CA2076037A1 - Method of site-specific alteration of rna and production of encoded polypeptides - Google Patents

Method of site-specific alteration of rna and production of encoded polypeptides

Info

Publication number
CA2076037A1
CA2076037A1 CA002076037A CA2076037A CA2076037A1 CA 2076037 A1 CA2076037 A1 CA 2076037A1 CA 002076037 A CA002076037 A CA 002076037A CA 2076037 A CA2076037 A CA 2076037A CA 2076037 A1 CA2076037 A1 CA 2076037A1
Authority
CA
Canada
Prior art keywords
rna
rna molecule
segment
rnase
modified
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002076037A
Other languages
French (fr)
Inventor
Thoru Pederson
Sudhir Agrawal
Sandra Mayrand
Paul C. Zamecnik
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.)
Worcester Foundation for Biomedical Research
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2076037A1 publication Critical patent/CA2076037A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8216Methods for controlling, regulating or enhancing expression of transgenes in plant cells
    • C12N15/8222Developmentally regulated expression systems, tissue, organ specific, temporal or spatial regulation
    • C12N15/823Reproductive tissue-specific promoters
    • C12N15/8233Female-specific, e.g. pistil, ovule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8216Methods for controlling, regulating or enhancing expression of transgenes in plant cells
    • C12N15/8222Developmentally regulated expression systems, tissue, organ specific, temporal or spatial regulation
    • C12N15/823Reproductive tissue-specific promoters
    • C12N15/8235Fruit-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/312Phosphonates
    • C12N2310/3125Methylphosphonates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/314Phosphoramidates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates

Abstract

A method of site-directed alteration (removal or removal followed by replacement) of selected nucleotides in an RNA molecule, as well as to mixed phosphate backbone oligonucleotides useful in the method. It further relates to a method of producing polypeptides or proteins encoded by the RNA molecule altered by the present method. Through use of the present method, site-directed cleavage of an RNA molecule is effected, followed by excision of the selected or target segment of the RNA molecule.

Description

U'091/12323 PCT/US9ltO0968 HETHOD O~ SITE SPECIFIC ALTERATION O~ RNA AND
PRODUCTION OF ENCODED POLYPEPTIDES

Description Back~round ____ _____ 05 Several aspects of contemporary molecular genetics and biotechnology make it desirable to be able to produce genetically-altered proteins. For example, mu~ated protein domains are sometimes hyper-immunogenic, facilitating the production of neutralizing antibody-based vaccines. Moreover, site-directed mutations, ideally one amino ~cid at a time, csn be a powerful approach to deciphering protein structure and/or enzyme-substrate reaction mechanisms.
Typically, deletions or substitutions of amino acids are made at the gene or DNA level, by recombinant DNA
techniques ~hich rely on the use of restriction endonucleases. However, restriction endonucleases available have a limited array of target sites in DNA , (usually palindromic hexanucleotide or octanucleotide z~ sequences). Deletion of a particular in-frame trinucleo-tide or trinucleotides ~ay not be possible because there ~ay be no suitably located restriction sites. As a result, presently-available methods of altering an amino . . . .: .
. . ~ .

.

WO9~/1232~ PCT/US91/00968 acid sequence by altering the DNA sequence which encodes it, are limited in their applicsbility.

Summary of the Inventlon ______ _ _ _______._ __ The present invention relates to a method of site-05 directed alteration (removal or re~o~al followed by replacement) of selected nucleotides in an RNA ~o~ecule, as well ns to ~i~ed phosphate backbone oligonucleotides useful in the method. It further relates to ~ method of producing polypept~des or proteins encoded by the RNA
molecule altered by the present-method. Through use of the present method, site-directed cleavage of an RNA
molecule is effected, followed by e~cision of the selected or target segment of the RNA molecule.
Follo~ing cleavage and excision, in onç embodiment, the two segmen~s of the resulting interrupted RNA.molecule ~re ~oined, through the sction of an appropriate ligase.
This results in production of a continuous RNA molecule, referred to as an altered RNA molecule, which is the same as the original RNA molecule except that it lacks the nucleotides originally pres~ent in the target segment of the RNA molecule. In a second embodiment, selected nucleotides can be introduced into the space or gap created by removal of the target RNA segment; a continuous RNA molecule is created by ligating the selected nucleotides introduced in this manner to the nYcleotide on each side of the gap.
ln the present ~etkod, an RNA molecule ~hose nucleo-tide sequence is to be altered in a site-directed manner -: . .
' .
, 3 PCT/~S91/00968 2076~37 is brou~ht into contact with an oligonucleotide, referred to as a mixed phosphate backbone oligonucleotide, in the presence of RN~se H. The mixed phosphste backbone oligonucleotide is complementary to all or a portion of 05 an RNA ~olecule ~hich includes a target segment to be altered. ln addition, the mixed phosphate backbone oligonucleotide includea ~n internal portion or ~egment of deoxynucleotides which is capsble of sctivatin~ RNase H ~nd is flanked on each side by a cequence of nucleo-tides ~hich is unable to sctivate RN~se H. The internalsequence includes two or ~ore consecutive phosphodiester linka~es, ~hich may be unmodified or modified. The flanking sequences are modified deo~yribonucleotide or ribonucleotide sequences. It has been shown that when lS such a mixed phosphate backbone oligonucleotide is contacted with a target segment of an RNA molecule, according to the method of th,e present invention~ the result is RNase H metiated excision of RNA target nucleotides complementary only to the internal sequence of oligonucleotides. This mekes it possible to excise precisely any tesired nucleotide or oligonucleotide from an RNA molecule. Followed by ~NA ligation, this results in a desired altered messenger RNA or other type of RNA. ;~
Thus, for the first time, it is possible to carry out -2S precise excision of a selected segment of sn RNA
molecule.
As a result, it is possible to selectively telete any desired number of nucleotides and, if desired, to ~ntroduce replacement nucleotides. The encoded amino acid sequence or polypeptide can be produced by express-ing the ~ltered RNA in vitro or in vivo. As a result, a ... .. .

.

WO91/12323 PCT/USgl/00968 selected a~ino acid sequence or selected polypeptide csn be produced by the present method by: l) producing an altered RNA ~olecule encoding a elected ~mino acld sequence or selected polypeptide snd 2) expressing the O5 ~ltered RNA ~olecule under appropr1ate conditlons.
Cell-free translation of the altercd RNA molecule can be cnrried out to produce deslred mutant proteins use~ul, for e~ample, for ~tudies of protein structure or func-tion. Aleernati~ely, an appropriate ~ixed phosphate backbone oli~onucleotide can be -taken up by or introduced into cultured cells or into c"eils of an animal or a plsnt, in ~hich endogenoùs RNase H and RNA ligase activities can produce altered RNAs; upon translation, corresponding genetically altered protein~ are produced.
lS This is useful, for example, as a means of producing defective viral or infectious/pathogenlc agent seplication or gene expression, which can be useful therapeutically or prophylactically.

Brief Descri~tion of the Drawings Figure l is a schematic representation of the present method for excision of a tar~eted segment of an RNA molecule in ~hich sequence I is RNA from which a targeted segment is to be excised and sequence II is a mixed phosphate backbone oligonucleotide.
Subscript "m designates an RNase H-resistant internucleoside phosphate; subscript "s" indicates an RNase H-suscept~ble internucleoside phosphate; "X"
designates any of the four ribonucleotides A, C, G or U;
and ~Y~ des~gnates a deoxyribonucleotide complementary to 30 the ribonucleotide directly abo~e it. ' Figure 2 is a schematic representatlon of repair of a genetic defect by the method of the present invention, in ~hlch IVa and IVb represent a double-stranded hybrid consisting of a smaller unmodified ribonucleotide (IVa) 05 hybr~dized to B lsrger nodifled, RNase H-resistant deo~yribonucleot~de (IVb) and V is the repa~red RNA
hybridized to the RNase N-r6sistant deo~yr~bonucleot~de.

Detailed Descri~tion of the Invention The present invent~on relstes to a method of site-specific or site-directed alteration (removal or removal follo~ed by replacement) of selected nucleotides (i.e., a target segment) in an RNA molecule, to produce an altered RNA sequence, as well as to mixed phosphate back~one oligonucleotides useful in the method. It further xelates to a method of produc~ng altered amino acid sequences or polypept~des by translating the altered RNA
seq~ence, which results in production of the encoded ~olecule.
In the present method, a selected or target segment of an RNA molecule, such as pre-mRNA, mRNA or viral RNA, is altered as follows: an RNA molecule which includes the target segment (l.e., a nucleotide or a nucleotide sequence to be altered) is combined ~ith an appropri-ately-selected mixed phosphate backbone oligonucleotide in the presence of RNase H. The mixed phosphate backbone oligonucleotide is ;complementary to all or a portion of the RNA ~olecule which includes the target BNA segment;
it is of sufficient length to hybridize to the target RNA
segment and sequences on either side and remain - . ~' ' U'091/12323 PCT/US91/~0968 2076~37 hybridized under the conditions used. The mixed phosphate backbone oli~onucleotide has ~o key components: an internsl portion or segment o f deoxynucleotides which is capable of acei~ating RN~se H
05 and t~o nucleotide cequences, ~hich flank the i~ternal seg~ent, ~h~ch are unable to act~vate RNase H. The internal deoxynucleotide segment includes two or ~ore phosphodieçter linkages, ~hlch ~ay be un~odLfied or modifled. The fianking nucleot~de sequence3 m~y be deo~yribonucleotide or ribonucleotide sequences and is modified. That is, some or all of the internucleoside bridging phosphate residues are modified phosphates, such as methyl phosphonates, phosphoromorpholidates, phosphoropiperzaidates and phosphoramidates. An essential feature of the mi~ed phosphate backbone olig'onucleotide is that the internal complementary segment is RNase H acti~ating and the flanking complementary sequences are unable to activate RNase H.
As demonstrated herein, site-directed alteration occurs ~hen the RNA molecule which includes the target segment and an appropriately selected mixed phosphate backbone oligonucleotide are combined in the presence of RNase H and maintained under appropriate conditions ~e.~., temperaturè, time, salt concentration) for 2~ complementary nucleotide sequences to hybridize and RNase H to be activated (i.e., to be able to clea~e aDd excise). That is, as a result, the nucleotides in the RNA molecule to ~hich tbe internal segment of the mixed phosphate backbone oligonucleotide i5 complementary are excised precisely from the RNA molecule.

.,. , ", . : :. .

, WO9l/1'~1~ Pcr/ US91/00968 2~760~7 , In one embodiment of the present ~ethod, the gap resulting from e~cision of the target RNA ~e~ent can be closed by the sct~it; of an appropriate ligase(s), resulting in a continuous RNA ~olecule refarred to as an ;05 altered RNA oolecule. The resultlng alter~d RNA ~olec~le differs from the RNA molecule only as to the target regment, Yhich ~s not pr~sent in the altered RNA ~ole-cule.
In ~ ~econd embodiment the gap created by the action 10 of RNase H as described above can be f~lled in by inero- -ducing a segment of replacement nucleotides, which can be of any length appropriate to fit into the gap created in the RNA molecule. The se~ent of replacement nucleotides is subsequently linked to the stjacent nucleotides of the RNA molecule by an sppropriPtely selected ligsse(s). The nucleotide present at each end of the segment of replace-ment nucleotides is ligated to the nucleotide present on the respecti~e end~ of the gap created in the RNA
molecule (underlined in Fi~ure 2(111~). The resulting altered RNA molecule tiffers from the RNA ~olecule in that the target segment (present in the RNA ~olecule) has been removed snd a replacement nucleotide cequence introduced in its place.
Altered RNA produced as des~ribed herein can be expressed, either in_vitro or i__~i~o, to produce the encoded polypeptide or protein; ~s used herein, the term polypeptide includes proteins. For example, the altered RNA can be introduced into an appropr~ate ~ector, which is in turn introduced into a host cell capable of trans-lating the alt~red RNA ~olecule and producing the encodedpolypeptide. ~olypeptides produced in this ~anner can be .... ~: .
' ~

WO9t/12323 PCr/US9l/00968 used for assessment of thelr structur~l/functional characteristics and a comparative assess~ent of polypep-tides ~hich d~ffer ln ~ defined ~anner ~e.~., by selected a~ino acids). Such polypeptides c~n also be used 05 therapeutically or prophylactically.
The following is M description, with reference to the fi~ures, of two enbodimenes of the present ~ethod of site-directed alteration of an ~NA molecule: a firfit embodiment in which a selected nucleotide qequence ~target segment) is re~oved and the resulting fragments ligated to produce sn altered RNA molecule lacking the target segment and 8 second embodiment in which a selected nucleotide sequence ~s removed and replaced by a selected nucleot~de or nucleotides, ~hich are ligsted to the nucleotide at each side of the gap created by removal of the target RNA segment.

