 (en) 2010-05-26 2011-12-01 Landers James P Method for detecting nucleic acids based on aggregate formation
NO2576783T3 (en) 2010-05-26 2018-04-28
CN103096913B (en) 2010-05-27 2017-07-18 Atyr 医药公司 Treatment, diagnosis and the innovation of antibody compositions related to the protein fragments of glutaminyl tRNA synzyme is found
KR101981705B1 (en) 2010-05-28 2019-05-24 사렙타 쎄러퓨틱스, 인코퍼레이티드 Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
CA2801066C (en) 2010-06-02 2021-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
US8957200B2 (en) 2010-06-07 2015-02-17 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
EP2580228B1 (en) 2010-06-08 2016-03-23 Ionis Pharmaceuticals, Inc. Substituted 2'-amino and 2'-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
US9638632B2 (en) 2010-06-11 2017-05-02 Vanderbilt University Multiplexed interferometric detection system and method
WO2011163466A1 (en) 2010-06-23 2011-12-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (nrg-1)
US8678238B2 (en) 2010-06-30 2014-03-25 Actamax Surgical Materials, Llc Self-contained hand-held yoke-connected device for dispensing a two-part adhesive aerosol
US9211554B2 (en) 2010-06-30 2015-12-15 Actamax Surgical Materials, Llc Self-contained hand-held direct drive device for dispensing a two-part adhesive aerosol
AU2011272941B2 (en) 2010-06-30 2014-05-29 Compugen Ltd. C10RF32 for the treatment of multiple sclerosis, rheumatoid arthritis and other autoimmune disorders
JP6116479B2 (en) 2010-07-12 2017-04-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of glycyl-tRNA synthetase
US8980860B2 (en) 2010-07-14 2015-03-17 Curna, Inc. Treatment of discs large homolog (DLG) related diseases by inhibition of natural antisense transcript to DLG
WO2012021554A1 (en) 2010-08-09 2012-02-16 Yale University Cyclic di-gmp-ii riboswitches, motifs, and compounds, and methods for their use
US8198429B2 (en) 2010-08-09 2012-06-12 Avi Biopharma, Inc. Antisense antiviral compounds and methods for treating a filovirus infection
WO2012027611A2 (en) 2010-08-25 2012-03-01 Atyr Pharma, Inc. INNOVATIVE DISCOVERY OF THERAPEUTIC, DIAGNOSTIC, AND ANTIBODY COMPOSITIONS RELATED TO PROTEIN FRAGMENTS OF TYROSYL-tRNA SYNTHETASES
WO2012031243A2 (en) 2010-09-03 2012-03-08 Avi Biopharma, Inc. dsRNA MOLECULES COMPRISING OLIGONUCLEOTIDE ANALOGS HAVING MODIFIED INTERSUBUNIT LINKAGES AND/OR TERMINAL GROUPS
WO2012038956A1 (en) 2010-09-20 2012-03-29 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method of treating neurodegenerative diseases
DK2625197T3 (en) 2010-10-05 2016-10-03 Genentech Inc Smoothened MUTANT AND METHODS OF USING THE SAME
US8993533B2 (en) 2010-10-06 2015-03-31 Curna, Inc. Treatment of sialidase 4 (NEU4) related diseases by inhibition of natural antisense transcript to NEU4
EP2625292B1 (en) 2010-10-07 2018-12-05 The General Hospital Corporation Biomarkers of cancer
EP3075396A1 (en) 2010-10-17 2016-10-05 Yeda Research and Development Co. Ltd. Methods and compositions for the treatment of insulin-associated medical conditions
US8648053B2 (en) 2010-10-20 2014-02-11 Rosalind Franklin University Of Medicine And Science Antisense oligonucleotides that target a cryptic splice site in Ush1c as a therapeutic for Usher syndrome
CA2815212A1 (en) 2010-10-22 2012-04-26 Curna, Inc. Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
DK2633052T3 (en) 2010-10-27 2018-07-16 Curna Inc TREATMENT OF INTERFERON-RELATED DEVELOPMENT REGULATOR 1 (IFRD1) -RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPT TO IFRD1
WO2012064824A1 (en) 2010-11-09 2012-05-18 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of eg5 and vegf genes
US20120122769A1 (en) 2010-11-12 2012-05-17 Avi Biopharma, Inc. Antisense antibacterial compounds and methods
WO2012065143A1 (en) 2010-11-12 2012-05-18 The General Hospital Corporation Polycomb-associated non-coding rnas
CA3077910A1 (en) 2010-11-17 2012-05-24 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
KR102010598B1 (en) 2010-11-23 2019-08-13 큐알엔에이, 인크. Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
WO2012097261A2 (en) 2011-01-14 2012-07-19 The General Hospital Corporation Methods targeting mir-128 for regulating cholesterol/lipid metabolism
WO2012106509A1 (en) 2011-02-02 2012-08-09 The Trustees Of Princeton University Sirtuin modulators as virus production modulators
WO2012106508A1 (en) 2011-02-02 2012-08-09 Pfizer Inc. Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
CN103501793A (en) 2011-02-08 2014-01-08 夏洛特-梅克伦堡医院(商业用名:卡罗来纳保健系统) Antisense oligonucleotides
US9562853B2 (en) 2011-02-22 2017-02-07 Vanderbilt University Nonaqueous backscattering interferometric methods
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
EP3460064B8 (en) 2011-04-03 2024-03-20 The General Hospital Corporation d/b/a Massachusetts General Hospital Efficient protein expression in vivo using modified rna (mod-rna)
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
WO2012149154A1 (en) 2011-04-26 2012-11-01 Swift Biosciences, Inc. Polynucleotide primers and probes
WO2012151324A1 (en) 2011-05-02 2012-11-08 Isis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with usher syndrome
WO2012151268A1 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system for high throughput optical and label free detection of analytes
WO2012151289A2 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system to detect aggregate formation on a substrate
CN103619356B (en) 2011-05-05 2017-09-12 萨勒普塔医疗公司 Peptide oligonucleotide conjugates
JP6188686B2 (en) 2011-06-09 2017-08-30 カッパーアールエヌエー,インコーポレイテッド Treatment of FXN-related diseases by inhibition of natural antisense transcripts to frataxin (FXN)
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2012170945A2 (en) 2011-06-10 2012-12-13 Isis Pharmaceuticals, Inc. Methods for modulating kallikrein (klkb1) expression
SG10201800715PA (en) 2011-06-21 2018-02-27 Alnylam Pharmaceuticals Inc Angiopoietin-like 3 (angptl3) irna compostions and methods of use thereof
EP2723351B1 (en) 2011-06-21 2018-02-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
MX344807B (en) 2011-06-21 2017-01-09 Alnylam Pharmaceuticals Inc Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes.
WO2012178033A2 (en) 2011-06-23 2012-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
CA2840614A1 (en) 2011-06-29 2013-01-03 Isis Pharmaceuticals, Inc. Methods for modulating kallikrein (klkb1) expression
EP2726503B1 (en) 2011-06-30 2019-09-04 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
WO2013012752A2 (en) 2011-07-15 2013-01-24 Sarepta Therapeutics, Inc. Methods and compositions for manipulating translation of protein isoforms from alternative initiation start sites
EP2739735A2 (en) 2011-08-01 2014-06-11 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
WO2013018060A2 (en) 2011-08-04 2013-02-07 Yeda Research And Development Co. Ltd. Micro-rnas and compositions comprising same for the treatment and diagnosis of serotonin-, adrenalin-, noradrenalin-, glutamate-, and corticotropin-releasing hormone- associated medical conditions
MX343071B (en) 2011-09-13 2016-10-21 Monsanto Technology Llc Methods and compositions for weed control.
ES2645927T3 (en) 2011-09-13 2017-12-11 Monsanto Technology Llc Procedures and compositions for weed control
US9840715B1 (en) 2011-09-13 2017-12-12 Monsanto Technology Llc Methods and compositions for delaying senescence and improving disease tolerance and yield in plants
BR112014005975A8 (en) 2011-09-13 2017-09-12 Monsanto Technology Llc PLANT CONTROL METHOD, METHOD OF REDUCING EXPRESSION OF A PDS GENE IN A PLANT, MICROBIAL EXPRESSION CASSETTE, METHOD OF MAKING A POLYNUCLEOTIDE, METHOD OF IDENTIFICATION OF POLYNUCLEOTIDES, AND COMPOSITIONS FOR WEED CONTROL
MX343072B (en) 2011-09-13 2016-10-21 Monsanto Technology Llc Methods and compositions for weed control.
US10829828B2 (en) 2011-09-13 2020-11-10 Monsanto Technology Llc Methods and compositions for weed control
US10806146B2 (en) 2011-09-13 2020-10-20 Monsanto Technology Llc Methods and compositions for weed control
US10760086B2 (en) 2011-09-13 2020-09-01 Monsanto Technology Llc Methods and compositions for weed control
US9920326B1 (en) 2011-09-14 2018-03-20 Monsanto Technology Llc Methods and compositions for increasing invertase activity in plants
AU2012308302A1 (en) 2011-09-14 2014-03-20 Northwestern University Nanoconjugates able to cross the blood-brain barrier
JP6129844B2 (en) 2011-09-14 2017-05-17 ラナ セラピューティクス インコーポレイテッド Multimeric oligonucleotide compounds
WO2013040548A2 (en) 2011-09-17 2013-03-21 Yale University Fluoride-responsive riboswitchs, fluoride transporters, and methods of use
US9801948B2 (en) 2011-09-21 2017-10-31 Yale University Antimicrobial compositions and methods of use thereof
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
EP2766482B1 (en) 2011-10-11 2016-12-07 The Brigham and Women's Hospital, Inc. Micrornas in neurodegenerative disorders
WO2013061328A2 (en) 2011-10-27 2013-05-02 Yeda Research And Development Co. Ltd. Method of treating cancer
CN104039739A (en) 2011-11-18 2014-09-10 萨勒普塔医疗公司 Functionally-modified oligonucleotides and subunits thereof
JP6317675B2 (en) 2011-11-30 2018-04-25 サレプタ セラピューティクス, インコーポレイテッド Oligonucleotides for treating prolonged repeat disease
CA2857664A1 (en) 2011-11-30 2013-06-06 Sarepta Therapeutics, Inc. Antisense oligonucleotides targeting within the smn2 pre-mrna for use ininduced exon inclusion in spinal muscle atrophy
US10465042B2 (en) 2011-12-02 2019-11-05 Yale University Poly(amine-co-ester) nanoparticles and methods of use thereof
WO2013082529A1 (en) 2011-12-02 2013-06-06 Yale University Enzymatic synthesis of poly(amine-co-esters) and methods of use thereof for gene delivery
US9895451B2 (en) 2011-12-02 2018-02-20 Yale University Formulations for targeted release of agents to low pH tissue environments or cellular compartments and methods of use thereof
EP2788487B1 (en) 2011-12-08 2018-04-04 Sarepta Therapeutics, Inc. Oligonucleotide analogues targeting human lmna
DK2790736T3 (en) 2011-12-12 2018-05-07 Oncoimmunin Inc In vivo delivery of oligonucleotides
WO2013096837A1 (en) 2011-12-22 2013-06-27 Isis Pharmaceuticals, Inc. Methods for modulating metastasis-associated-in-lung-adenocarcinoma-transcript-1(malat-1) expression
US20150025231A1 (en) 2012-01-11 2015-01-22 Cold Spring Harbor Laboratory Compositions and methods for modulation of ikbkap splicing
WO2013114367A2 (en) 2012-02-01 2013-08-08 Compugen Ltd. C10rf32 antibodies, and uses thereof for treatment of cancer
WO2013120003A1 (en) 2012-02-08 2013-08-15 Isis Pharmaceuticals, Inc. Modulation of rna by repeat targeting
SG11201404608WA (en) 2012-02-13 2014-09-26 Gamida Cell Ltd Mesenchymal stem cells conditioned medium and methods of generating and using the same
EP2844745A2 (en) 2012-02-22 2015-03-11 Brainstem Biotec Ltd. Generation of neural stem cells and motor neurons
JP6329911B2 (en) 2012-02-22 2018-05-23 ブレインステム バイオテック リミテッド MicroRNA for the production of astrocytes
CN104395480B (en) 2012-03-13 2018-01-30 斯威夫特生物科学公司 For the method and composition of size-controlled homopolymeric tailing to be carried out to substrate polynucleotide by nucleic acid polymerase
EP2639238A1 (en) 2012-03-15 2013-09-18 Universität Bern Tricyclic nucleosides and oligomeric compounds prepared therefrom
CN110438125A (en) 2012-03-15 2019-11-12 科纳公司 By inhibiting the natural antisense transcript of brain derived neurotrophic factor (BDNF) to treat BDNF related disease
WO2013142514A1 (en) 2012-03-19 2013-09-26 Isis Pharmaceuticals, Inc. Methods and compositions for modulating alpha-1-antitrypsin expression
CN104411831B (en) 2012-03-20 2020-08-11 萨勒普塔医疗公司 Boronic acid conjugates of oligonucleotide analogs
EP2831231A1 (en) 2012-03-30 2015-02-04 Isis Pharmaceuticals, Inc. Compositions and methods for modulating tau expression for reducing seizure and modifying a neurodegenerative syndrome
WO2013151771A1 (en) 2012-04-05 2013-10-10 Massachusetts Institute Of Technology Immunostimulatory compositions and methods of use thereof
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
EP2850092B1 (en) 2012-04-09 2017-03-01 Ionis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
AU2013248981B2 (en) 2012-04-20 2018-11-29 Aptamir Therapeutics, Inc. Mirna modulators of thermogenesis
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
US9273949B2 (en) 2012-05-11 2016-03-01 Vanderbilt University Backscattering interferometric methods
KR102028784B1 (en) 2012-05-16 2019-10-04 트랜슬레이트 바이오 인코포레이티드 Compositions and methods for modulating gene expression
DK2850186T3 (en) 2012-05-16 2019-04-08 Translate Bio Ma Inc COMPOSITIONS AND PROCEDURES FOR MODULATING SMN GENFAMILY EXPRESSION
WO2013173789A2 (en) 2012-05-17 2013-11-21 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
WO2013177248A2 (en) 2012-05-22 2013-11-28 Isis Pharmaceuticals, Inc. Modulation of enhancer rna mediated gene expression
US10240161B2 (en) 2012-05-24 2019-03-26 A.B. Seeds Ltd. Compositions and methods for silencing gene expression
WO2013181665A1 (en) 2012-06-01 2013-12-05 Isis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with fibronectin
US9487780B2 (en) 2012-06-01 2016-11-08 Ionis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with fibronectin
WO2013184209A1 (en) 2012-06-04 2013-12-12 Ludwig Institute For Cancer Research Ltd. Mif for use in methods of treating subjects with a neurodegenerative disorder
LT3461895T (en) 2012-06-25 2020-09-10 Ionis Pharmaceuticals, Inc. Modulation of ube3a-ats expression
US20140038182A1 (en) 2012-07-17 2014-02-06 Dna Logix, Inc. Cooperative primers, probes, and applications thereof
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
US20150297629A1 (en) 2012-07-27 2015-10-22 Isis Pharmaceuticals, Inc. Modulation of renin-angiotensin system (ras) related diseases by angiotensinogen
CA2880833A1 (en) 2012-08-03 2014-02-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
EP2885312A4 (en) 2012-08-15 2016-01-20 Isis Pharmaceuticals Inc Method of preparing oligomeric compounds using modified capping protocols
AU2013306006B2 (en) 2012-08-20 2017-07-06 The Regents Of The University Of California Polynucleotides having bioreversible groups
AR092658A1 (en) 2012-09-25 2015-04-29 Genzyme Corp OLIGONUCLEOTIDOS ANTISENTIDO MORFOLINO UNITED TO PEPTIDE FOR THE TREATMENT OF MYOTONIC DYSTROPHY
WO2014059364A1 (en) 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Methods of treating kennedy's disease
US9175291B2 (en) 2012-10-11 2015-11-03 Isis Pharmaceuticals Inc. Modulation of androgen receptor expression
WO2014059356A2 (en) 2012-10-12 2014-04-17 Isis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
EP4144845B1 (en) 2012-10-12 2024-04-24 Ionis Pharmaceuticals, Inc. Antisense compounds and uses thereof
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
CN104870647A (en) 2012-10-18 2015-08-26 孟山都技术公司 Methods and compositions for plant pest control
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
MX2015008035A (en) 2012-12-20 2016-03-17 Sarepta Therapeutics Inc Improved exon skipping compositions for treating muscular dystrophy.
US10041068B2 (en) 2013-01-01 2018-08-07 A. B. Seeds Ltd. Isolated dsRNA molecules and methods of using same for silencing target molecules of interest
US10683505B2 (en) 2013-01-01 2020-06-16 Monsanto Technology Llc Methods of introducing dsRNA to plant seeds for modulating gene expression
US9856474B2 (en) 2013-01-16 2018-01-02 Iowa State University Research Foundation, Inc. Deep intronic target for splicing correction on spinal muscular atrophy gene
US10980804B2 (en) 2013-01-18 2021-04-20 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
US10000767B2 (en) 2013-01-28 2018-06-19 Monsanto Technology Llc Methods and compositions for plant pest control
KR102190852B1 (en) 2013-01-31 2020-12-14 아이오니스 파마수티컬즈, 인코포레이티드 Method of preparing oligomeric compounds using modified coupling protocols
KR102169899B1 (en) 2013-02-14 2020-10-26 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of apolipoprotein c-iii (apociii) expression in lipoprotein lipase deficient (lpld) populations
WO2014130922A1 (en) 2013-02-25 2014-08-28 Trustees Of Boston University Compositions and methods for treating fungal infections
US10251906B2 (en) 2013-03-12 2019-04-09 University Of Utah Research Foundation Compositions and methods for inducing apoptosis
EP2971185A4 (en) 2013-03-13 2017-03-08 Monsanto Technology LLC Methods and compositions for weed control
AU2014248958A1 (en) 2013-03-13 2015-10-01 Monsanto Technology Llc Methods and compositions for weed control
BR112015022156A2 (en) 2013-03-14 2017-11-14 Isis Pharmaceuticals Inc compositions and methods for modulating tau expression
WO2014160129A2 (en) 2013-03-14 2014-10-02 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
JP6449231B2 (en) 2013-03-14 2019-01-09 サレプタ セラピューティクス, インコーポレイテッド Exon skipping composition for treating muscular dystrophy
EP2970964B8 (en) 2013-03-14 2019-03-06 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
US20140283211A1 (en) 2013-03-14 2014-09-18 Monsanto Technology Llc Methods and Compositions for Plant Pest Control
EA201591792A1 (en) 2013-03-15 2016-02-29 Сарепта Терапьютикс, Инк. IMPROVED COMPOSITIONS FOR THE TREATMENT OF MUSCULAR DISTROPHIA
US10568328B2 (en) 2013-03-15 2020-02-25 Monsanto Technology Llc Methods and compositions for weed control
JP2016522679A (en) 2013-04-04 2016-08-04 プレジデント アンド フェローズ オブ ハーバード カレッジ Therapeutic use of genome editing with the CRISPR / Cas system
US9127276B2 (en) 2013-05-01 2015-09-08 Isis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
BR112015029139B1 (en) 2013-05-22 2022-07-12 Alnylam Pharmaceuticals, Inc DOUBLE-STRAND RNAI AGENT FOR INHIBITING SERPINA1 EXPRESSION IN A CELL, ITS USES, AS WELL AS PHARMACEUTICAL COMPOSITION AND IN VITRO METHOD OF INHIBITING SERPINA1 EXPRESSION IN A CELL
CN105452463B (en) 2013-05-22 2019-06-21 阿尔尼拉姆医药品有限公司 TMPRSS6 IRNA composition and its application method
US20160113911A1 (en) 2013-06-06 2016-04-28 The General Hospital Corporation Methods and compositions for the treatment of cancer
JP6869720B2 (en) 2013-06-13 2021-05-12 アンチセンス セラピューティクス リミテッド Combination therapy
EP3656386A1 (en) 2013-06-21 2020-05-27 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating apolipoprotein c-iii expression for improving a diabetic profile
WO2015002971A2 (en) 2013-07-02 2015-01-08 Isis Pharmaceuticals, Inc. Modulators of growth hormone receptor
US10077236B2 (en) 2013-07-15 2018-09-18 The Regents Of The University Of California Azacyclic constrained analogs of FTY720
CN105980567B (en) 2013-07-19 2021-04-16 孟山都技术有限公司 Compositions and methods for controlling phyllometaca
TWI657819B (en) 2013-07-19 2019-05-01 美商Ionis製藥公司 Compositions for modulating tau expression
US9850496B2 (en) 2013-07-19 2017-12-26 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
AU2014292926B2 (en) 2013-07-25 2020-03-05 Exicure Operating Company Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
EP4079745A1 (en) 2013-08-08 2022-10-26 The Scripps Research Institute A method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
TW201536329A (en) 2013-08-09 2015-10-01 Isis Pharmaceuticals Inc Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
CA2921839A1 (en) 2013-08-28 2015-03-05 Ionis Pharmaceuticals, Inc. Modulation of prekallikrein (pkk) expression
EP3041935A1 (en) 2013-09-05 2016-07-13 Sage Therapeutics, Inc. Antisense-induced exon2 inclusion in acid alpha-glucosidase
US10059947B2 (en) 2013-09-11 2018-08-28 Synthena Ag Nucleic acids and methods for the treatment of Pompe disease
EP3808338A1 (en) 2013-09-11 2021-04-21 Eagle Biologics, Inc. Liquid protein formulations containing ionic liquids
PL3043827T3 (en) 2013-09-13 2020-03-31 Ionis Pharmaceuticals, Inc. Modulators of complement factor b
US9708360B2 (en) 2013-09-30 2017-07-18 Geron Corporation Phosphorodiamidate backbone linkage for oligonucleotides
MX2016004230A (en) 2013-10-02 2016-10-21 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the lect2 gene.