~ethod for Excisin~ a SPecific Small Se~ment of Rlbo-_________ _ _______ ____ _______________ _____________ nucleotides from an RNA Molecule Figure l ls a schematic representation of sn embodi-ment of the present method.~ The sequence designated I is a segment of an RNA molecule, ~hich includes a t~rget segcent (~GACGUCA) to be altered. The RNA may be a pre-RNA, a viral ~NA, or one of a Yariety of RNAs present in animal, plant or bacterlal cells. The sequence designated II is a se~ment of DNA which is a mixed phosphate backbone oli~odeoxynucleotide complementary to the RNA molecule I. It can be synthesized us~ng known techniques, such as chemical or enzymatic methods. The mixed phosphate backbone oligodeoxynucleotide includes an inte~nal sequence which is capable of activating RNase H
.

.
. . - . .
. , ~ . .

WO 91/12323 PCI/US91/OOg68 g and is flanked on each side by a DNA ~egment which is incapable of activating RNase h, ~he total length of the ~i~ed phosphate backbone oligodeoxynucleotides varies, dopending on the length of the target RNA segment to be 05 gltered, but must be sufficient to hybrid$ze to the RNA
nolecule containing the target RNA segment Hnd remain hybrldiz~d under the cond~tions u~ed. The ~nternal ~egDent ~ust be of sufficient length--et least two nucleotides--to be capable of actl~ating RNase H, ~s de~onstrstet herein and by others (Ualder, ~.Y. snd J.A.
~alder, Proc. Natl. acad. Sci USA, 85:5011-5015 ~1988);
Furdon, P.J. et al., Nucleic Acids_Res., 17:9193 9204 (1989)).
In the internal segment, the internucleoside bridg-lS ing phosphate residues may be unmodified phosphates or any phosphate modific~tion capable of activa~ing RNase ~, such a~ phosphorothioates. The flanking n~cleotide sequences can be deo~yribonucleotides, as descr~bed in this embodiment or can be ribonucleotides and their modifications. The flanking sequences sre connected by methyl phosphonates (PC), phosphoromorpholldates (PM), phosphoropiperaz~dates, phosphoramidates, or other modifications of in~ernucleoside phosphates which are not able to activate RNase H. The RNA molecule I and the ~odified backbone oligodeoxynuclestide II are combinet in the presence of RNase H, such as endogenous RNase H in a cell, and excision of the target segment occurs. A
hairpin loop may form by pairing of complementary nucleo-tides, with the result that the two newly-formed segments are brought into proximity to each oeher. lt ~ay not be necessary that a hairpin loop form, however, in order for '~ ' - ' '~ '.:

, WO 91/1232~ PCI~US91/00968 2076037 -~

the se~ments produced by the activity of RNase H to be brought together becauce the two RNA segments are tether-ed by their base-pairing with the ~xed phosphate backbone oligodeo~ynucleotide. In the presence of an 05 appropriate ligase(s), ~uch as an endogenous ligase(s), the two newly-created cegments are ~oined, by ligation of the nuclcotide (~nterlined ~n Fi~ure 1~ on ~ach ~ide of the gap.
The definition of ~activating RNsse H" ls based on the induced-fit theory of Koshland, in which "enzyme sites Yere envisaged as somewhat flexible and undefined before binding occurred, locking actlve site residues into defined positions around the substrate" (Koshlant, D.E., Jr., Proc. Nael _Acad. Sci. USA, 44:98 (1958);
Zeffren, E. and P.L. Hall, The Study of_Enzyme Mechsn isms, p. 201, John Wiley and Sons, New York, (1973)).

Method_for excisi R_a_d_re~lacin~_a_s~ecific_small se~ment of ribon~cleotides from_an RNA molecule A second e~bodiment of the present invention is represented schenatically ~n Figure 2. This embodiment is useful, as described below, in repairing a genetic defect by re~o~ing the defective nucleotides and replac-in~ them with others, such as those present in the normal RNA or those wh~ch result in RNA cncoding a desired polypept~de. As shown, sn RNA molecule to be altered, such as pre-mRNA or ~RNA in which a tefect is present, is combined with an appropriate mixed phosphate backbone oligodeoxyr~bonucleotide ln which the internal segment is capable of activ~ting RNase H and the flankin~ nucleotide sequences are unable to acti~ate RNase H, in the presence of RNase H. As described above, the target se~ment is e~cised in a site-d~rected msnner, producing a spl$t RNA
molecule, as represented in Figure 2 as sequence lIIa.
In this embodinene, a replacement oligomer, ~hich ls the S scries of nucleotides to be inserted into the gAp created by excision cf the target ~egment, is ~ntroduced into cells. $hrough hybridizstion e~ch-nge (as described ln detail belo~), the r-pl-cement oligoner ~s introduced into the g~p created as a result of the RNase H activity.
E~pression of the resulting altered RNA (which includes the desired/nondefective cequence in place of the defecti~e sequence) results in production of the desired/nondefective polypeptide.
Certa$n genetic tisorders (inborn errors of ~etabol-ism~ can be corrected by the method of the presentin~ention. For example, cystic fibrosis is usually due to a 8ene mutation in which a specific phenylalanine codon is deleted (Riordon, J.R. et al., Science, 245:
1066-1073 (1989)). Through use of the present me~hod, ~he mutant cystic fibrosLs mRNA present ln an indiv~du-al's cells can be cleaved at the missing codon by site-directed RNase H alteration, as described below. Intro-duction into the individual's cells of the appropriate oligoribonucleotide (i.e., one encodin~ a phenylalanine), followed by endo~enous RNA ligase acti~ity results in ligation and, thus, production of a wild-type mRNA
encoding a normsl protein product.
Other genetic tefects can be remedied in a similar manner. Examples of genetic defects for which this would be appropriate are: the substitution of serine for glyc~ne 844 in a severe ~ariant of osteogenesis , . ' ~ : -.

' ' .

Wo 91/12323 PCI/l,'S~1/00968 inperfecta (Pack, ~. et al., J. Biol. Chem., 264:
19694-19699 (1989)); a glycine to serine substitution in pro ~ (II) collagen in a for~ of dwarfism (Vissing, H. et al., J. Blol. Chem., 264:18265-18267 (1989)); ~ glyc~ne _ _ _ 05 833 conversion to aspartate in a mild varlant of Ehlers-Danlos syndro~e IV (Tromp, G. et al., J. Biol.
Che~., 264:19313-19317 (1989)); an ~ to G translt~on in an lnit~tion codon ~utction in the Apo C~ ene Df ~
patient with a deficièncy of apolipoprotein C-II (Fo~o.
S.S. et Al., J Biol Chem., 264:10839-10842 (1989)); and an aberrant GT splice-donor signal flanking exon 19 in retinoblastoma RB-88 (Ysndell, D.W. et al., N.E. J. Med., 321:1689-1695 (1989)). Such precise genet~c tefects can be repaired at the RNA level by the method described.

Correction of the s~ecific defect sssoc~ste_ with cystic fibrosis Riordan et al. ha~e found that in approximately 70%
of the cases of cystic fibrosis, a trinucleotide (TTT) is missing fro~ a seg~ent of the gene of chromosome 7, as compared with its normal, wild-type counterpart, Thus, the ~NA transcription prod~ct of TTT, na~ely VUU, is also missing.
An approach to correcting the defect associated with cystic ibrosis is ~s follows: a mixed phosphate backbone oligodeoxyribonucleotide is synthesized, which consists of an ~nternal segment composed of either ~nmodified bridging phosphodiester bonds, or of modified ones, such as phosphorothioates, (or certain other modific~tlons) Yhich ~re c~pable of activating RNase H.

, .. .
- . . .

.
; ~

2076~37 As an example, Figure 2(I) illustrates 8 section of pre-mRNA from cystic fibrosis in which the UUU trinucleo-tide is ~iSsiDg. The ni~ed phosphate backbone oligamer A-TAG-T~A-CCA-CA~-~ ls cynthesized. It incl~des an 05 internAl se~me~t, bearln~ the subscript ~s~, ~hich ~ncludes the ~nternucleotido phosphates (unmodified, phosphorothioate or other) ~hlch re capable of ~cti~at-lng RNase H. The flanking se~ents have the subscrlpt designation ~m~, ~hich s~gnifies that the internucleoside bridging phosphate oodific~tions render this ~e8ment of DNA incapable of activating RNase H. These ~m~ ~odifica-tions may be ~ethyl phosphonates, phosphoromorpholidates, phosphoropiperazidates, phosphoro-n-butylamidates, or other modifications of the lnternucleoside phosphate which result in inability to ~ctlvate RNase H. In Figure 2(1I) is shown a synthetic oligodeoxynucleotide consist-ing of six internal deoxynucleotides (subscript s) capable of activatin~ RNase H in the portion of RNA to which they are hybridized. These residues are flanked on both sides by oligod~oxynucleotides whose lnternucleoside bridging phosphate modificitions are designated by subscript ~m~, to indicate that these segments of the synthetic oligomer are incapable of activating RNase H.
RNase H acti~ation requires a DNA-RNA hybrid in which at least 2 consecutive nucleotides of the DNA strand have RNase H sensiti~e internucleoside linkages, as demonstrated herein (See Exemplification) and by others.
Walder R.Y. and J.A. Walder, Proc__ atl-~_ca_. Sci. USA~
85:5011-5015 (1988); Furdon, P.~. et al., Nucleic Acids Res., 17:9193 9204 (1989)~. Alternati~ely the sequence designated II can be a central RNase H sensitive section, ~ 091/12323 PCT/~S91/00968 ss described abo~e, flanked by two segments of ribonucleotides (subscript r), one on either side of the internal seg~ent. In this case, the oligomer of Figure 2(II) would be represented ~s follows:
05 ~-VAG-TAA-CCA-CCA-A. Hybridlzed ribonucleotide oligomers do not activate RNsse H; therefore, the sbove oligoocr ~ould also consist of ~Na~e H ~ensitive and reslstant ~egments.
As is described ~above ~lth reference to Figure l, the target segment i5 e~cised ln a site-spec~fic ~anner, resulting in an interrupted RNA ~olecule; in this case, AUC-GGU have been removed,;producin~ a 6-nucleotide gap.
For exa~ple, in an indivitual, the synthetic oligomer (II), dissolved ~n physiological saline solution (e.g., at a concentration of l x lO - l x lO M) is injected (e.g, intravenously) into an individual with cyst~c fibrosis. The oligomer ~ould become distributed lnto various organs and tissues of the individusl. HybrLdiza- ;
tion of the synthetic oligomer with pre-mRNA or mRNA at 20 the targetet site ~ould occur, activation of RNase H
would follow, and cleavage and excision of the six nucleotide segment of RNA w~uld result.
Subsequently, another construct, such as that designated IVa in Figure 2, is administered to the individual in such a manner that it enters cells (e.g., by Intravenous injection). This construct is a hybrid, partially double-stranded synthetic oligomer which consists of a r~bonucleo~ide unmodified nonamer ~ith a 5'-phosphate, hybridized to a 27-~er deoxyribonucleotide, in which all of the residues are modified so as to be nuclease resistant.