SG11201602631XA (en) 2013-10-04 2016-05-30 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the alas1 gene
EP3055426B1 (en) 2013-10-09 2019-06-19 The United States of America as represented by The Secretary Department of Health and Human Services Detection of hepatitis delta virus (hdv) for the diagnosis and treatment of sjögren's syndrome and lymphoma
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
US9758546B2 (en) 2013-10-21 2017-09-12 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
EP3502270B1 (en) 2013-10-21 2020-03-18 The General Hospital Corporation Methods relating to circulating tumor cell clusters and the treatment of cancer
WO2015066708A1 (en) 2013-11-04 2015-05-07 Northwestern University Quantification and spatio-temporal tracking of a target using a spherical nucleic acid (sna)
MX2016005778A (en) 2013-11-04 2016-12-20 Monsanto Technology Llc Compositions and methods for controlling arthropod parasite and pest infestations.
EP3750997A1 (en) 2013-12-02 2020-12-16 Ionis Pharmaceuticals, Inc. Antisense compounds and uses thereof
CA2932122C (en) 2013-12-03 2022-04-19 Northwestern University Liposomal particles, methods of making same and uses thereof
WO2015085183A2 (en) 2013-12-06 2015-06-11 Swift Biosciences, Inc. Cleavable competitor polynucleotides
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
UA119253C2 (en) 2013-12-10 2019-05-27 Біолоджикс, Інк. Compositions and methods for virus control in varroa mite and bees
AU2014362262B2 (en) 2013-12-12 2021-05-13 Alnylam Pharmaceuticals, Inc. Complement component iRNA compositions and methods of use thereof
CN106456694B (en) 2013-12-20 2020-06-30 通用医疗公司 Methods and assays relating to circulating tumor cells
CN111394355A (en) 2013-12-24 2020-07-10 Ionis制药公司 Modulation of angiopoietin-like 3 expression
CN105979770B (en) 2014-01-15 2019-07-05 孟山都技术公司 For using the method and composition of the Weeds distribution of EPSPS polynucleotides
DK3102197T3 (en) 2014-02-04 2018-11-19 Genentech Inc Smoothened mutant and methods for its use
WO2015118537A2 (en) 2014-02-05 2015-08-13 Yeda Research And Development Co. Ltd. Micro-rnas and compositions comprising same for the treatment and diagnosis of serotonin-, adrenalin-, noradrenalin-, glutamate-, and corticotropin-releasing hormone- associated medical conditions
CN113057959A (en) 2014-02-11 2021-07-02 阿尔尼拉姆医药品有限公司 Ketohexokinase (KHK) iRNA compositions and methods of use thereof
WO2015142910A1 (en) 2014-03-17 2015-09-24 Isis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
US10006027B2 (en) 2014-03-19 2018-06-26 Ionis Pharmaceuticals, Inc. Methods for modulating Ataxin 2 expression
CN106103717A (en) 2014-03-19 2016-11-09 Ionis制药公司 For regulating the compositions that ataxin 2 is expressed
CN106459071A (en) 2014-03-31 2017-02-22 波士顿生物技术公司 New tricyclic quinone derivative
WO2015153339A2 (en) 2014-04-01 2015-10-08 Monsanto Technology Llc Compositions and methods for controlling insect pests
SG11201608109TA (en) 2014-04-01 2016-10-28 Ionis Pharmaceuticals Inc Compositions for modulating sod-1 expression
EP3129493B1 (en) 2014-04-09 2021-07-07 The Scripps Research Institute Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
WO2015164693A1 (en) 2014-04-24 2015-10-29 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
HUE052709T2 (en) 2014-05-01 2021-05-28 Ionis Pharmaceuticals Inc Conjugates of modified antisense oligonucleotides and their use for modulating pkk expression
KR102369736B1 (en) 2014-05-01 2022-03-02 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating complement factor b expression
SI3137605T1 (en) 2014-05-01 2021-02-26 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
WO2015168514A1 (en) 2014-05-01 2015-11-05 Isis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
KR102149571B1 (en) 2014-05-01 2020-08-31 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating growth hormone receptor expression
WO2015175510A1 (en) 2014-05-12 2015-11-19 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
EP3620178A3 (en) 2014-05-16 2020-07-22 Oregon State University Antisense antibacterial compounds and methods
CA2948568A1 (en) 2014-05-19 2015-11-26 David Greenberg Antisense antibacterial compounds and methods
US10570169B2 (en) 2014-05-22 2020-02-25 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
SG10202104570TA (en) 2014-05-22 2021-06-29 Alnylam Pharmaceuticals Inc Angiotensinogen (agt) irna compositions and methods of use thereof
US20170182189A1 (en) 2014-05-23 2017-06-29 Genzyme Corporation Inhibiting or downregulating glycogen synthase by creating premature stop codons using antisense oligonucleotides
CN106659758A (en) 2014-06-02 2017-05-10 儿童医疗中心有限公司 Methods and compositions for immunomodulation
AU2015272128B2 (en) 2014-06-10 2021-10-28 Erasmus University Medical Center Rotterdam Antisense oligonucleotides useful in treatment of Pompe disease
AU2015280252A1 (en) 2014-06-23 2017-01-12 Monsanto Technology Llc Compositions and methods for regulating gene expression via RNA interference
WO2015200697A1 (en) 2014-06-25 2015-12-30 The General Hospital Corporation Targeting human satellite ii (hsatii)
EP3161138A4 (en) 2014-06-25 2017-12-06 Monsanto Technology LLC Methods and compositions for delivering nucleic acids to plant cells and regulating gene expression
CN107001442B (en) 2014-07-15 2021-04-27 耶路撒冷希伯来大学伊森姆研究发展公司 Isolated polypeptide of CD44 and application thereof
US9951327B1 (en) 2014-07-17 2018-04-24 Integrated Dna Technologies, Inc. Efficient and rapid method for assembling and cloning double-stranded DNA fragments
US10378012B2 (en) 2014-07-29 2019-08-13 Monsanto Technology Llc Compositions and methods for controlling insect pests
US10653747B2 (en) 2014-07-31 2020-05-19 Uab Research Foundation ApoE mimetic peptides and higher potency to clear plasma cholesterol
EP3183007B1 (en) 2014-08-19 2020-06-17 Northwestern University Protein/oligonucleotide core-shell nanoparticle therapeutics
WO2016030899A1 (en) 2014-08-28 2016-03-03 Yeda Research And Development Co. Ltd. Methods of treating amyotrophic lateral scleroses
KR102631505B1 (en) 2014-08-29 2024-02-01 알닐람 파마슈티칼스 인코포레이티드 Methods of treating transthyretin(ttr) mediated amyloidosis
EP3185910A4 (en) 2014-08-29 2018-01-10 Children's Medical Center Corporation Methods and compositions for the treatment of cancer
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
EP3191591A1 (en) 2014-09-12 2017-07-19 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
EP3663403A1 (en) 2014-09-26 2020-06-10 University of Massachusetts Rna-modulating agents
WO2016054259A1 (en) 2014-10-01 2016-04-07 Arsia Therapeutics, Inc. Polysaccharide and nucleic acid formulations containing viscosity-lowering agents
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CA2968114A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2016081621A1 (en) 2014-11-18 2016-05-26 Yale University Formulations for targeted release of agents under low ph conditions and methods of use thereof
US10682422B2 (en) 2014-11-18 2020-06-16 Yale University Formulations for targeted release of agents under low pH conditions and methods of use thereof
AU2015349680A1 (en) 2014-11-21 2017-06-08 Northwestern University The sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
WO2016086104A1 (en) 2014-11-25 2016-06-02 Ionis Pharmaceuticals, Inc. Modulation of ube3a-ats expression
CN107532162A (en) 2014-12-12 2018-01-02 托德·M·伍尔夫 For the composition and method using oligonucleotides editor's cell amplifying nucleic acid
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
WO2016108930A2 (en) 2014-12-31 2016-07-07 Geller Bruce L Antisense antibacterial compounds and methods
US10793855B2 (en) 2015-01-06 2020-10-06 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
US10538763B2 (en) 2015-01-16 2020-01-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of DUX4
US9434947B2 (en) 2015-01-20 2016-09-06 Oregon Health & Science University Modulation of KCNH2 isoform expression by oligonucleotides as a therapeutic approach for long QT syndrome
JP6942632B2 (en) 2015-01-22 2021-09-29 モンサント テクノロジー エルエルシー LEPTINOTARSA control composition and its method
EP3247988A4 (en) 2015-01-23 2018-12-19 Vanderbilt University A robust interferometer and methods of using same
US10676726B2 (en) 2015-02-09 2020-06-09 Duke University Compositions and methods for epigenome editing
AU2016219263B2 (en) 2015-02-13 2022-12-01 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (PNPLA3) iRNA compositions and methods of use thereof
US20180200387A1 (en) 2015-02-23 2018-07-19 Crispr Therapeutics Ag Materials and methods for treatment of human genetic diseases including hemoglobinopathies
US10450342B2 (en) 2015-02-23 2019-10-22 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
MX2017011010A (en) 2015-02-26 2017-10-20 Ionis Pharmaceuticals Inc Allele specific modulators of p23h rhodopsin.