......
'' ' , 1 :' -' :' ' ' ' ': '' ' ", 2~76o37 As diagrammed in Figure 2, a hybridization exch~nge occurs, in ~hich the longer, double-stranded oligomer construction IV would replace the shortsr oligomer IIIb (A-IAG-T~A-CCA-CAA-A) in hybridizing ~ith the natursl RNA
05 (IIIa) at.the cr~tical, excised defect~Ye slte shown in Fi~ure 2(IIIa). The latter bears a 5iX nucleotide gap;
the tYo ~egments are held ~n place by the ol~gomer A-~AG-TAA-CCA-CAA-A. Due to the greHter hybridization length of the hybrld shown ln F~gure 2(IVb), a hybritiza-tion exchange occurs and the latter oligomer constructreplaces the A-TAG-TAA-CCA-CAA~A. The nonamer (IVa) fills in the gap left by the nucleotide excision. The complementary pattern of the oligomer construct with the cystic fibrosis R~A ~ill separate its two seg~ent.s sufficiently to permit a nine nucleotide segment to be inserted ~here a six segment nucleotide was removed. The 5' phosphate on this unmod~fied RNA nonamer will permit ATP and RNA ligase, both present endogenously, perhaps assisted by other intracellular factors, to make the two co~alent phosphodiester bonds necessary to repair the excision defect. The f~nal result is to insert a trinucleotide UUU, at the tesired location into the RNA
transcript of the cystic fibrosis patient's genome.
~ybridization exchange occurs because of the greater hybridization association constant and higher Tm of the 18 complementary nucleotides in the oligo~e-r construct as comparet ~ith that of the eight nucleotides in the A-TA~-TAA.-CCA-CAA-A. It is slso possible that a natural RNA repair process may fill in the gap, if the unmodified rebonucleotide nonamer is omitted, and just the oligomer : . .. : .~ . .

W091/l23~3 PCT/US91/00968 2076037 _ TTA-TAG-TAG-AAA-CCA-CAA-ACG-A~A ls used for the transhybridization and repair steps.

node of administration of ~odified backbone olI~onucleo-tide 05 The ~anner in ~hich ~odified backbone ollgonucleo-tides ~re provided will depend on the context ~n ~hlch they are used ~i.e., in vitro, in ~o).
The modified backb~one oligonucleotide is generally dissolved in water or'a suitable buffèred medium, such as Dulbecco's medium, Eagle's medium, or a similar physio-logical saline ~edium, typically at a concentr~tion of 10 5 to 10 8 mol~r. In the case of a tissue cultuxe system, the dissolved oligomer is ster~lized by filtra-tion through a bacterial filter, nnd is added to the other components of the tissue culture incubation medium.
In the case of a seed, the oligomer is d~ssolved in ~ater and added to the seeds spread on sterile filter paper iDside a sterile covered glass or plastlc dish. In the case of a plant, dissolved oligomèr, in aqueous media, is added to the soil or other nutrient material in which the plant is gro~ing. In the case of an animal or man, the oligomer, dissolved ln physiological saline, may be iniected subcutaneously, intraper~toneally, intra-muscularly, intravenously, or possible by capsule orally.
It has been shown that oligomers such as the above-described enter living cells, and are found in signifi-cant concentrations both in the cytoplasm and in the nucleus ~ithin mi~utes after administration (Zamecnik,' P.C. et al., Proc._Natl. Acad._Sci._USA, 83:4143-4146 (1986); Coodchild, J- et_al., C rre_t_Co m__ica ions in , :

.;
- ::

WO 91/123'3 PCI/l!S91/00968 .

Molecular Biolo~v Aneisense RNA and DNA, Cold Spring Harbor, pp. 135 139 (1988~; Wickstrom, E.L. et al., Proc.
Natl._Acad. Sci. USA, 85:1028-1032 (1988)).
The follo~ing exempllfication demonstrates that S site-spec~f$c excision of nucleotides ~t the RNA le~el ha~e been carried out, us~n~ the present ~ethod ~nd ~odified bac~bone oligonuclsotides ~s described herein.

E~EMPLIFICASION

Materials and Methods _____________________ Oli~odeoxynucleotide Synthesis ___ _____ ____________ _______ Oligodeoxynucleotides were synthesized on an auto-matet instrument (model 8700, Milligen, MA~. Nor~al phosphodiester (PO) oligodeoxynucleotides and the analogous phosphorothioate (PS) or phosphoramidate oligodeoxynucleotides were synthesized using H-phos-phonate chemistry (Agrawal, S. et_al., Proc Nael. Acad.
Sci _~SQ, 86:7790-7794 (1989); and Agrawal, S. et al., Proc Natl. Acad. Sci. USA, 85:7079-7083 (1988)).
01igodeoxynucleos~de methylphosphonate (PC) ~nalogues uere a~sembled by using nucleoside methylphosphonamidites (Agrawal. S. snd J. Goodchild, Tetra_edron Lett., 28:
3539-3542 (1987~). Oligodeoxynucleotides containing both PO and PC internucleoside linkages were assembled by using nucleoside-~-cyanoethylphosphoramidites and nucleo-side methylphosphonamidites, and oligodeoxynucleot-ides containing both PC and PS linkages were synthesized from nucleoside methylphosphonamidites (Agrawal, S. and J.
Goodchild, Tetrahe_ro__Lett., 28:3539-3542 (19B7)~ and , WO 91/12323 PC~/l,'S9t/00968 nucleoside H-phosphonates. Oligodeoxynucleotides con-taining both P0 and phosphor~midate linka~es were syn-thes~zed by us~ng nucleoside-B-cyanoethylphosphora~idites and nucleoside H-phosphonates.

05 RNase H Assays __ _ _ The 13-~er oli~odeoxynucl~otides complement ry to nucleotides 2-14 of hunan Ul s~all nuclear RNA ~count~ng the G cap as nucleotide (0)) ~ere stded at 100 ~g/~l to a HeLa cell nuciear extract (Dignam, J.D. et_al., Nucleic Acids_Res., 11:1475 1479 (1983)~ coneaining 0.5 mM ATP, 20 mM crestine phosphate, 3.2 ~M MgC12, ant 1000 units of RNase per ml and ~ere incubated under the cond~tions specified. RNA was isolated from the nucle~r extract by phenol~chloroform extraction ~nd ethanol precipitation, followed by electrophoresis In a 10~ polyacrylamid~ ~et con~aining 8.3 M urea. The RNAs were visualized by ethidiu~ bromide staining.
A second RNase H assay was also carried out in nuclear extracts by using an exogenous P-labeled RNA.
A 5}4-nucleotide test RNA (here~fter ter~ed ~514 RNA~ for convenience) was generated by SP6 RNA polymerase trans-cription of a HindIII-linearized pGEM-2 clone, pT7H~6.
514 RNA is antisense to the first two exons and intron of human ~-globin pre-mRNA ~nd was chosen for the reasons described below. 514 RNA labeled with a ¦~- 2P~GTP, l~-32PlCTP, snd l~-32P]UTP was added to the nuclear extract containing the specified oligodeoxynucleotide at an oligo~er-to-514 RNA ~olar ratio of 3000:1, unless other~ise noted. After incubation as specified, RNA was extracted and the P-labeled 514 RNA or its clea~age :' , ~ : '~

U'091~1232~ PCT/~;S91/~0968 2076~37 -l9-products were visualized by electrophore~is and auto-radiography.

~esules Act~on of RNase H on Oli~odeoxynucleotide Ul Small 05 Nuclear RNA Hybrids HeLa cell ~uclear e~tracts cont~in RNase H sctivity that can ~ct on DNA-RNA hybrids that form after addit-on of oligodeoxynucleotides complementary to rertain endo-genous nuclear RNAs (Krainer, A.R. and T. ~aniatis, Cell, 42:725-736 (1985)). The ability of this RNase H activity __ to cleave the'5'-terminal nucleotides of endo~enous Ul small nuclear RNA, after incubation of the various ~odi-fied oligomers ~n the nuclear extract, uas investigated.
Incubation of the P0 oligomer $n the nuclear extract led to cleavage of a large proportion of the Ul RNA to a product (Ul~) hav~ng the mobility expected for removal of the first 15 nucleotides (cap, nucleotide l, and nucleotides 2-14). There was a lack of effect on thP
mobility of any other RNAs present, demonstrating the high ~equence specificity of the Ul oligomer-directed RNase H cleavage. Ul cleavage ~as also observed with the PS oligomer, although to a consistently lesser extent than with the P0 oligomer. In contrast, no cleavage was obser~ed ~ith the PG, phosphoro~-morpholidate (PM), or phosphoro-N-butylam~dAte (PB) oligomers. Reducing the temperature of ~ncubation to 20-C and extending the time to 60 minutes did ~ot increase the extent of cleavage observed with the PS oligomer, nor did it reveal cleavages with the PC, PM or PB oligomers. The pattern .

:-- . :

WOgl/12323 PCT/US91/00968 of UI cleavages seen with the vario~s oligomers w~s also not aleered by adding E._coli RNase H (Yharmacia, final concentrstion 8 units/ml~ to the nuclear ~xtract at the outset of incubation.

S RNase H Action at an Internal RNA Site Hybridized with __ ___ ____ _________ __________ _____ _______ or~al and Modified Oligodeo~ynucleotides She foregoing Ul RNA cloavsges lnvolve the 5' estFe~ity of a s~all RNA. Indeed, the 5' end of Ul RNA
is sn exceptionally fsvored target for ollgodeoxy-Ducleotide-directed RN~se H cleavage aince al~ost all the other regions of this RNA molecule are tightly complexed vith proteins in the Ul small nuclear ribonucleoprotein particle (Patton, J.R. et al., Mol. Cell. Biol., 7:
4030-6037 (1987); Patton, J.R. et al., Proc. Natl. Acad.
lS Sci. USA, 85:747-751 (1988); Patton, J.R. et al., Mol.
Cell. Biol., 9:3360-3368 (1989)). Therefore, the action of RNase H on hybrids formed by normal or modified oligomers at internal sites in a longer RNA was also in-~estigated.
For this purpose a test RNA which is 514 nucleotides long (termed 514 RNA) ~as uged. 514 RNA is antisense to the first t~o exons and intron of human ~-glo~in pre-mRNA. The underlying reasoning was that, because it is antisense to a pre-mRNA, 514 RNA ~ould not undergo splicing when ~dded to nuciear extract (the cleava~e-ligation steps of whlch ~ould otherwise complicate analysis of oligodeoxynucleotide-directed RNase H clea~-ages).
A series of normal and modified lS-~er oligodeoxy-nucleotides complementary to nucleotides 349-363 of 514 ': . . , ' `'i:
. .

WO91~12323 PCT/US91/00968 RNA vas synthesized chemically (Table 1, oligomers A-E).
This particular 514 RNA site ~as selected because oligomer-directed RNase H cleavage would generate two frdgments of readlly distinguishable lengths (lS0 and 348 05 nucleotides) ~nd also because this 15-nucleotide ~equence does ~ot occur else~here in 514 RNA (Lawn, R.M. et al., Cell, 21:647-651 (1980)). ~elting curves ln 0.16 M Na f~r four of these oli~odeo~yDucleotides ~fter duplex formstion with the co~plementnry (P0) oligomer reve~led that the P0, PC, PS and P~ oligomer-containing duplexes had tms of 53-C, 46-C, 43-C and 38-C, respectively, indicating a lower duplex ~tability for the modified oligomers, in part confir~lng previous reports (Stein, C.A. et al., Nucleic_Acids Res., 16:3209 3221 (1988);
lS Froehler, B. et_al., Nucleic_Ac~ds Res., 16:4831 4839 (1988); Quartin, R.S. ~nd J.G. Wetmur, Biochemistry, 2B:1040-1047 (1989); Agrawal, S. et_al., Nucleosides Nucleot~_es, 8:819-823 (1989)), . , . :
:: . .
. :- - . . :
, .

, ~'091/12323 PCT/US9l/0096~

2~7 ~ ~ I 22-___ ___________ _____ ___ ___ _ ___ __ _____ _ __ _ .
Ineernucleos~de Oli~omer Sequence Linka~e ___ __ _ _ _ __ ____ _ _ _ _ __ __ _ _ _ AGTA TCA ~GG TTA CAA PO
05 ~ GIA TCA AGG TTA CAA PS
CGTA TCA AGG TTA CAA PM
DGTA TCA ~GG TTA CAA PB
EGTA TCA AGG TTA CAA PC
F*GTA TCA TAT GAG ACA PO
G* GTA GCA AGG CTA CAA PO
H*GTA TGA GAC ATA TAC PO
__ _ _____ ___________________ ________________ Underlined nucle~tides indicate base pairing mismatches; PB is pbospho-N-butylamidate.
The beries of normal snd modified ol$godeoxy-nucleotides shown in Table 1 were incubated with P-labeled 514 RNA in nuclear extracts, under the conditions specified tn the ~aterisls and ~ethods section, for either 30 minutes or 3.5 hours.
The results of incubating these oligomers with 514 RNA in HeLa nuclear extract ~re as follows: After 30 minutes of incubation without any oligomer, intact input 514 RNA was the only labeled ~pecies v~sualized. After 3.5 hours of incubation without any oligomer, the 514 RNA
~as completely degraded by the action of ribonuclease kno~n to be present ~n the extract. Incubation of 514 RNA in the extract for 30 minutes together ~ith either the PO or PS oligomers resulted in precise cleavage of the substrate RNA into two fragments of the sizes . .

, ... . ~ ,.
., , . ~ , . .
:

W~91/123'~ PCT/~IS91/0096~

2~76037 23- , expected fro~ ehe location of the oligomer-complementary sequence. Surprisingly, in the case of the PS oligomer, these two fragments vere still present, albeit ln sli~htly degrnded form, after 3.5 hours of incubatlon.
05 Incubation of 514 RNA with PM ol~omer for 30 minutes resulted in partial cleavage (see the figure legend for additional details). No cleavage was observed with the PB or PC oligomers under these conditions.
PO oligomers that vere only pareially complementary eo 514 RNA (oligomers F, G and H ~n Table 1) were also tested. None of these oligomers, containing 4, 5 or 6 uninterrupted complementary nucleotides out of the 15 (Table 1), elicited RNase H cleavage of 514 RNA.
The finding that the PS oligomer was less effective than the P0 oligomer in eliciting RNase H cleavage in the Ul RNA assay raised the possibility that the more complete RNase H cleavages observed with both P0 and PS
oligomers in the S14 RNA assay might reflect the particular reaction conditions employed. Therefore, a ran~e of oligomer-to-514 RNA molar ratios tests (0.1:1-1000:1), all below that used in the above-described (3000:1), was investigated. Results showed that virtually complete 514 RNA cleavage occurred with the PO oligomer at an oligomer-to-~NA ratio of lO0:1, whereas a comparabl~ extent of 514 cleavage with the PS oli~omer occurred at a oligomer-to-RNA ratio of 1000:1. A very similar, ~ncomplete extent of cleavage was obser~ed with the P0 and PS oligomers at ratios of 10:1 and 100:1, respectively. The possibility that these results might reflect a preferential instability of the PS oligomer turing incubation in ~he nuclear extracts was ', .

WO91~1232~ PCT/US91/00968 2076~'~7 examined by experiments in which either the P0 or the PS
oligomer ~as preincubated in nuclear extract or 30 minutes, followed by addition of P-labeled 514 RNA and incubation for an atditional 30 min. This revealed the 05 same e~tent of difference in RNase H cleavage as described above.

~Restriction Endonuclease.Like~ Cleava~e with Oli~omers Containin~ RNase H Sensitive and -Resistant Internucleo-s~de_Linka~es-The extreme differences be~ween the R~ase H sensi-tivity of DNA-RNA hybrids containing PO or PS oligodeoxy-nucleotides, contrasted with ones with PC, PM or PB
oligomers, led to an investlgat~on of how RNase H acts on a DNA-RNA hybrid in which only a small proportion of lS internucleoside linkages in the DNA strand were RNase H-sensitive. Table 2 shows the series of oligomers that were synthesized to address this issue,.

WO91t12323 PCT/US91/00968 2076~37 _________ ____ _ _____ ___ ______ Oli~omer Sequence __ ___ _ _ _ ____________ AGG T
J A AGG TT

L GTA ICA ~GG TTA CAA
~ GTA TCA AGG TTA CAA
N GTA TCA AGG TTA CAA
O GTA TCA AGG TTA CAA
p GTA TCA AGG TTA CAA
___ _ __ Q GTA TCA AGG TTA CAA
R GTA TCA AGG TTA CAA
__ _____________ _ _ _ _ ____ _ _ Vnderlined nucleotides are PC; dashed nucleotides are PS; Boxed nucleotides are phosphoromorpholidates;
lS Double-underlined nucleotites are PM (oligomer Q) or PB;
the remaining nucleotides are PO.
The oligomers listed yere tested for their capacity to elicit RNase H action after hybridization to 514 RNA, as in the preced1ng experiments.
20 Results demonstrated that neither a tetramer nor a hexa~er (all PO) oligodeoxynucleoeide co~plementary to ~14 R~A was able to induce RNase H cleavage in this nuclear extract system. ~hen PO/PC-containing penta-decamers containing either two or four coDsecutive PO
linkages were used, a ~ow but readily detectable level of RNase H cleavage occurred. In contrast, a pentadecamer containing six consecutive PO linkages elicited complete RNase H cleavage of the substrate RNA. Note thst the six WO91/123~3 PCT/~'S91/00968 207~37 PO nucleotides in this oligomer (oligomer M in Table 2) are identical in sequence to the RNase H-inactive hexamer (oligomer J), from which it is inferred that the potency of the pentadec~mer reflects its increased hybrid ~5 stability with 514 RNA owing to the additional nine complenentary nucleotides.
Similar tests ~ere performed ~ith mixed PO~PC
pent-dec-mers co~pl~mentsry to a different site ~n 514 RNA (i,e,, nucleotides 463-477). These tests revealed an effect of the number of PO linksges on RNase H cleavage similar to that described above.
Additional variants of mixed PO/PC oligomers were also tested. Pentadecamer PO/PC oligomers with five or six consecutive PO linkages at either the extreme 5' or 3' end were highly effective in eliciting RNase H cleav-age, A PS/PC pentadecamer with six consecutive PS
linkages at the extreme 5' end ~oligomer P in Table 2) was only partially active. Comparison of these results with lanes 2 and 3 in Figure 4B confirms the above-described sesults showing that all-PS oligomers are less effective than all-P0 oligomers in eliciting RNase H
cleavage. RNase H cleavage~of 514 RNA was also observed with a 15-mer containing nine consecutive PM or PB
linkages followed by six PO-linked nucleotides (oli~omers Q and R in Table 2).

. . .

' ' ' : . '"' ~ ~ . ,. `
.: '' ' ' . , , ~ ' :
... . .. . .

Claims (12)

1. A method of site-specific alteration of a target RNA
segment of an RNA molecule, comprising the steps of:
a) combining the RNA molecule with a fixed phosphate backbone oligonucleotide which is complementary to all or a portion of the RNA
molecule, in the presence of RNase H, the mixed phosphate backbone oligonucleotide including an internal segment which activates RNase H and two flanking nucleotide sequences which are unable to activate RNase H, one of the two flanking nucleotide sequences being present on either side of the internal segment, under conditions appropriate for hybridization of complementary nucleotide sequences and activation of RNase H, thereby excising the target RNA segment, creating a gap in the RNA
molecule and producing an interrupted RNA
molecule which has two segments; and b) contacting the interrupted RNA molecule with an appropriate ligase, under conditions appropri-ate for joining the two segments of the interrupted RNA molecule by the ligase, thereby producing an altered RNA molecule.
2. The method of Claim 1 wherein the mixed phosphate backbone oligonucleotide is a mixed phosphate backbone oligodeoxynucleotide; the internucleoside bridging phosphate residues of the internal segment are unmodified phosphates and the internucleoside bridging phosphate residues of the flanking sequences are modified phosphate residues.
3. The method of Claim 2 wherein the bridging phosphate residues of the flanking sequences are modified phosphates selected from the group consisting of:
methyl phosphonates, phosphoromorpholidates, phosphoropiperazidates, and phosphoramidates.
4. The method of Claim 1 wherein the mixed phosphate backbone oligonucleotide is a mixed phosphate backbone oligodeoxynucleotide; the internucleoside bridging phosphate residues of the internal segment are modified phosphates and the internucleoside bridging phosphate residues of the flanking sequences are modified phosphates.
5. The method of Claim 4 wherein the internucleoside bridging phosphate residues of the internal segment are modified phosphates which are phosphorothioates and the internucleoside bridging phosphate residues of the flanking sequences are modified phosphates selected from the group consisting of: methyl phosphonates, phosphoromorpholidates, phosphoro-piperazidates, and phosphoramidates.
6. A method of producing a selected polypeptide com-prising expressing an altered RNA molecule produced by the method of Claim 1.
7. A method of producing a selected polypeptide encoded by an altered RNA molecule, comprising the steps of:
a) providing an altered RNA molecule produced by a method of site-specific alteration of a target RNA segment of an RNA molecule; and b) expressing the altered RNA modlecule provided in step a).
8. A method of site-specific alteration of Claim 1, further comprising replacing the target RNA segment excised in step a) by introducing a segment of replacement nucleotides into the gap in the RNA
molecule and b) linking the nucleotide present at each end of the segment of replacement nucleotides to the respective nucleotide present at the gap created in the RNA molecule.
9. A mixed phosphate backbone oligonucleotide useful in the method of Claim 1.
10. A mixed phosphate backbone oligonucleotide consisting essentially of an internal segment of deoxynucleotides which activates RNase H and two modified nucleotide sequences which do not activate RNase H, in which the two modified nucleotide sequences flank the internal segment, one on each side of the internal segment.
11. The mixed phosphate backbone oligonucleotide of Claim 10 wherein the internucleoside bridging phosphate residues of the internal segment are unmodified phosphates and the internucleoside bridging phosphate residues of the two modified nucleotide sequences are modified phosphates selected from the group consisting of: methyl phosphonates, phosphoromorpholidates, phosphoropiperazitates, and phosphoramidates.
12. The mixed phosphate backbone oligonucleotide of Claim 10 wherein the internucleoside bridging phosphate residues of the internal segment are modified phosphates which are phosphorothioates and the internucleoside bridging phosphate residues of the two modified nucleotide sequences are modified phosphates selected from the group consisting of:
methyl phosphonates, phosphoromorpholidates, phos-phoropiperazidates, and phosphoramidates.
CA002076037A 1990-02-15 1991-02-13 Method of site-specific alteration of rna and production of encoded polypeptides Abandoned CA2076037A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US480,269 1990-02-15
US07/480,269 US5149797A (en) 1990-02-15 1990-02-15 Method of site-specific alteration of rna and production of encoded polypeptides

Publications (1)

Publication Number Publication Date
CA2076037A1 true CA2076037A1 (en) 1991-08-16

Family

ID=23907315

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002076037A Abandoned CA2076037A1 (en) 1990-02-15 1991-02-13 Method of site-specific alteration of rna and production of encoded polypeptides