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
US10781445B2 (en) 2015-03-11 2020-09-22 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Decoy oligonucleotides for the treatment of diseases
US10036066B2 (en) 2015-03-17 2018-07-31 Dako Denmark A/S In situ hybridization detection method
MA41795A (en) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc EXCLUSION OF AN EXON INDUCED BY ANTISENSE COMPOUNDS IN MYOSTATIN
US20180064748A1 (en) 2015-03-27 2018-03-08 Yeda Research And Development Co. Ltd. Methods of treating motor neuron diseases
KR20180020125A (en) 2015-03-27 2018-02-27 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Modified T cells and methods for their manufacture and use
US10961532B2 (en) 2015-04-07 2021-03-30 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
WO2016164746A1 (en) 2015-04-08 2016-10-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US20180126014A1 (en) 2015-04-15 2018-05-10 Yale University Compositions for enhancing delivery of agents across the blood brain barrier and methods of use thereof
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
KR102258516B1 (en) 2015-04-16 2021-05-31 아이오니스 파마수티컬즈, 인코포레이티드 Compositions for modulating c9orf72 expression
UA126962C2 (en) 2015-05-04 2023-03-01 Монсанто Текнолоджі Елелсі Compositions and methods for controlling arthropod parasite and pest infestations
EP3294280A1 (en) 2015-05-11 2018-03-21 Yeda Research and Development Co., Ltd. Citrin inhibitors for the treatment of cancer
CN107921092B (en) 2015-05-19 2022-04-08 萨勒普塔医疗公司 Peptide oligonucleotide conjugates
US10849917B2 (en) 2015-06-01 2020-12-01 Sarepta Therapeutics, Inc. Antisense-induced exon exclusion in type VII collagen
EP3302053B1 (en) 2015-06-02 2021-03-17 Monsanto Technology LLC Compositions and methods for delivery of a polynucleotide into a plant
EP3302030A4 (en) 2015-06-03 2019-04-24 Monsanto Technology LLC Methods and compositions for introducing nucleic acids into plants
EP3302489A4 (en) 2015-06-04 2019-02-06 Sarepta Therapeutics, Inc. Methods and compounds for treatment of lymphocyte-related diseases and conditions
WO2016201301A1 (en) 2015-06-12 2016-12-15 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
EP3310918B1 (en) 2015-06-18 2020-08-05 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
EP3314250A4 (en) 2015-06-26 2018-12-05 Beth Israel Deaconess Medical Center, Inc. Cancer therapy targeting tetraspanin 33 (tspan33) in myeloid derived suppressor cells
US11414657B2 (en) 2015-06-29 2022-08-16 Ionis Pharmaceuticals, Inc. Modified CRISPR RNA and modified single CRISPR RNA and uses thereof
US10590425B2 (en) 2015-06-29 2020-03-17 Caris Science, Inc. Therapeutic oligonucleotides
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
WO2017011276A1 (en) 2015-07-10 2017-01-19 Ionis Pharmaceuticals, Inc. Modulators of diacyglycerol acyltransferase 2 (dgat2)
US10941176B2 (en) 2015-07-28 2021-03-09 Caris Science, Inc. Therapeutic oligonucleotides
EP3331546B1 (en) 2015-08-03 2023-10-04 Biokine Therapeutics Ltd. Cxcr4 inhibitor for the treatment of cancer
US20180237774A1 (en) 2015-08-04 2018-08-23 Yeda Research And Development Co. Ltd. Methods of screening for riboswitches and attenuators
AU2016312530A1 (en) 2015-08-24 2018-03-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotide nanoparticles for the modulation of gene expression and uses thereof
CN114525280A (en) 2015-09-02 2022-05-24 阿尔尼拉姆医药品有限公司 iRNA compositions of programmed cell death 1 ligand 1(PD-L1) and methods of use thereof
CN108366990B (en) 2015-09-24 2021-09-03 加利福尼亚大学董事会 Synthetic sphingolipid molecules, drugs, methods of their synthesis and methods of treatment
RU2018113709A (en) 2015-09-24 2019-10-30 Айонис Фармасьютикалз, Инк. KRAS EXPRESSION MODULATORS
JP2018532402A (en) 2015-09-24 2018-11-08 クリスパー セラピューティクス アーゲー Novel families of RNA programmable endonucleases and their use in genome editing and other applications
EP4285912A2 (en) 2015-09-25 2023-12-06 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
EP3653216A1 (en) 2015-09-30 2020-05-20 Sarepta Therapeutics, Inc. Methods for treating muscular dystrophy
CN108271351B (en) 2015-10-08 2021-10-26 Ionis 制药公司 Compounds and methods for modulating angiotensinogen expression
EP3858993A1 (en) 2015-10-09 2021-08-04 Sarepta Therapeutics, Inc. Compositions and methods for treating duchenne muscular dystrophy and related disorders
CA3000931A1 (en) 2015-10-28 2017-05-04 Crispr Therapeutics Ag Materials and methods for treatment of duchenne muscular dystrophy
EP3370734B1 (en) 2015-11-05 2023-01-04 Children's Hospital Los Angeles Antisense oligo for use in treating acute myeloid leukemia
EP3371305A1 (en) 2015-11-06 2018-09-12 Crispr Therapeutics AG Materials and methods for treatment of glycogen storage disease type 1a
JP2018531605A (en) 2015-11-06 2018-11-01 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Regulation of apolipoprotein (a) expression
EP4119569A1 (en) 2015-11-06 2023-01-18 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
US11174484B2 (en) 2015-11-10 2021-11-16 B. G. Negev Technologies And Applications Ltd., At Ben- Gurion University Means and methods for reducing tumorigenicity of cancer stem cells
WO2017087708A1 (en) 2015-11-19 2017-05-26 The Brigham And Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
AU2016359629B2 (en) 2015-11-23 2023-03-09 Ranjan BATRA Tracking and manipulating cellular RNA via nuclear delivery of CRISPR/Cas9
JP6932698B2 (en) 2015-12-01 2021-09-08 クリスパー・セラピューティクス・アクチェンゲゼルシャフトCRISPR Therapeutics AG Materials and Methods for the Treatment of Alpha-1 Antitrypsin Deficiency
US11058709B1 (en) 2015-12-04 2021-07-13 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
CA3007152A1 (en) 2015-12-07 2017-06-15 Erasmus University Medical Center Rotterdam Enzymatic replacement therapy and antisense therapy for pompe disease
US20190030176A1 (en) 2015-12-15 2019-01-31 Sarepta Therapeutics, Inc. Peptide oligonucleotide conjugates
US11761007B2 (en) 2015-12-18 2023-09-19 The Scripps Research Institute Production of unnatural nucleotides using a CRISPR/Cas9 system
CA3010084A1 (en) 2015-12-23 2017-06-29 Oregon State University Antisense antibacterial compounds and methods
WO2017112888A1 (en) 2015-12-23 2017-06-29 David Greenberg Antisense antibacterial compounds and methods
BR112018012894A2 (en) 2015-12-23 2018-12-04 Crispr Therapeutics Ag Materials and Methods for Treatment of Amyotrophic Lateral Sclerosis and / or Frontotemporal Lobular Degeneration
CA3006599A1 (en) 2016-01-05 2017-07-13 Ionis Pharmaceuticals, Inc. Methods for reducing lrrk2 expression
WO2017132483A1 (en) 2016-01-29 2017-08-03 Vanderbilt University Free-solution response function interferometry
EP3411078A1 (en) 2016-02-02 2018-12-12 Crispr Therapeutics AG Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
JP2019509721A (en) 2016-02-04 2019-04-11 キュリス,インコーポレイテッド Mutant smoothened and method of using the same
EP3416689B1 (en) 2016-02-18 2023-01-18 CRISPR Therapeutics AG Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
WO2017147087A1 (en) 2016-02-25 2017-08-31 The Brigham And Women's Hospital, Inc. Treatment methods for fibrosis targeting smoc2
WO2017147594A1 (en) 2016-02-26 2017-08-31 Yale University COMPOSITIONS AND METHODS OF USING piRNAS IN CANCER DIAGNOSTICS AND THERAPEUTICS
EP3423106B1 (en) 2016-03-01 2022-07-27 Alexion Pharmaceuticals, Inc. Biodegradable activated polymers for therapeutic delivery
EP3426677B1 (en) 2016-03-08 2023-12-06 Kemyth Biotech Co., Ltd. Use of pneumolysin peptides as antagonists against toll-like receptor 4 and methods of treating toll-like receptor 4 related diseases
CA3013797A1 (en) 2016-03-09 2017-09-14 Ionis Pharmaceuticals, Inc. Methods and compositions for inhibiting pmp22 expression
US10961271B2 (en) 2016-03-16 2021-03-30 Ionis Pharmaceuticals, Inc. Methods of modulating KEAP1
EP3429632B1 (en) 2016-03-16 2023-01-04 CRISPR Therapeutics AG Materials and methods for treatment of hereditary haemochromatosis
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
US10731166B2 (en) 2016-03-18 2020-08-04 Caris Science, Inc. Oligonucleotide probes and uses thereof
US20190117799A1 (en) 2016-04-01 2019-04-25 The Brigham And Women's Hospital, Inc. Stimuli-responsive nanoparticles for biomedical applications
US20190142856A1 (en) 2016-04-13 2019-05-16 Ionis Pharmaceuticals, Inc. Methods for reducing c9orf72 expression
MX2018012729A (en) 2016-04-18 2019-07-04 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies.