Country Status (10)

Country Link
US (3) US5149797A (en)
EP (1) EP0515511B1 (en)
JP (1) JPH05506355A (en)
AT (1) ATE141326T1 (en)
CA (1) CA2076037A1 (en)
DE (1) DE69121391T2 (en)
DK (1) DK0515511T3 (en)
ES (1) ES2090314T3 (en)
GR (1) GR3021278T3 (en)
WO (1) WO1991012323A1 (en)

Families Citing this family (552)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6399754B1 (en) * 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
US5955589A (en) * 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
US5623065A (en) * 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5149797A (en) * 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
US5965722A (en) 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
WO1994008003A1 (en) * 1991-06-14 1994-04-14 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
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
US20060270624A1 (en) * 1991-12-24 2006-11-30 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
AU669353B2 (en) * 1991-12-24 1996-06-06 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US6277603B1 (en) 1991-12-24 2001-08-21 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5700922A (en) * 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5856455A (en) * 1991-12-24 1999-01-05 Isis Pharmaceuticals, Inc. Gapped 2'-modified oligonucleotides
US6346614B1 (en) 1992-07-23 2002-02-12 Hybridon, Inc. Hybrid oligonucleotide phosphorothioates
US5652355A (en) * 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
TW244371B (en) * 1992-07-23 1995-04-01 Tri Clover Inc
US5523389A (en) * 1992-09-29 1996-06-04 Isis Pharmaceuticals, Inc. Inhibitors of human immunodeficiency virus
CA2150903A1 (en) * 1992-12-04 1994-06-23 American Home Products Corporation Compounds for the treatment of leukemias
NZ263985A (en) * 1993-03-31 1997-07-27 Hybridon Inc Oligonucleotide complementary to an essential amino acid sequence of influenza virus
ES2290951T3 (en) * 1993-05-11 2008-02-16 The University Of North Carolina At Chapel Hill OLIGONUCLEOTID ANTISENTIDO THAT COMBAT THE CUTTING AND OPENING PUMP AND METHODS OF THE SAME USE.
US6410322B1 (en) * 1993-07-27 2002-06-25 Hybridon Inc Antisense oligonucleotide inhibition of vascular endothelial growth factor expression
US6605708B1 (en) 1993-07-28 2003-08-12 Hybridon, Inc. Building blocks with carbamate internucleoside linkages and oligonucleotides derived therefrom
DE69433036T2 (en) 1993-09-03 2004-05-27 Isis Pharmaceuticals, Inc., Carlsbad AMINODERIVATIZED NUCLEOSIDES AND OLIGONUCLEOSIDES
AU7845194A (en) * 1993-09-28 1995-04-18 General Hospital Corporation, The Using antisense oligonucleotides to modulate nerve growth and to reverse beta/A4 amyloid-induced morphology
CA2176259A1 (en) * 1993-11-16 1995-05-26 Lyle J. Arnold, Jr. Chimeric oligonucleoside compounds
AU8102694A (en) * 1993-11-17 1995-06-06 Id Biomedical Corporation Cycling probe cleavage detection of nucleic acid sequences
US5550047A (en) * 1994-02-18 1996-08-27 University Of Massachusetts Oligonucleotides with anti-Epstein-Barr virus activity
US5801235A (en) * 1994-05-25 1998-09-01 Hybridon, Inc. Oligonucleotides with anti-cytomegalovirus activity
US5691316A (en) * 1994-06-01 1997-11-25 Hybridon, Inc. Cyclodextrin cellular delivery system for oligonucleotides
US6645943B1 (en) 1994-10-25 2003-11-11 Hybridon, Inc. Method of down-regulating gene expression
US6608035B1 (en) * 1994-10-25 2003-08-19 Hybridon, Inc. Method of down-regulating gene expression
US5591721A (en) * 1994-10-25 1997-01-07 Hybridon, Inc. Method of down-regulating gene expression
US5750341A (en) 1995-04-17 1998-05-12 Lynx Therapeutics, Inc. DNA sequencing by parallel oligonucleotide extensions
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US20030055240A1 (en) * 1995-06-06 2003-03-20 Roberts Peter C. HPV specific oligonucleotides
US6509149B2 (en) * 1995-06-06 2003-01-21 Hybridon, Inc. HPV-specific oligonucleotides
US7074768B2 (en) 1995-08-17 2006-07-11 Idera Pharmaceuticals, Inc. Modified protein kinase A-specific oligonucleotides and methods of their use
US6624293B1 (en) 1995-08-17 2003-09-23 Hybridon, Inc. Modified protein kinase A-specific oligonucleotides and methods of their use
US5652356A (en) * 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
CN1214688A (en) 1996-02-14 1999-04-21 伊希斯药物有限公司 Sugar-modified gapped oligonucleotides
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US20040266706A1 (en) * 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
US9096636B2 (en) * 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US20030044941A1 (en) 1996-06-06 2003-03-06 Crooke Stanley T. Human RNase III and compositions and uses thereof
US20040161777A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Modified oligonucleotides for use in RNA interference
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US5856462A (en) * 1996-09-10 1999-01-05 Hybridon Incorporated Oligonucleotides having modified CpG dinucleosides
US5886165A (en) * 1996-09-24 1999-03-23 Hybridon, Inc. Mixed backbone antisense oligonucleotides containing 2'-5'-ribonucleotide- and 3'-5'-deoxyribonucleotides segments
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
CA2294988C (en) 1997-07-01 2015-11-24 Isis Pharmaceuticals Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US6013786A (en) * 1997-08-22 2000-01-11 Hybridon, Inc. MDM2-specific antisense oligonucleotides
US20030036516A1 (en) * 1997-09-10 2003-02-20 Sudhir Agrawal Method for using oligonucleotides having modified cpg dinucleotides
US6306831B1 (en) 1997-09-12 2001-10-23 Qik Technologies, Inc. Transplacental delivery of oligonucleotides
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
JP2002515514A (en) 1998-05-21 2002-05-28 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 local delivery of oligonucleotides
AU745880B2 (en) 1998-05-21 2002-04-11 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US5962672A (en) * 1998-09-18 1999-10-05 Isis Pharmaceuticals Inc. Antisense modulation of RhoB expression
BR9914465A (en) * 1998-09-29 2001-10-09 Gamida Cell Ltd Methods to control the proliferation and differentiation of stem cells and progenitor cells and a pharmaceutical composition to induce differentiation in a cell population
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
US5959097A (en) * 1998-11-20 1999-09-28 Isis Pharmaceuticals, Inc. Antisense modulation of MEK2 expression
US5962673A (en) * 1998-11-20 1999-10-05 Isis Pharmaceuticals Inc. Antisense modulation of inhibitor-kappa B kinase-alpha expression
US5977341A (en) * 1998-11-20 1999-11-02 Isis Pharmaceuticals Inc. Antisense modulation of inhibitor-kappa B kinase-beta expression
US5981732A (en) * 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6127124A (en) * 1999-01-20 2000-10-03 Isis Pharmaceuticals, Inc. Fluorescence based nuclease assay
JP2002539833A (en) 1999-03-31 2002-11-26 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル Isolated DNAs encoding kalin regulators ROC1 and ROC2, isolated proteins encoded by them, and uses thereof
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US6046321A (en) * 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6013788A (en) * 1999-04-09 2000-01-11 Isis Pharmaceuticals Inc. Antisense modulation of Smad3 expression
US20030078216A1 (en) * 1999-05-03 2003-04-24 Macleod A. Robert Inhibition of histone deactylase
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US7101989B1 (en) 1999-07-09 2006-09-05 University Of North Carolina At Chapel Hill DsrA protein and polynucleotides encoding the same
US6677445B1 (en) 1999-08-27 2004-01-13 Chiron Corporation Chimeric antisense oligonucleotides and cell transfecting formulations thereof
US20010011126A1 (en) * 1999-09-10 2001-08-02 Bock Elisabeth Marianne Neurogenic compositions and methods
US7332275B2 (en) 1999-10-13 2008-02-19 Sequenom, Inc. Methods for detecting methylated nucleotides
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
EP1438404A2 (en) 2000-03-24 2004-07-21 Methylgene, Inc. Inhibition of specific histone deacetylase isoforms
WO2001074346A2 (en) * 2000-04-03 2001-10-11 Hybridon, Inc. Sensitization of cells to cytotoxic agents using oligonucleotides directed to nucleotide excision repair or transcritpion coupled repair genes
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
AU2001275530A1 (en) * 2000-06-08 2001-12-17 Board Of Regents, The University Of Texas System Inhibitors of c-reactive protein induced inflammation
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
US6596490B2 (en) * 2000-07-14 2003-07-22 Applied Gene Technologies, Inc. Nucleic acid hairpin probes and uses thereof
US6380377B1 (en) 2000-07-14 2002-04-30 Applied Gene Technologies, Inc. Nucleic acid hairpin probes and uses thereof
US20030152557A1 (en) * 2001-01-12 2003-08-14 Besterman Jeffrey M. Methods for inhibiting histone deacetylase-4
US20030148970A1 (en) * 2001-01-12 2003-08-07 Besterman Jeffrey M. Methods for specifically inhibiting histone deacetylase-4
WO2002059337A1 (en) * 2001-01-26 2002-08-01 Georgetown University School Of Medicine Anti-apoptopic gene scc-s2 and diagnostic and therapeutic uses thereof
AU2002258728A1 (en) 2001-04-06 2002-10-21 Georgetown University Gene brcc-3 and diagnostic and therapeutic uses thereof
WO2002081639A2 (en) * 2001-04-06 2002-10-17 Georgetown University Gene brcc2 and diagnostic and therapeutic uses thereof
AU2002305151A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene scc-112 and diagnostic and therapeutic uses thereof
AU2002303262A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene shinc-1 and diagnostic and therapeutic uses thereof
GB2392911A (en) * 2001-05-11 2004-03-17 Methylgene Inc Inhibitors of dna methyltransferase isoforms
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
ATE522623T1 (en) 2001-06-20 2011-09-15 Genentech Inc COMPOSITIONS AND METHODS FOR THE TREATMENT AND DIAGNOSIS OF LUNG TUMORS
EP1404698A4 (en) 2001-06-21 2004-12-22 Isis Pharmaceuticals Inc Antisense modulation of superoxide dismutase 1, soluble expression
SE0102327D0 (en) 2001-06-28 2001-06-28 Active Biotech Ab A novel engineered superantigen for human therapy
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
US20040266718A1 (en) * 2001-08-06 2004-12-30 Zuomei Li Inhibition of specific histone deacetylase isoforms
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US7868204B2 (en) * 2001-09-14 2011-01-11 Methylgene Inc. Inhibitors of histone deacetylase
KR20040048411A (en) * 2001-09-14 2004-06-09 메틸진, 인크. Inhibitors of histone deacetylase
US6897220B2 (en) * 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
EP2153843B1 (en) 2001-09-18 2012-08-15 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
NZ585001A (en) 2001-10-09 2011-08-26 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expression
EP1488233A4 (en) 2001-10-09 2006-06-21 Genentech Inc Novel acidic mammalian proteins and polynucleotides encoding the same
WO2005049075A2 (en) 2003-11-17 2005-06-02 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
US7276489B2 (en) * 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
JP2005525095A (en) 2002-01-02 2005-08-25 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
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
EP1465982A4 (en) * 2002-01-25 2006-06-07 Gamida Cell Ltd Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US7244565B2 (en) * 2002-04-10 2007-07-17 Georgetown University Gene shinc-3 and diagnostic and therapeutic uses thereof
US7138512B2 (en) * 2002-04-10 2006-11-21 Georgetown University Gene SHINC-2 and diagnostic and therapeutic uses thereof
EP2011886A3 (en) 2002-04-16 2009-02-11 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7432342B2 (en) 2002-05-03 2008-10-07 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof and related documents
US20030228317A1 (en) * 2002-05-22 2003-12-11 Prafulla Gokhale Gene BRCC-1 and diagnostic and therapeutic uses thereof
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
US20040072770A1 (en) * 2002-07-03 2004-04-15 Besterman Jeffrey M. Methods for specifically inhibiting histone deacetylase-7 and 8
US20040009483A1 (en) * 2002-07-12 2004-01-15 Ilsley Diane D. Method of linear mRNA amplification using total RNA
EP2269619A1 (en) 2002-08-12 2011-01-05 Jennerex Biotherapeutics ULC Methods and compositions concerning poxviruses and cancer
WO2004016586A2 (en) * 2002-08-16 2004-02-26 Board Of Regents The University Of Texas System Compositions and methods related to flavivirus envelope protein domain iii antigens
EP1537208A1 (en) * 2002-09-13 2005-06-08 Replicor, Inc. Non-sequence complementary antiviral oligonucleotides
US20050196382A1 (en) * 2002-09-13 2005-09-08 Replicor, Inc. Antiviral oligonucleotides targeting viral families
ATE497778T1 (en) 2002-09-16 2011-02-15 Agennix Inc LACTOFERRIN COMPOSITIONS AND METHODS FOR TREATING DIABETIC ULCUS
US7229976B2 (en) 2002-09-26 2007-06-12 Isis Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
CA2499880A1 (en) * 2002-09-27 2004-04-08 University Of North Carolina At Chapel Hill Methods and compositions for modification of splicing of pre-mrna
WO2004035525A1 (en) * 2002-10-17 2004-04-29 Methylgene Inc. Inhibitors of histone deacetylase
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
WO2004044139A2 (en) 2002-11-05 2004-05-27 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
AU2003294281B2 (en) 2002-11-13 2010-05-20 Kastle Therapeutics, Llc Antisense modulation of apolipoprotein B expression
SI1569695T1 (en) 2002-11-13 2013-08-30 Genzyme Corporation Antisense modulation of apolipoprotein b expression
CA2506127C (en) 2002-11-15 2013-07-09 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
EP1601789A4 (en) * 2003-01-16 2007-10-31 Idera Pharmaceuticals Inc Modulation of immunostimulatory properties of oligonucleotide-based compounds by utilizing modified immunostimulatory dinucleotides
US7354907B2 (en) * 2003-02-07 2008-04-08 Idera Pharmaceuticals, Inc. Short immunomodulatory oligonucleotides
WO2004072284A1 (en) 2003-02-11 2004-08-26 Antisense Therapeutics Ltd Modulation of insulin like growth factor i receptor expression
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
ATE479752T1 (en) 2003-03-07 2010-09-15 Alnylam Pharmaceuticals Inc THERAPEUTIC COMPOSITIONS
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US20050118154A1 (en) 2003-04-02 2005-06-02 Mien-Chie Hung Antitumor effect of mutant Bik
WO2004091515A2 (en) 2003-04-09 2004-10-28 Alnylam Pharmaceuticals, Inc. iRNA CONJUGATES
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
AU2004232964B2 (en) 2003-04-17 2011-09-22 Alnylam Pharmaceuticals, Inc. Protected monomers
EP2660322A3 (en) 2003-04-17 2013-11-13 Alnylam Pharmaceuticals Inc. Modified iRNA agents
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
MXPA05012421A (en) * 2003-05-16 2006-02-22 Hybridon Inc Synergistic treatment of cancer using immunomers in conjunction with chemotherapeutic agents.
BRPI0410886A (en) 2003-06-03 2006-07-04 Isis Pharmaceuticals Inc double stranded compound, pharmaceutical composition, pharmaceutically acceptable salt, methods of modifying human survivin-encoding nucleic acid, inhibiting suvivin expression in cells or tissues, and treating a condition associated with suvivin expression or overexpression, and single stranded RNA oligonucleotide
ES2569927T3 (en) 2003-06-11 2016-05-13 Idera Pharmaceuticals, Inc. Stabilized immunomodulatory oligonucleotides
US7790691B2 (en) * 2003-06-20 2010-09-07 Isis Pharmaceuticals, Inc. Double stranded compositions comprising a 3′-endo modified strand for use in gene modulation
WO2005009355A2 (en) 2003-07-15 2005-02-03 Hybridon, Inc. Synergistic stimulation of the immune system using immunostimulatory oligonucleotides and/or immunomer compounds in conjunction with cytokines and/or chemotherapeutic agents or radiation therapy
EP1648914A4 (en) 2003-07-31 2009-12-16 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
EP1677822B1 (en) 2003-09-18 2014-04-23 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
KR100750788B1 (en) 2003-09-18 2007-08-20 아이시스 파마수티컬즈 인코포레이티드 4 MODULATION OF eIF4E EXPRESSION
AU2004276337B2 (en) * 2003-09-24 2009-11-12 Methylgene Inc. Inhibitors of histone deacetylase
WO2005038013A1 (en) * 2003-10-07 2005-04-28 Isis Pharmaceuticals, Inc. Artisense oligonucleotides optimized for kidney targeting
DK1678194T3 (en) 2003-10-10 2013-10-07 Alchemia Oncology Pty Ltd MODULATING SYNTHESIS AND DEGRADING HYALURONANE IN THE TREATMENT OF DISEASE
EP1689888A2 (en) 2003-10-23 2006-08-16 Illumigen Biosciences Inc. Detection of mutations in a gene associated with resistance to viral infection, oas1
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
US20050120397A1 (en) * 2003-11-24 2005-06-02 Hermann Steller Compounds and methods for regulation of spermatid differentiation
CN101094594B (en) 2003-12-08 2012-08-15 海布里顿公司 Modulation of immunostimulatory properties by small oligonucleotide-based compounds
US7695932B2 (en) * 2003-12-24 2010-04-13 Massachusetts Institute Of Technology Gene targets for enhanced carotenoid production
US7741070B2 (en) * 2003-12-24 2010-06-22 Massachusetts Institute Of Technology Gene targets for enhanced carotenoid production
US7608699B2 (en) * 2003-12-29 2009-10-27 Rockefeller University Synthetic nuclear localization signal derived from lentiviral integrase and methods of use thereof
EP2363480A3 (en) 2004-01-20 2015-10-07 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
AU2005216075B2 (en) * 2004-02-20 2011-03-10 Idera Pharmaceuticals, Inc. Potent mucosal immune response induced by modified immunomodulatory oligonucleotides
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
WO2005089268A2 (en) 2004-03-15 2005-09-29 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of rna by rnase h
US7253204B2 (en) * 2004-03-26 2007-08-07 Methylgene Inc. Inhibitors of histone deacetylase
US8314226B2 (en) 2004-03-29 2012-11-20 The General Hospital Corporation Oligonucleotide complex compositions and methods of use as gene alteration tools
US20050267300A1 (en) 2004-04-05 2005-12-01 Muthiah Manoharan Processes and reagents for oligonucleotide synthesis and purification
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
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
US7498425B2 (en) * 2004-06-15 2009-03-03 Idera Pharmaceuticals, Inc. Immunostimulatory oligonucleotide multimers
CA2570786C (en) * 2004-06-15 2013-05-28 Idera Pharmaceuticals, Inc. Modified cpg-containing oligonucleotide multimers in immune stimulation
US7785843B2 (en) * 2004-06-23 2010-08-31 Sequenom, Inc. Target-specific compomers and methods of use
EP3808845A1 (en) 2004-06-28 2021-04-21 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
USRE48960E1 (en) 2004-06-28 2022-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8680062B2 (en) 2004-07-06 2014-03-25 Deliversir Ltd. System for delivering therapeutic agents into living cells and cells nuclei
AU2005272912A1 (en) 2004-08-10 2006-02-23 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP1791567B1 (en) 2004-08-10 2015-07-29 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
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
JP2008513507A (en) 2004-09-17 2008-05-01 アイシス ファーマシューティカルズ インコーポレイティッド Enhanced antisense oligonucleotides
WO2006086821A1 (en) 2004-10-20 2006-08-24 Antisense Therapeutics Ltd ANTISENS MODULATION OF INTEGRIN α4 EXPRESSION
US8003619B2 (en) * 2004-12-09 2011-08-23 Alnylam Pharmaceuticals, Inc. Method of stimulating an immune response and inhibiting expression of a gene using an oligonucleotide
CN101175769A (en) 2005-03-10 2008-05-07 健泰科生物技术公司 Methods and compositions for modulating vascular integrity
WO2006102760A1 (en) * 2005-04-01 2006-10-05 Methylgene Inc. Inhibitors of histone deacetylase
CA2606270A1 (en) * 2005-04-19 2006-10-26 Massachusetts Institute Of Technology Amphiphilic polymers and methods of use thereof
JP5329949B2 (en) 2005-05-31 2013-10-30 エコーレ ポリテクニーク フェデラーレ デ ローザンヌ Triblock copolymers for cytoplasmic delivery of gene-based drugs
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
ES2450593T3 (en) 2005-07-01 2014-03-25 Index Pharmaceuticals Ab Immunostimulant method
PL2179737T3 (en) 2005-07-01 2014-01-31 Index Pharmaceuticals Ab Modulating responsiveness to steroids
US20070026012A1 (en) 2005-08-01 2007-02-01 Cornell Research Foundation, Inc. Compositions and methods for monitoring and altering protein folding and solubility
US8980246B2 (en) 2005-09-07 2015-03-17 Sillajen Biotherapeutics, Inc. Oncolytic vaccinia virus cancer therapy
JP2009507853A (en) * 2005-09-07 2009-02-26 ジェンネレックス インコーポレイティッド Systemic treatment of metastatic and / or systemic disseminated cancer using GM-CSF expressing poxvirus
CA2625969A1 (en) * 2005-10-28 2007-05-03 Index Pharmaceuticals Ab Composition and method for the prevention, treatment and/or alleviation of an inflammatory disease
US7470674B2 (en) * 2005-11-07 2008-12-30 Idera Pharmaceuticals, Inc. Immunostimulatory properties of oligonucleotide-based compounds comprising modified immunostimulatory dinucleotides
CA2628306C (en) 2005-11-07 2014-06-17 Sudhir Agrawal Immunostimulatory oligonucleotide-based compounds with glycerol-linked dinucleotides and modified guanosines at cpg and uses thereof
US7776834B2 (en) * 2005-11-07 2010-08-17 Idera Pharmaceuticals, Inc. Immunostimulatory properties of oligonucleotide-based compounds comprising modified immunostimulatory dinucleotides
EP1966377A2 (en) 2005-11-21 2008-09-10 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
MX2008008278A (en) * 2005-12-20 2008-10-01 Idera Pharmaceuticals Inc Novel synthetic agonists of toll-like receptors containing cg dinucleotide modifications.
JP2009521218A (en) * 2005-12-20 2009-06-04 イデラ ファーマシューティカルズ インコーポレイテッド Immunostimulatory action of palindromic immune modulatory oligonucleotides (IMO (TM)) containing palindromic segments of different lengths
AU2006342447B2 (en) 2005-12-28 2013-06-20 Translational Therapeutics, Inc Translational dysfunction based therapeutics
JP5713377B2 (en) 2005-12-28 2015-05-07 ザ スクリプス リサーチ インスティテュート Natural antisense and non-coding RNA transcripts as drug targets
PT2161038E (en) 2006-01-26 2014-03-10 Isis Pharmaceuticals Inc Compositions and their uses directed to huntingtin
JP5213723B2 (en) 2006-01-27 2013-06-19 アイシス ファーマシューティカルズ, インコーポレーテッド Oligomer compounds and compositions for use in modulating microRNA
KR101304071B1 (en) * 2006-01-27 2013-09-06 아이시스 파마수티컬즈 인코포레이티드 6-modified bicyclic nucleic acid analogs
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
ES2452820T3 (en) * 2006-04-07 2014-04-02 Methylgene, Inc. Benzamide derivatives as histone deacetylase inhibitors