KR102522059B1 (en) 2016-04-18 2023-04-14 사렙타 쎄러퓨틱스 인코퍼레이티드 Antisense oligomers and methods of their use to treat diseases associated with the acid alpha-glucosidase gene
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
NZ747685A (en) 2016-04-29 2023-05-26 Sarepta Therapeutics Inc Oligonucleotide analogues targeting human lmna
WO2017191503A1 (en) 2016-05-05 2017-11-09 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
WO2017197128A1 (en) 2016-05-11 2017-11-16 Yale University Poly(amine-co-ester) nanoparticles and methods of use thereof
BR112018074270B1 (en) 2016-05-24 2021-02-02 Sarepta Therapeutics, Inc process for preparing oligomeric compounds as well as said compounds
US11472824B2 (en) 2016-05-24 2022-10-18 Sarepta Therapeutics, Inc. Processes for preparing phosphorodiamidate morpholino oligomers
MA45183A (en) 2016-05-24 2019-04-10 Sarepta Therapeutics Inc PROCESSES FOR THE PREPARATION OF MORPHOLINO OLIGOMERS OF PHOSPHORODIAMIDATE
MA45362A (en) 2016-05-24 2019-04-10 Sarepta Therapeutics Inc PROCESSES FOR THE PREPARATION OF MORPHOLINO OLIGOMERS OF PHOSPHORODIAMIDATE
CA3024178A1 (en) 2016-05-24 2017-12-14 Sarepta Therapeutics, Inc. Pharmaceutical composition comprising eteplirsen
CN109563114B (en) 2016-05-24 2022-08-12 萨勒普塔医疗公司 Process for preparing oligomers
WO2017205879A2 (en) 2016-05-24 2017-11-30 Sarepta Therapeutics, Inc. Processes for preparing phosphorodiamidate morpholino oligomers
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
EP3469083A1 (en) 2016-06-10 2019-04-17 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
US10925973B2 (en) 2016-06-15 2021-02-23 University Of Utah Research Foundation Compositions and methods for using albumin-based nanomedicines
EP3471781A4 (en) 2016-06-17 2020-05-06 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
ES2929047T3 (en) 2016-06-24 2022-11-24 Scripps Research Inst Novel nucleoside triphosphate transporter and uses thereof
EP3478313B1 (en) 2016-06-29 2022-05-04 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis (als) and other related disorders
US11564997B2 (en) 2016-06-29 2023-01-31 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
US11427838B2 (en) 2016-06-29 2022-08-30 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of myotonic dystrophy type 1 (DM1) and other related disorders
AU2017290231A1 (en) 2016-06-30 2019-02-07 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
AU2017292173B2 (en) 2016-07-06 2022-01-13 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of pain related disorders
US11801313B2 (en) 2016-07-06 2023-10-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of pain related disorders
WO2018007871A1 (en) 2016-07-08 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of transthyretin amyloidosis
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
IL264216B2 (en) 2016-07-15 2024-04-01 Ionis Pharmaceuticals Inc Compounds and methods for modulation of smn2
WO2018015936A2 (en) 2016-07-21 2018-01-25 Maxcyte, Inc. Methods and compositions for modifying genomic dna
WO2018020323A2 (en) 2016-07-25 2018-02-01 Crispr Therapeutics Ag Materials and methods for treatment of fatty acid disorders
NL2017294B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Natural cryptic exon removal by pairs of antisense oligonucleotides.
NL2017295B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Antisense oligomeric compound for Pompe disease
WO2018039629A2 (en) 2016-08-25 2018-03-01 Northwestern University Micellar spherical nucleic acids from thermoresponsive, traceless templates
KR102493872B1 (en) 2016-09-02 2023-01-30 다이서나 파마수이티컬, 인크. 4´-phosphate analogs and oligonucleotides comprising the same
AU2017330276B2 (en) 2016-09-20 2022-01-20 The Regents Of The University Of Colorado, A Body Corporate Synthesis of backbone modified morpholino oligonucleotides and chimeras using phosphoramidite chemistry
JOP20190065A1 (en) 2016-09-29 2019-03-28 Ionis Pharmaceuticals Inc Compounds and methods for reducing tau expression
EP3522898A4 (en) 2016-10-06 2020-05-27 Ionis Pharmaceuticals, Inc. Method of conjugating oligomeric compounds
US11459568B2 (en) 2016-10-31 2022-10-04 University Of Massachusetts Targeting microRNA-101-3p in cancer therapy
JOP20190104A1 (en) 2016-11-10 2019-05-07 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CA3045306A1 (en) 2016-11-29 2018-06-07 Boston Biomedical, Inc. Naphthofuran derivatives, preparation, and methods of use thereof
EP3330276A1 (en) 2016-11-30 2018-06-06 Universität Bern Novel bicyclic nucleosides and oligomers prepared therefrom
EP3548620A4 (en) 2016-12-02 2020-07-22 Cold Spring Harbor Laboratory Modulation of lnc05 expression
JP7206214B2 (en) 2016-12-13 2023-01-17 シアトル チルドレンズ ホスピタル (ディービーエイ シアトル チルドレンズ リサーチ インスティテュート) Methods for exogenous drug activation of chemo-inducible signaling complexes expressed in engineered cells in vitro and in vivo
US20200085758A1 (en) 2016-12-16 2020-03-19 The Brigham And Women's Hospital, Inc. Co-delivery of nucleic acids for simultaneous suppression and expression of target genes
KR20230166146A (en) 2016-12-16 2023-12-06 알닐람 파마슈티칼스 인코포레이티드 Methods for treating or preventing ttr-associated diseases using transthyretin(ttr) irna compositions
HUE060065T2 (en) 2016-12-19 2023-01-28 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
BR112019012647A2 (en) 2016-12-19 2019-11-19 Sarepta Therapeutics Inc exon jump oligomer conjugates for muscular dystrophy
CN110636866A (en) 2016-12-19 2019-12-31 萨勒普塔医疗公司 Exon skipping oligomer conjugates for muscular dystrophy
EP3571321A1 (en) 2017-01-23 2019-11-27 Regeneron Pharmaceuticals, Inc. Hydroxysteroid 17-beta dehydrogenase 13 (hsd17b13) variants and uses thereof
WO2018154418A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
AU2018224387A1 (en) 2017-02-22 2019-09-05 Crispr Therapeutics Ag Compositions and methods for gene editing
US11559588B2 (en) 2017-02-22 2023-01-24 Crispr Therapeutics Ag Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
WO2018154462A2 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
US11407997B2 (en) 2017-02-22 2022-08-09 Crispr Therapeutics Ag Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
WO2018165564A1 (en) 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
JOP20190215A1 (en) 2017-03-24 2019-09-19 Ionis Pharmaceuticals Inc Modulators of pcsk9 expression
WO2018183969A1 (en) 2017-03-30 2018-10-04 California Institute Of Technology Barcoded rapid assay platform for efficient analysis of candidate molecules and methods of making and using the platform
CN110913898B (en) 2017-04-18 2024-04-05 阿尔尼拉姆医药品有限公司 Methods of treating subjects having Hepatitis B Virus (HBV) infection
EP3612215A4 (en) 2017-04-20 2021-05-26 aTyr Pharma, Inc. Compositions and methods for treating lung inflammation
WO2018193428A1 (en) 2017-04-20 2018-10-25 Synthena Ag Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
EP3612546B1 (en) 2017-04-20 2022-07-13 Synthena AG Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
US20200384115A1 (en) 2017-04-21 2020-12-10 The Broad Institute , Inc. Targeted delivery to beta cells
CA3062595A1 (en) 2017-05-10 2018-11-15 The Regents Of The University Of California Directed editing of cellular rna via nuclear delivery of crispr/cas9
CA3062506A1 (en) 2017-05-12 2019-05-23 Crispr Therapeutics Ag Materials and methods for engineering cells and uses thereof in immuno-oncology
US20200131555A1 (en) 2017-07-11 2020-04-30 Synthorx, Inc. Incorporation of unnatural nucleotides and methods thereof
MX2020000387A (en) 2017-07-13 2020-08-17 Univ Northwestern General and direct method for preparing oligonucleotide-functiona lized metal-organic framework nanoparticles.
EP3652317A1 (en) 2017-07-13 2020-05-20 Alnylam Pharmaceuticals, Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
EP3651799A1 (en) 2017-07-13 2020-05-20 Massachusetts Institute of Technology Targeting the hdac2-sp3 complex to enhance synaptic function
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
SG11202000939PA (en) 2017-08-03 2020-02-27 Synthorx Inc Cytokine conjugates for the treatment of proliferative and infectious diseases
NL2019390B1 (en) 2017-08-04 2019-02-21 Univ Leiden Screening Method
WO2019032827A1 (en) 2017-08-09 2019-02-14 Massachusetts Institute Of Technology Albumin binding peptide conjugates and methods thereof
WO2019036613A1 (en) 2017-08-18 2019-02-21 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
US10517889B2 (en) 2017-09-08 2019-12-31 Ionis Pharmaceuticals, Inc. Modulators of SMAD7 expression
WO2019060442A1 (en) 2017-09-19 2019-03-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (ttr) mediated amyloidosis
EA201991450A1 (en) 2017-09-22 2019-12-30 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXONISM SKIP IN MUSCULAR DYSTROPHY
US20210145852A1 (en) 2017-09-28 2021-05-20 Sarepta Therapeutics, Inc. Combination Therapies for Treating Muscular Dystrophy
US20200248178A1 (en) 2017-09-28 2020-08-06 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
JP2020536057A (en) 2017-09-28 2020-12-10 サレプタ セラピューティクス, インコーポレイテッド Combination therapy to treat muscular dystrophy
MA50833A (en) 2017-10-17 2020-08-26 Bayer Healthcare Llc COMPOSITIONS AND METHODS FOR GENE EDITING FOR HEMOPHILIA A
WO2019079637A2 (en) 2017-10-18 2019-04-25 Sarepta Therapeutics, Inc. Antisense oligomer compounds
EP3701029A1 (en) 2017-10-26 2020-09-02 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
EP3707256A1 (en) 2017-11-09 2020-09-16 CRISPR Therapeutics AG Self-inactivating (sin) crispr/cas or crispr/cpf1 systems and uses thereof
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
WO2019099610A1 (en) 2017-11-16 2019-05-23 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
US10953036B2 (en) 2017-11-20 2021-03-23 University Of Georgia Research Foundation, Inc. Compositions and methods of modulating HIF-2A to improve muscle generation and repair
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
AU2018372763A1 (en) 2017-11-21 2020-05-28 Bayer Healthcare Llc Materials and methods for treatment of autosomal dominant Retinitis Pigmentosa
CA3084955A1 (en) 2017-12-05 2019-06-13 Vertex Pharmaceuticals Incorporated Crispr-cas9 modified cd34+ human hematopoietic stem and progenitor cells and uses thereof
US11725208B2 (en) 2017-12-14 2023-08-15 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
WO2019118935A1 (en) 2017-12-14 2019-06-20 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and their use in genome editing and other applications
MX2020006012A (en) 2017-12-18 2020-09-14 Alnylam Pharmaceuticals Inc High mobility group box-1 (hmgb1) irna compositions and methods of use thereof.
CN111836892A (en) 2017-12-21 2020-10-27 克里斯珀医疗股份公司 Materials and methods for treating type 2A uker syndrome
WO2019123430A1 (en) 2017-12-21 2019-06-27 Casebia Therapeutics Llp Materials and methods for treatment of usher syndrome type 2a and/or non-syndromic autosomal recessive retinitis pigmentosa (arrp)
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
CA3088180A1 (en) 2018-01-12 2019-07-18 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting transferrin
AU2019206731A1 (en) 2018-01-15 2020-07-30 Ionis Pharmaceuticals, Inc. Modulators of DNM2 expression
EP3740472A1 (en) 2018-01-19 2020-11-25 Synthena AG Tricyclo-dna nucleoside precursors and processes for preparing the same
US20190233816A1 (en) 2018-01-26 2019-08-01 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
US11566236B2 (en) 2018-02-05 2023-01-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
US11268077B2 (en) 2018-02-05 2022-03-08 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2019155465A1 (en) 2018-02-08 2019-08-15 Yeda Research And Development Co. Ltd. Methods of identifying and using agents for treating diseases associated with intestinal barrier dysfunction
JP7317029B2 (en) 2018-02-12 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド Modified compounds and uses thereof
EP3752616A1 (en) 2018-02-16 2020-12-23 CRISPR Therapeutics AG Compositions and methods for gene editing by targeting fibrinogen-alpha
SG11202007728QA (en) 2018-02-26 2020-09-29 Synthorx Inc Il-15 conjugates and uses thereof
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
EP3759127A4 (en) 2018-03-02 2022-03-30 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-beta precursor protein
CN112105625A (en) 2018-03-07 2020-12-18 赛诺菲 Nucleotide precursors, nucleotide analogs, and oligomeric compounds containing the same
CN112424348A (en) 2018-03-19 2021-02-26 克里斯珀医疗股份公司 Novel RNA-programmable endonuclease system and uses thereof
WO2019183440A1 (en) 2018-03-22 2019-09-26 Ionis Pharmaceuticals, Inc. Methods for modulating fmr1 expression
WO2019186514A2 (en) 2018-03-30 2019-10-03 Rheinische Friedrich-Wilhelms-Universitat Bonn Aptamers for targeted activaton of t cell-mediated immunity
JP2021520781A (en) 2018-04-06 2021-08-26 チルドレンズ メディカル センター コーポレーションChildren’S Medical Center Corporation Compositions and Methods for Somatic Cell Reprogramming and Imprinting Modulation
CA3094020A1 (en) 2018-04-11 2019-10-17 Ionis Pharmaceuticals, Inc. Modulators of ezh2 expression
WO2019204668A1 (en) 2018-04-18 2019-10-24 Casebia Therapeutics Limited Liability Partnership Compositions and methods for knockdown of apo(a) by gene editing for treatment of cardiovascular disease
AU2019266307A1 (en) 2018-05-09 2020-11-19 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ATXN3 expression
BR112020020957B1 (en) 2018-05-09 2022-05-10 Ionis Pharmaceuticals, Inc Oligomeric compounds, population and pharmaceutical composition thereof and their uses
TW202016304A (en) 2018-05-14 2020-05-01 美商阿尼拉製藥公司 Angiotensinogen (agt) irna compositions and methods of use thereof
US10765760B2 (en) 2018-05-29 2020-09-08 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
EP4219717A3 (en) 2018-06-13 2023-12-20 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
AU2019287635A1 (en) 2018-06-14 2020-12-17 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing STMN2 expression
SG11202011864XA (en) 2018-06-27 2020-12-30 Ionis Pharmaceuticals Inc Compounds and methods for reducing lrrk2 expression
KR20210027389A (en) 2018-06-28 2021-03-10 크리스퍼 테라퓨틱스 아게 Compositions and methods for genome editing by insertion of donor polynucleotides
WO2020023737A1 (en) 2018-07-25 2020-01-30 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing atxn2 expression
TW202020153A (en) 2018-07-27 2020-06-01 美商薩羅塔治療公司 Exon skipping oligomers for muscular dystrophy
US10857174B2 (en) 2018-07-27 2020-12-08 United States Government As Represented By The Department Of Veterans Affairs Morpholino oligonucleotides useful in cancer treatment
EP3830278A4 (en) 2018-08-01 2022-05-25 University of Georgia Research Foundation, Inc. Compositions and methods for improving embryo development
TW202023574A (en) 2018-08-13 2020-07-01 美商阿尼拉製藥公司 Hepatitis b virus (hbv) dsrna agent compositions and methods of use thereof
WO2020037125A1 (en) 2018-08-16 2020-02-20 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the lect2 gene
CN113329739A (en) 2018-08-20 2021-08-31 罗贡股份有限公司 Antisense oligonucleotides targeting SCN2A for the treatment of SCN1A encephalopathy
CA3110661A1 (en) 2018-08-29 2020-03-05 University Of Massachusetts Inhibition of protein kinases to treat friedreich ataxia
WO2020051507A1 (en) 2018-09-06 2020-03-12 The Broad Institute, Inc. Nucleic acid assemblies for use in targeted delivery
EP3620520A1 (en) 2018-09-10 2020-03-11 Universidad del Pais Vasco Novel target to treat a metabolic disease in an individual
SG11202102531WA (en) 2018-09-14 2021-04-29 Univ Northwestern Programming protein polymerization with dna
AU2019344776A1 (en) 2018-09-18 2021-01-21 Alnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA compositions and methods of use thereof
TW202023573A (en) 2018-09-19 2020-07-01 美商Ionis製藥公司 Modulators of pnpla3 expression
AU2019362000A1 (en) 2018-10-17 2021-05-20 Bayer Healthcare Llc Compositions and methods for delivering transgenes
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
TW202039840A (en) 2018-11-15 2020-11-01 美商伊奧尼斯醫藥公司 Modulators of irf5 expression
US20220025366A1 (en) 2018-11-21 2022-01-27 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing prion expression
IL263184A (en) 2018-11-21 2020-05-31 Yarden Yosef Method of treating cancer and compositions for same
US20210332495A1 (en) 2018-12-06 2021-10-28 Northwestern University Protein Crystal Engineering Through DNA Hybridization Interactions
EP3894558A1 (en) 2018-12-13 2021-10-20 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
EP3898977A1 (en) 2018-12-20 2021-10-27 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of kcnt1 related disorders
SI3897672T1 (en) 2018-12-20 2024-02-29 Humabs Biomed Sa Combination hbv therapy
MX2021008628A (en) 2019-01-16 2021-11-17 Genzyme Corp Serpinc1 irna compositions and methods of use thereof.