EP2505646A1 (en) 2006-05-05 2012-10-03 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of CRP
AU2007249349B2 (en) 2006-05-11 2012-03-08 Isis Pharmaceuticals, Inc. 5'-Modified bicyclic nucleic acid analogs
US7666854B2 (en) * 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
US8198253B2 (en) 2006-07-19 2012-06-12 Isis Pharmaceuticals, Inc. Compositions and their uses directed to HBXIP
WO2008017473A2 (en) 2006-08-08 2008-02-14 Gunther Hartmann Structure and use of 5' phosphate oligonucleotides
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
AU2007294858B2 (en) 2006-09-14 2011-05-12 Medgenics Medical Israel, Ltd Long lasting drug formulations
ES2551892T3 (en) 2006-09-15 2015-11-24 Ottawa Health Research Institute Oncolytic rhabdovirus
EP2338499A1 (en) 2006-10-10 2011-06-29 Gunther Hartmann 5' triphosphate oligonucleotide induces anti-viral response
BRPI0717474A2 (en) * 2006-10-20 2014-03-11 Arizona Board Of Regentes For And On Behalf Of Arizona State University Modified cyanobacteria
US8399452B2 (en) 2006-10-28 2013-03-19 Methylgene Inc. Dibenzo[b,f][1,4]oxazepin-11-yl-N-hydroxybenzamides as HDAC inhibitors
US7645578B2 (en) * 2006-10-31 2010-01-12 Agilent Technologies, Inc. Cleavage of RNA at redundant sites
KR20090094383A (en) 2006-12-19 2009-09-04 메틸진 인크. Inhibitors of histone deacetylase and prodrugs thereof
EP1935428A1 (en) * 2006-12-22 2008-06-25 Antisense Pharma GmbH Oligonucleotide-polymer conjugates
EP2641971A1 (en) 2007-01-29 2013-09-25 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
EP2121987B1 (en) 2007-02-09 2012-06-13 Northwestern University Particles for detecting intracellular targets
US8030344B2 (en) * 2007-03-13 2011-10-04 Methylgene Inc. Inhibitors of histone deacetylase
US9556210B2 (en) 2007-04-23 2017-01-31 Sabag-Rfa Ltd. System for delivering therapeutic agents into living cells and cells nuclei
WO2014064687A1 (en) 2012-10-22 2014-05-01 Deliversir Ltd A system for delivering therapeutic agents into living cells and cells nuclei
US9156865B2 (en) 2007-04-23 2015-10-13 Deliversir Ltd System for delivering therapeutic agents into living cells and cells nuclei
EP2144633B1 (en) * 2007-04-23 2014-07-16 Deliversir Ltd A system for delivering therapeutic agents into living cells and cells nuclei
EP2160464B1 (en) 2007-05-30 2014-05-21 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
EP2173760B2 (en) 2007-06-08 2015-11-04 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
AU2008272918B2 (en) * 2007-07-05 2012-09-13 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
ES2439591T3 (en) 2007-08-15 2014-01-23 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
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
WO2009045536A2 (en) * 2007-10-05 2009-04-09 The University Of North Carolina At Chapel Hill Receptor targeted oligonucleotides
EP2219680A2 (en) * 2007-11-13 2010-08-25 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
EP2222851B1 (en) 2007-11-20 2017-06-28 Ionis Pharmaceuticals, Inc. Modulation of cd40 expression
US8546556B2 (en) * 2007-11-21 2013-10-01 Isis Pharmaceuticals, Inc Carbocyclic alpha-L-bicyclic nucleic acid analogs
EP4074344A1 (en) 2007-12-04 2022-10-19 Arbutus Biopharma Corporation Targeting lipids
WO2009100320A2 (en) * 2008-02-07 2009-08-13 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
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
US8846639B2 (en) 2008-04-04 2014-09-30 Isis Pharmaceutical, Inc. Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
AU2009234266B2 (en) 2008-04-11 2015-08-06 Tekmira Pharmaceuticals Corporation Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
WO2009141146A1 (en) 2008-05-21 2009-11-26 Gunther Hartmann 5' triphosphate oligonucleotide with blunt end and uses thereof
US20110262485A1 (en) 2008-08-04 2011-10-27 University Of Miami Sting (stimulator of interferon genes), a regulator of innate immune responses
AU2009275387B2 (en) 2008-08-25 2010-07-08 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
WO2011028218A1 (en) 2009-09-02 2011-03-10 Alnylam Pharmaceuticals, Inc. Process for triphosphate oligonucleotide synthesis
JP2012513953A (en) 2008-09-23 2012-06-21 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 Chemical modification of monomers and oligonucleotides using cycloaddition
DK2356129T3 (en) * 2008-09-24 2013-05-13 Isis Pharmaceuticals Inc Substituted alpha-L bicyclic nucleosides
DK2361256T3 (en) 2008-09-24 2013-07-01 Isis Pharmaceuticals Inc Cyclohexenyl-nucleic acid analogues
CN102245186B (en) 2008-10-15 2014-08-27 Isis制药公司 Modulation of factor 11 expression
EP4174178A1 (en) 2008-10-24 2023-05-03 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for dmd
US8987435B2 (en) 2008-10-24 2015-03-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
JP5763539B2 (en) 2008-10-24 2015-08-12 アイシス ファーマシューティカルズ, インコーポレーテッド 5 'and 2' bis-substituted nucleosides and oligomeric compounds produced therefrom
KR20230147211A (en) 2008-11-10 2023-10-20 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
KR101692880B1 (en) 2008-11-24 2017-01-04 노오쓰웨스턴 유니버시티 Polyvalent rna-nanoparticle compositions
CN102317458B (en) 2008-12-04 2018-01-02 库尔纳公司 Pass through treatment of the suppression of erythropoietin(EPO) (EPO) natural antisense transcript to EPO relevant diseases
US20110237649A1 (en) 2008-12-04 2011-09-29 Opko Curna, Llc Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
MX358603B (en) 2008-12-04 2018-08-28 Curna Inc Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene.
WO2010080554A1 (en) 2008-12-17 2010-07-15 Avi Biopharma, 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
KR101546673B1 (en) * 2009-01-15 2015-08-25 삼성전자주식회사 Toner for electrophotographic and process for preparing the same
WO2010090969A1 (en) 2009-02-06 2010-08-12 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
WO2010091308A2 (en) 2009-02-06 2010-08-12 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
PL2396038T3 (en) 2009-02-12 2016-05-31 Curna Inc Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
CN102439149B (en) 2009-02-12 2018-01-02 库尔纳公司 By suppressing to treat the related diseases of GDNF for the natural antisense transcript of the glial derived neurotrophic factor (GDNF)
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
EP2403946A4 (en) 2009-03-04 2012-11-14 Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt 1
EP2408919B1 (en) 2009-03-16 2017-10-18 CuRNA, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases by inhibition of natural antisense transcript to nrf2
US9708604B2 (en) 2009-03-17 2017-07-18 Curna, Inc. Treatment of delta-like 1 homolog (DLK1) related diseases by inhibition of natural antisense transcript to DLK1
EP3524275A1 (en) 2009-04-22 2019-08-14 Massachusetts Institute Of Technology Innate immune supression enables repeated delivery of long rna molecules
WO2010127195A2 (en) 2009-05-01 2010-11-04 Curna, Inc. Antisense oligonucleotides of hemoglobins
US20120046236A1 (en) 2009-05-06 2012-02-23 Opko Curna Llc Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
CN106237345A (en) 2009-05-06 2016-12-21 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
JP5922017B2 (en) 2009-05-18 2016-05-24 クルナ・インコーポレーテッド Treatment of reprogramming factor-related diseases by suppression of natural antisense transcripts against the reprogramming factor
JP2012527248A (en) 2009-05-22 2012-11-08 クルナ・インコーポレーテッド Treatment of TFE3 and insulin receptor substrate 2 (IRS2) related diseases by inhibition of natural antisense transcripts against transcription factor E3 (TFE3)
JP5960049B2 (en) 2009-05-28 2016-08-02 クルナ・インコーポレーテッド Treatment of antiviral gene-related diseases by suppression of natural antisense transcripts against antiviral genes
NZ622843A (en) 2009-06-10 2015-10-30 Tekmira Pharmaceuticals Corp Improved lipid formulation
US20120171170A1 (en) 2009-06-16 2012-07-05 Opko Curna, Llc Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
CN102612560B (en) 2009-06-16 2017-10-17 库尔纳公司 By suppressing to treat the related diseases of PON1 for the natural antisense transcript of PON1 (PON1)
CA2765889A1 (en) 2009-06-24 2010-12-29 Opko Curna, Llc Treatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
CA2765815A1 (en) 2009-06-26 2010-12-29 Opko Curna, Llc Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
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)
US9012421B2 (en) 2009-08-06 2015-04-21 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
EP2470657B1 (en) 2009-08-25 2019-10-23 CuRNA, Inc. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
SI2470656T1 (en) 2009-08-27 2015-07-31 Idera Pharmaceuticals, Inc. Composition for inhibiting gene expression and uses thereof
ES2599076T3 (en) 2009-09-02 2017-01-31 Genentech, Inc. Smoothened mutant and methods of use thereof
BR112012005774B8 (en) 2009-09-14 2023-02-14 Jennerex Inc use of an anti-angiogenic kinase inhibitor in cancer treatment
CA2778442A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
AU2010313154B2 (en) 2009-10-30 2016-05-12 Northwestern University Templated nanoconjugates
HUE040445T2 (en) 2009-11-12 2019-03-28 Univ Western Australia Antisense molecules and methods for treating pathologies
CN107312777B (en) 2009-11-13 2020-11-13 萨雷普塔治疗公司 Antisense antiviral compounds and methods of treating influenza virus infection
MX2012006072A (en) 2009-11-30 2012-07-23 Genentech Inc Antibodies for treating and diagnosing tumors expressing slc34a2 (tat211 = seqid2 ).
WO2011071860A2 (en) 2009-12-07 2011-06-16 Alnylam Pharmaceuticals, Inc. Compositions for nucleic acid delivery
MX337062B (en) 2009-12-10 2016-02-11 Ottawa Hospital Res Inst Oncolytic rhabdovirus.
KR101823702B1 (en) 2009-12-16 2018-01-30 큐알엔에이, 인크. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
DK2516648T3 (en) 2009-12-23 2018-02-12 Curna Inc TREATMENT OF HEPATOCYTE GROWTH FACTOR (HGF) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT AGAINST HGF
JP6031356B2 (en) 2009-12-23 2016-11-24 カッパーアールエヌエー,インコーポレイテッド Treatment of uncoupling protein 2 (UCP2) -related diseases by inhibition of natural antisense transcripts against UCP2.
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
CN102770540B (en) 2009-12-29 2017-06-23 库尔纳公司 P63 relevant diseases are treated by suppressing the natural antisense transcript of oncoprotein 63 (p63)
NO2521784T3 (en) 2010-01-04 2018-05-05
US8912157B2 (en) 2010-01-06 2014-12-16 Curna, Inc. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
US8779118B2 (en) 2010-01-11 2014-07-15 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
DK2524039T3 (en) 2010-01-11 2018-03-12 Curna Inc TREATMENT OF GENDER HORMON-BINDING GLOBULIN (SHBG) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTS TO SHBG
KR101853510B1 (en) 2010-01-25 2018-06-20 큐알엔에이, 인크. Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
WO2011100131A2 (en) 2010-01-28 2011-08-18 Alnylam Pharmacuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
WO2011094580A2 (en) 2010-01-28 2011-08-04 Alnylam Pharmaceuticals, Inc. Chelated copper for use in the preparation of conjugated oligonucleotides
EP2539452B1 (en) 2010-02-22 2016-07-27 CuRNA, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
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
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
BR112012020373A8 (en) 2010-02-23 2018-01-02 Genentech Inc isolated antibody, cell, isolated nucleic acid, method of identification, cell proliferation inhibition, therapeutic treatment, determination of the presence of a tat419 protein, diagnosis of the presence of cancer and distribution of cytotoxic agent
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
EP2545173A2 (en) 2010-03-12 2013-01-16 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
US8906875B2 (en) 2010-03-12 2014-12-09 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
EP3517613A1 (en) 2010-04-09 2019-07-31 CuRNA, Inc. Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
WO2011139714A2 (en) 2010-04-26 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-trna synthetase
CN103096911B (en) 2010-04-27 2018-05-29 Atyr 医药公司 Treatment relevant with the protein fragments of Isoleucyl-tRNA synthetase, diagnosis and the innovation of antibody compositions are found
EP3173419A1 (en) 2010-04-28 2017-05-31 Ionis Pharmaceuticals, Inc. Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
JP6008837B2 (en) 2010-04-28 2016-10-19 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 alanyl tRNA synthetase
US9127033B2 (en) 2010-04-28 2015-09-08 Isis Pharmaceuticals, Inc. 5′ modified nucleosides and oligomeric compounds prepared therefrom
US9156873B2 (en) 2010-04-28 2015-10-13 Isis Pharmaceuticals, Inc. Modified 5′ diphosphate nucleosides and oligomeric compounds prepared therefrom
JP5896175B2 (en) 2010-04-29 2016-03-30 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 transthyretin expression
CN103118693B (en) 2010-04-29 2017-05-03 Atyr 医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl tRNA synthetases
US20130156845A1 (en) 2010-04-29 2013-06-20 Alnylam Pharmaceuticals, Inc. Lipid formulated single stranded rna
EP2563912B1 (en) 2010-04-29 2018-09-05 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl trna synthetases
CA2797978C (en) 2010-05-03 2019-12-03 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-trna synthetases
CN102958941A (en) 2010-05-03 2013-03-06 霍夫曼-拉罗奇有限公司 Compositions and methods for the diagnosis and treatment of tumor
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
WO2011139986A2 (en) 2010-05-03 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases
KR20130101442A (en) 2010-05-03 2013-09-13 큐알엔에이, 인크. Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CA2798139C (en) 2010-05-04 2019-09-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-trna synthetase complex
CN109593814A (en) 2010-05-12 2019-04-09 哥伦比亚大学纽约管理委员会 Screening analysis, the method and pharmaceutical composition for identifying medicament
CA2805086C (en) 2010-05-13 2020-10-20 Sarepta Therapeutics, Inc. Antisense modulation of interleukins 17 and 23 signaling
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
TWI531370B (en) 2010-05-14 2016-05-01 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
JP6027965B2 (en) 2010-05-17 2016-11-16 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 leucyl-tRNA synthetase
DK2576783T3 (en) 2010-05-26 2018-03-12 Curna Inc TREATMENT OF ATONAL HOMOLOGY 1- (ATOH1) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTS AT ATOH1
EP2575856B1 (en) 2010-05-27 2017-08-16 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-trna synthetases
US8957200B2 (en) 2010-06-07 2015-02-17 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
US8846637B2 (en) 2010-06-08 2014-09-30 Isis Pharmaceuticals, Inc. Substituted 2′-amino and 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
CN103260649A (en) 2010-06-15 2013-08-21 迈德詹尼克斯医疗以色列有限公司 Long lasting drug formulations
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
CN103068982B (en) 2010-07-14 2017-06-09 库尔纳公司 DLG relevant diseases are treated by suppressing the natural antisense transcript of the big homologue of plate-like (DLG)
WO2012016188A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
CN103108650A (en) 2010-08-25 2013-05-15 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-trna synthetases
EP2426203B1 (en) 2010-09-02 2016-07-13 Université de Mons Agents useful in treating facioscapulohumeral muscular dystrophy
WO2012047968A2 (en) 2010-10-05 2012-04-12 Genentech, Inc. Mutant smoothened and methods of using the same
CA2813901C (en) 2010-10-06 2019-11-12 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
WO2012054723A2 (en) 2010-10-22 2012-04-26 Opko Curna Llc Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
EP2633052B1 (en) 2010-10-27 2018-04-11 CuRNA, Inc. Treatment of interferon-related developmental regulator 1 (ifrd1) related diseases by inhibition of natural antisense transcript to ifrd1
JP6336755B2 (en) 2010-11-12 2018-06-06 ザ ジェネラル ホスピタル コーポレイション Non-coding RNA related to polycomb
JP6126009B2 (en) 2010-11-17 2017-05-10 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 α-synuclein expression
US8987225B2 (en) 2010-11-23 2015-03-24 Curna, Inc. Treatment of NANOG related diseases by inhibition of natural antisense transcript to NANOG
CN103429258B (en) 2011-01-04 2016-03-09 新罗杰公司 By use oncolytic vaccinia virus generate for tumor antigen antibody and generate tumour-specific CDC
EP3202760B1 (en) 2011-01-11 2019-08-21 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
US9045749B2 (en) 2011-01-14 2015-06-02 The General Hospital Corporation Methods targeting miR-128 for regulating cholesterol/lipid metabolism
EP2668199B1 (en) 2011-01-27 2017-09-06 Ramot at Tel Aviv University, Ltd. Glycogen synthase kinase-3 inhibitors
EP2668200B1 (en) 2011-01-27 2017-03-15 Ramot at Tel-Aviv University Ltd. Glycogen synthase kinase-3 inhibitors
CN103391777A (en) 2011-02-02 2013-11-13 普林斯顿大学理事会 Sirtuin modulators as virus production modulators
TWI593416B (en) 2011-02-02 2017-08-01 艾克厘德製藥公司 Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
US8877722B2 (en) 2011-03-25 2014-11-04 Idera Pharmaceuticals, Inc. Compositions for inhibiting gene expression and uses thereof
EP2508530A1 (en) 2011-03-28 2012-10-10 Rheinische Friedrich-Wilhelms-Universität Bonn Purification of triphosphorylated oligonucleotides using capture tags
US20140088174A1 (en) 2011-04-05 2014-03-27 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Compounds and methods for altering activin receptor-like kinase signaling
ES2627529T3 (en) 2011-06-08 2017-07-28 Children's Hospital Of Eastern Ontario Research Institute Inc. Compositions for glioblastoma treatment
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
US9593330B2 (en) 2011-06-09 2017-03-14 Curna, Inc. Treatment of frataxin (FXN) related diseases by inhibition of natural antisense transcript to FXN
US20140227293A1 (en) 2011-06-30 2014-08-14 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
EP3922722B1 (en) 2011-08-11 2023-06-28 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
WO2013033230A1 (en) 2011-08-29 2013-03-07 Isis Pharmaceuticals, Inc. Oligomer-conjugate complexes and their use
AU2012308302A1 (en) 2011-09-14 2014-03-20 Northwestern University Nanoconjugates able to cross the blood-brain barrier
EP3533873A1 (en) 2011-09-14 2019-09-04 Translate Bio MA, Inc. Multimeric oligonucleotide compounds
WO2013043720A1 (en) 2011-09-20 2013-03-28 The University Of North Carolina At Chapel Hill Regulation of sodium channels by plunc proteins
US9701623B2 (en) 2011-09-27 2017-07-11 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
CA2851280C (en) 2011-10-11 2021-05-18 The Brigham And Women's Hospital, Inc. Micrornas in neurodegenerative disorders
EP2596806A1 (en) 2011-11-25 2013-05-29 Index Pharmaceuticals AB Method for prevention of colectomy
US10557136B2 (en) 2011-12-12 2020-02-11 Oncolmmunin Inc. In vivo delivery of oligonucleotides
WO2013121426A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
EP2849778B1 (en) 2012-03-13 2017-12-27 James Cook University Ac-TMP-2 FOR USE IN TREATING INFLAMMATION
EP2639238A1 (en) 2012-03-15 2013-09-18 Universität Bern Tricyclic nucleosides and oligomeric compounds prepared therefrom
US20150031750A1 (en) 2012-03-15 2015-01-29 The Scripps Research Institute Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
WO2013138662A1 (en) 2012-03-16 2013-09-19 4S3 Bioscience, Inc. Antisense conjugates for decreasing expression of dmpk
CA2866392C (en) 2012-03-30 2023-10-24 Washington University Methods for modulating tau expression for reducing seizure and modifying a neurodegenerative syndrome
EP2850092B1 (en) 2012-04-09 2017-03-01 Ionis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
JP2015518485A (en) 2012-04-20 2015-07-02 アプタミアール セラピューティクス インコーポレイテッド Thermogenic miRNA regulator
EP3597741A1 (en) 2012-04-27 2020-01-22 Duke University Genetic correction of mutated genes
EP3511416A1 (en) 2012-05-16 2019-07-17 Translate Bio MA, Inc. Compositions and methods for modulating gene expression
JP2015523854A (en) 2012-05-16 2015-08-20 ラナ セラピューティクス インコーポレイテッド Compositions and methods for modulating SMN gene family expression
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
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
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
EP2885312A4 (en) 2012-08-15 2016-01-20 Isis Pharmaceuticals Inc Method of preparing oligomeric compounds using modified capping protocols
EP2892617B1 (en) 2012-09-06 2018-06-13 The University of Chicago Antisense polynucleotides to induce exon skipping and methods of treating dystrophies
EP2712870A1 (en) 2012-09-27 2014-04-02 Rheinische Friedrich-Wilhelms-Universität Bonn Novel RIG-I ligands and methods for producing them
PL2900686T3 (en) 2012-09-28 2021-01-25 The University Of North Carolina At Chapel Hill Aav vectors targeted to oligodendrocytes
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
BR112015014987A2 (en) 2012-12-20 2017-08-15 Sarepta Therapeutics Inc IMPROVED EXON-SKIPPING COMPOSITIONS FOR THE TREATMENT OF MUSCULAR DYSTROPHY
US10363293B2 (en) 2013-02-21 2019-07-30 Turnstone Limited Partnership Vaccine composition
NZ775701A (en) 2013-03-14 2022-08-26 Sarepta Therapeutics Inc Exon skipping compositions for treating muscular dystrophy
ES2807379T3 (en) 2013-03-14 2021-02-22 Ionis Pharmaceuticals Inc Compositions and methods to regulate the expression of Tau
TR201903009T4 (en) 2013-03-14 2019-03-21 Sarepta Therapeutics Inc Exon skipping compositions for the treatment of muscular dystrophy.
US9506058B2 (en) 2013-03-15 2016-11-29 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
BR112015027321A8 (en) 2013-05-01 2018-01-02 Isis Pharmaceuticals Inc COMPOUNDS AND COMPOSITIONS FOR MODULING APOLIPOPROTEIN(A) EXPRESSION AND THEIR USES
DK3007704T3 (en) 2013-06-13 2021-03-29 Antisense Therapeutics Ltd Combination therapy for acromegaly
EP3013850B1 (en) 2013-06-24 2018-07-25 Ramot at Tel-Aviv University Ltd. Glycogen synthase kinase-3 inhibitors
TWI702046B (en) 2013-07-19 2020-08-21 美商Ionis製藥公司 Compositions for modulating tau expression
CA2924130A1 (en) 2013-09-18 2015-03-26 James Cook University Anti-inflammatory proteins and methods of use
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
WO2015061246A1 (en) 2013-10-21 2015-04-30 Isis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
US20170268000A1 (en) 2013-12-02 2017-09-21 Ionis Pharmaceuticals, Inc. Antisense compounds and uses thereof
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
EP2883883A1 (en) 2013-12-16 2015-06-17 Cardio3 Biosciences S.A. Therapeutic targets and agents useful in treating ischemia reperfusion injury
US9636298