CA3128093A1 (en) 2019-01-31 2020-08-06 Ionis Pharmaceuticals, Inc. Modulators of yap1 expression
WO2020157760A1 (en) 2019-01-31 2020-08-06 Bar Ilan University Neoantigens created by aberrant-induced splicing and uses thereof in enhancing immunotherapy
KR20210123299A (en) 2019-02-06 2021-10-13 신톡스, 인크. IL-2 conjugates and methods of use thereof
AU2020221340A1 (en) 2019-02-15 2021-09-16 Bayer Healthcare Llc Gene editing for hemophilia A with improved Factor VIII expression
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
JP2022521010A (en) 2019-02-21 2022-04-04 イッスム・リサーチ・デベロプメント・カムパニー・オブ・ザ・ヘブリュー・ユニバシティー・オブ・エルサレム リミテッド Methods for reducing drug-induced nephrotoxicity
SG11202108450SA (en) 2019-02-27 2021-09-29 Ionis Pharmaceuticals Inc Modulators of malat1 expression
SG11202109741VA (en) 2019-03-12 2021-10-28 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
MA55515A (en) 2019-03-28 2022-02-09 Sarepta Therapeutics Inc METHODS OF TREATING MUSCULAR DYSTROPHY WITH CASIMERSEN
US20220228140A1 (en) 2019-03-29 2022-07-21 Mitsubishi Tanabe Pharma Corporation Compound, method and pharmaceutical composition for modulating expression of dux4
KR20210145213A (en) 2019-03-29 2021-12-01 다이서나 파마수이티컬, 인크. Compositions and methods for treating a KRAS-associated disease or disorder
CN117431244A (en) 2019-03-29 2024-01-23 Ionis制药公司 Compounds and methods for modulating UBE3A-ATS
WO2020214763A1 (en) 2019-04-18 2020-10-22 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US11814464B2 (en) 2019-04-29 2023-11-14 Yale University Poly(amine-co-ester) polymers and polyplexes with modified end groups and methods of use thereof
AU2020268798A1 (en) 2019-05-03 2021-11-04 Dicerna Pharmaceuticals, Inc. Double-stranded nucleic acid inhibitor molecules with shortened sense strands
EP3966327A1 (en) 2019-05-08 2022-03-16 Vertex Pharmaceuticals Incorporated Crispr/cas all-in-two vector systems for treatment of dmd
KR20220036914A (en) 2019-05-13 2022-03-23 비르 바이오테크놀로지, 인코포레이티드 Compositions and methods for treating hepatitis B virus (HBV) infection
WO2020243292A1 (en) 2019-05-28 2020-12-03 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing fus expression
BR112021025130A2 (en) 2019-06-14 2022-03-15 Scripps Research Inst Reagents and methods for replication, transcription and translation in semisynthetic organisms
EP3983542A2 (en) 2019-06-17 2022-04-20 CRISPR Therapeutics AG Methods and compositions for improved homology directed repair
JP2022536495A (en) 2019-06-19 2022-08-17 サレプタ セラピューティクス, インコーポレイテッド How to treat muscular dystrophy
WO2021021673A1 (en) 2019-07-26 2021-02-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021022109A1 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. SERPIN FAMILY F MEMBER 2 (SERPINF2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021025899A1 (en) 2019-08-02 2021-02-11 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer pharmaceutical compositions
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
JP2022544587A (en) 2019-08-15 2022-10-19 アイオーニス ファーマシューティカルズ, インコーポレーテッド Bond-modified oligomeric compounds and uses thereof
KR20220047598A (en) 2019-08-15 2022-04-18 신톡스, 인크. Immuno-oncology Combination Therapy Using IL-2 Conjugates
US20210054040A1 (en) 2019-08-23 2021-02-25 Synthorx, Inc. Novel il-15 conjugates and uses thereof
CA3149421A1 (en) 2019-08-30 2021-03-04 Yale University Compositions and methods for delivery of nucleic acids to cells
WO2021046122A1 (en) 2019-09-03 2021-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
EP4025582A1 (en) 2019-09-05 2022-07-13 Sanofi Oligonucleotides containing nucleotide analogs
KR20220061158A (en) 2019-09-10 2022-05-12 신톡스, 인크. IL-2 conjugates and methods of use for treating autoimmune diseases
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021074772A1 (en) 2019-10-14 2021-04-22 Astrazeneca Ab Modulators of pnpla3 expression
EP4045652A1 (en) 2019-10-18 2022-08-24 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
BR112022007540A2 (en) 2019-10-22 2022-07-12 Alnylam Pharmaceuticals Inc COMPONENTS COMPLEMENTARY C3 IRNA COMPOSITIONS AND METHODS OF USE THEREOF
CA3155921A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
EP4051796A1 (en) 2019-11-01 2022-09-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
JP2022554272A (en) 2019-11-04 2022-12-28 シンソークス, インコーポレイテッド Interleukin 10 conjugates and uses thereof
CN114981431A (en) 2019-11-13 2022-08-30 阿尔尼拉姆医药品有限公司 Methods and compositions for treating Angiotensinogen (AGT) -related disorders
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
AU2020391215A1 (en) 2019-11-27 2022-06-02 Bayer Healthcare Llc Methods of synthesizing RNA molecules
JP2023506181A (en) 2019-12-13 2023-02-15 アルナイラム ファーマシューティカルズ, インコーポレイテッド Human chromosome 9 open reading frame 72 (C9ORF72) iRNA agent compositions and methods of use thereof
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2021122944A1 (en) 2019-12-18 2021-06-24 Alia Therapeutics Srl Compositions and methods for treating retinitis pigmentosa
IL294599A (en) 2020-01-15 2022-09-01 Dicerna Pharmaceuticals Inc 4’-o-methylene phosphonate nucleic acids and analogues thereof
US20230057461A1 (en) 2020-01-27 2023-02-23 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Rab13 and net1 antisense oligonucleotides to treat metastatic cancer
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
EP4103714A1 (en) 2020-02-10 2022-12-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
WO2021163281A1 (en) 2020-02-12 2021-08-19 Accutar Biotechnology Inc. Antisense oligonucleotides and their use for treating pendred syndrome
EP4107265A1 (en) 2020-02-18 2022-12-28 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
CR20220485A (en) 2020-02-28 2022-11-10 Ionis Pharmaceuticals Inc Compounds and methods for modulating smn2
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
EP4114948A1 (en) 2020-03-06 2023-01-11 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
JP2023519274A (en) 2020-03-26 2023-05-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド CORONAVIRUS iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
JP2023520582A (en) 2020-04-06 2023-05-17 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for silencing MYOC expression
EP4133076A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Angiotensin-converting enzyme 2 (ace2) irna compositions and methods of use thereof
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
US20230159933A1 (en) 2020-04-07 2023-05-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
CN115955972A (en) 2020-04-27 2023-04-11 阿尔尼拉姆医药品有限公司 Apolipoprotein E (APOE) iRNA agent compositions and methods of use thereof
AU2021265813A1 (en) 2020-04-30 2022-11-10 Alnylam Pharmaceuticals, Inc. Complement factor B (CFB) iRNA compositions and methods of use thereof
CA3181546A1 (en) 2020-05-01 2021-11-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating atxn1
US20230227824A1 (en) 2020-05-12 2023-07-20 Mitsubishi Tanabe Pharma Corporation Compound, method and pharmaceutical composition for regulating expression of ataxin 3
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
CA3162416C (en) 2020-05-15 2023-07-04 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
EP4150086A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
WO2021231691A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rsi)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
US20230183707A1 (en) 2020-05-21 2023-06-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
US20230203484A1 (en) 2020-05-22 2023-06-29 Wave Life Sciences Ltd. Double stranded oligonucleotide compositions and methods relating thereto
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
JP2023530234A (en) 2020-06-05 2023-07-14 ザ・ブロード・インスティテュート・インコーポレイテッド Compositions and methods for treating neoplasms
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
CA3184289A1 (en) 2020-06-18 2021-12-23 Alnylam Pharmaceuticals, Inc. Xanthine dehydrogenase (xdh) irna compositions and methods of use thereof
AU2021293780A1 (en) 2020-06-19 2023-02-02 Yale University Poly(amine-co-ester) polymers with modified end groups and enhanced pulmonary delivery
KR20230042023A (en) 2020-06-24 2023-03-27 비르 바이오테크놀로지, 인코포레이티드 Engineered hepatitis B virus neutralizing antibodies and uses thereof
AU2021296622A1 (en) 2020-06-25 2023-02-23 Synthorx, Inc. Immuno oncology combination therapy with IL-2 conjugates and anti-EGFR antibodies
WO2022006134A2 (en) 2020-06-29 2022-01-06 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating plp1
WO2022031433A1 (en) 2020-08-04 2022-02-10 Dicerna Pharmaceuticals, Inc. Systemic delivery of oligonucleotides
MX2023002480A (en) 2020-08-31 2023-05-18 Univ Yale Compositions and methods for delivery of nucleic acids to cells.
TW202227102A (en) 2020-09-22 2022-07-16 瑞典商阿斯特捷利康公司 Method of treating fatty liver disease
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
US20220290136A1 (en) 2020-09-30 2022-09-15 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis
EP3978608A1 (en) 2020-10-05 2022-04-06 SQY Therapeutics Oligomeric compound for dystrophin rescue in dmd patients throughout skipping of exon-51
WO2022076291A1 (en) 2020-10-05 2022-04-14 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
KR20230084204A (en) 2020-10-09 2023-06-12 신톡스, 인크. Immuno-oncology therapy using IL-2 conjugates
BR112023006364A2 (en) 2020-10-09 2023-05-09 Synthorx Inc IMMUNO-ONCOLOGY COMBINATION THERAPY WITH IL-2 AND PEMBROLIZUMAB CONJUGATES
WO2022079719A1 (en) 2020-10-15 2022-04-21 Yeda Research And Development Co. Ltd. Method of treating myeloid malignancies
JP2023546199A (en) 2020-10-20 2023-11-01 サノフイ Novel ligands for asialoglycoprotein receptors
WO2022087041A1 (en) 2020-10-21 2022-04-28 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
WO2022087329A1 (en) 2020-10-23 2022-04-28 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
EP4244355A1 (en) 2020-11-13 2023-09-20 Alnylam Pharmaceuticals, Inc. Coagulation factor v (f5) irna compositions and methods of use thereof
EP4136092A4 (en) 2020-11-18 2023-10-11 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
US20240002853A1 (en) 2020-11-23 2024-01-04 Alpha Anomeric Sas Nucleic acid duplexes
EP4256053A1 (en) 2020-12-01 2023-10-11 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
GB2603454A (en) 2020-12-09 2022-08-10 Ucl Business Ltd Novel therapeutics for the treatment of neurodegenerative disorders
JP2024501288A (en) 2020-12-23 2024-01-11 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー Compositions of modified TREM and uses thereof
WO2022140535A1 (en) 2020-12-23 2022-06-30 Sarepta Therapeutics, Inc. Compositions comprising exon skipping oligonucleotide conjugates for treating muscular dystrophy
EP4274896A1 (en) 2021-01-05 2023-11-15 Alnylam Pharmaceuticals, Inc. Complement component 9 (c9) irna compositions and methods of use thereof
KR20230146048A (en) 2021-02-12 2023-10-18 알닐람 파마슈티칼스 인코포레이티드 Superoxide dismutase 1 (SOD1) IRNA compositions and methods of using them to treat or prevent superoxide dismutase 1- (SOD1-)-related neurodegenerative diseases
EP4291243A1 (en) 2021-02-12 2023-12-20 Synthorx, Inc. Lung cancer combination therapy with il-2 conjugates and an anti-pd-1 antibody or antigen-binding fragment thereof
WO2022174101A1 (en) 2021-02-12 2022-08-18 Synthorx, Inc. Skin cancer combination therapy with il-2 conjugates and cemiplimab
EP4298220A1 (en) 2021-02-25 2024-01-03 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods of use thereof
IL305153A (en) 2021-02-26 2023-10-01 Alnylam Pharmaceuticals Inc KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
IL305418A (en) 2021-03-04 2023-10-01 Alnylam Pharmaceuticals Inc Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
WO2022192038A1 (en) 2021-03-12 2022-09-15 Northwestern University Antiviral vaccines using spherical nucleic acids
IL307239A (en) 2021-03-29 2023-11-01 Alnylam Pharmaceuticals Inc Huntingtin (htt) irna agent compositions and methods of use thereof
WO2022212153A1 (en) 2021-04-01 2022-10-06 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
BR112023022284A2 (en) 2021-04-26 2023-12-26 Alnylam Pharmaceuticals Inc COMPOSITIONS OF TRANSMEMBRANE PROTEASE IRNA, SERINE 6 (TMPRSS6) AND METHODS OF USE THEREOF
EP4330396A1 (en) 2021-04-29 2024-03-06 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
CA3216839A1 (en) 2021-04-30 2022-11-03 Sarepta Therapeutics, Inc. Treatment methods for muscular dystrophy
EP4334448A1 (en) 2021-05-03 2024-03-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (ttr) mediated amyloidosis
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
TW202317762A (en) 2021-06-02 2023-05-01 美商艾拉倫製藥股份有限公司 Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
TW202313117A (en) 2021-06-03 2023-04-01 美商欣爍克斯公司 Head and neck cancer combination therapy comprising an il-2 conjugate and cetuximab
TW202308663A (en) 2021-06-04 2023-03-01 美商艾拉倫製藥股份有限公司 Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
AR126070A1 (en) 2021-06-08 2023-09-06 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR TREATING OR PREVENTING STARGARDT DISEASE AND/OR DISORDERS ASSOCIATED WITH RETINOL BORDER PROTEIN 4 (RBP4)
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
AU2022293556A1 (en) 2021-06-18 2024-01-18 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ifnar1 expression
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
AU2022303164A1 (en) 2021-06-30 2024-01-18 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2023285431A1 (en) 2021-07-12 2023-01-19 Alia Therapeutics Srl Compositions and methods for allele specific treatment of retinitis pigmentosa
TW202333748A (en) 2021-07-19 2023-09-01 美商艾拉倫製藥股份有限公司 Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
IL309905A (en) 2021-07-23 2024-03-01 Alnylam Pharmaceuticals Inc Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
WO2023014677A1 (en) 2021-08-03 2023-02-09 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
TW202337474A (en) 2021-08-04 2023-10-01 美商艾拉倫製藥股份有限公司 Irna compositions and methods for silencing angiotensinogen (agt)
TW202334413A (en) 2021-08-13 2023-09-01 美商艾拉倫製藥股份有限公司 Factor xii (f12) irna compositions and methods of use thereof
WO2023034870A2 (en) 2021-09-01 2023-03-09 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing dmpk expression
WO2023034515A2 (en) 2021-09-03 2023-03-09 Sarepta Therapeutics, Inc. Delivery of anitsense oligomers by mirror image peptides
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
WO2023044094A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
WO2023055774A1 (en) 2021-09-30 2023-04-06 Sarepta Therapeutics, Inc. Antisense oligonucleotides having one or more abasic units
WO2023070086A1 (en) 2021-10-22 2023-04-27 Sarepta Therapeutics, Inc. Morpholino oligomers for treatment of peripheral myelin protein 22 related diseases
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
AR127477A1 (en) 2021-10-29 2024-01-31 Alnylam Pharmaceuticals Inc COMPOSITIONS OF RNAi AGAINST COMPLEMENT FACTOR B (CFB) AND METHODS OF USE THEREOF
WO2023086295A2 (en) 2021-11-10 2023-05-19 University Of Rochester Antisense oligonucleotides for modifying protein expression
WO2023086292A2 (en) 2021-11-10 2023-05-19 University Of Rochester Gata4-targeted therapeutics for treatment of cardiac hypertrophy
GB202117758D0 (en) 2021-12-09 2022-01-26 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023118349A1 (en) 2021-12-21 2023-06-29 Alia Therapeutics Srl Type ii cas proteins and applications thereof
WO2023122750A1 (en) 2021-12-23 2023-06-29 Synthorx, Inc. Cancer combination therapy with il-2 conjugates and cetuximab
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023159189A1 (en) 2022-02-18 2023-08-24 Yale University Branched poly(amine-co-ester) polymers for more efficient nucleic expression
WO2023168427A1 (en) 2022-03-03 2023-09-07 Yale University Compositions and methods for delivering therapeutic polynucleotides for exon skipping
WO2023178230A1 (en) 2022-03-17 2023-09-21 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer conjugates
WO2023177866A1 (en) 2022-03-18 2023-09-21 Dicerna Pharmaceuticals, Inc. Decarboxylative acetoxylation using mn(ii) or mn(iii) reagent for synthesis of 4'-acetoxy- nucleoside and use thereof for synthesis of corresponding 4'-(dimethoxyphosphoryl)methoxy- nucleotide
WO2023194359A1 (en) 2022-04-04 2023-10-12 Alia Therapeutics Srl Compositions and methods for treatment of usher syndrome type 2a
WO2023205451A1 (en) 2022-04-22 2023-10-26 Entrada Therapeutics, Inc. Cyclic peptides for delivering therapeutics
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2024015924A2 (en) 2022-07-15 2024-01-18 Entrada Therapeutics, Inc. Hybrid oligonucleotides
WO2024039776A2 (en) 2022-08-18 2024-02-22 Alnylam Pharmaceuticals, Inc. Universal non-targeting sirna compositions and methods of use thereof
WO2024050261A1 (en) 2022-08-29 2024-03-07 University Of Rochester Antisense oligonucleotide-based anti-fibrotic therapeutics
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
WO2024056880A2 (en) 2022-09-16 2024-03-21 Alia Therapeutics Srl Enqp type ii cas proteins and applications thereof
WO2024064237A2 (en) 2022-09-21 2024-03-28 Sarepta Therapeutics, Inc. Dmd antisense oligonucleotide-mediated exon skipping efficiency

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8311228D0 (en) * 1983-04-25 1983-06-02 Fujisawa Pharmaceutical Co Morpholine derivatives
DE3650349T2 (en) * 1985-03-15 1995-12-14 Antivirals Inc IMMUNOTEST FOR POLYNUCLEOTID AND METHOD.
ATE220407T1 (en) * 1989-12-20 2002-07-15 Antivirals Inc UNCHARGED MORPHOLINE BASED POLYMERS WITH CHIRAL PHOSPHORUS CONTAINING BRIDGES BETWEEN SUBUNITS
JPH09118898A (en) * 1995-08-23 1997-05-06 Kao Corp Bleaching agent composition for herd surface

Also Published As

Publication number Publication date
ATE157098T1 (en) 1997-09-15
EP0592511B1 (en) 1997-08-20
US5166315A (en) 1992-11-24
CA2110988A1 (en) 1993-01-07
AU2253192A (en) 1993-01-25
DE69221730T2 (en) 1998-02-26
WO1993000352A1 (en) 1993-01-07
KR100225090B1 (en) 1999-10-15
DE69221730D1 (en) 1997-09-25
TW466243B (en) 2001-12-01
EP0592511A1 (en) 1994-04-20
AU665560B2 (en) 1996-01-11
JPH06508526A (en) 1994-09-29

Similar Documents

Publication Publication Date Title
CA2110988C (en) Sequence-specific binding polymers for duplex nucleic acids
US5405938A (en) Sequence-specific binding polymers for duplex nucleic acids
US5142047A (en) Uncharged polynucleotide-binding polymers
US5217866A (en) Polynucleotide assay reagent and method
EP0962463B1 (en) Uncharged morpholino-based polymers having phosphorus-containing chiral intersubunit linkages
US5235033A (en) Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5705333A (en) Peptide-based nucleic acid mimics(PENAMS)
US5034506A (en) Uncharged morpholino-based polymers having achiral intersubunit linkages
US11384105B2 (en) Processes for preparing oligomers
US5470974A (en) Uncharged polynucleotide-binding polymers
US5378841A (en) Alpha-morpholino ribonucleoside derivatives and polymers thereof
CA3076694A1 (en) Divalent nucleobase compounds and uses therefor
Verheijen et al. 2′, 5′-Oligoadenylate-peptide nucleic acids (2–5A-PNAs) activate RNase L
Eritja Solid-phase synthesis of modified oligonucleotides
Vaijayanthi et al. Recent advances in oligonucleotide synthesis and their applications
Buchardt et al. Synthetic Procedures for Peptide Nucleic Acids
Koh Design of novel bases for recognition of GC base pairs by oligonucleotide-directed triple helix formation

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed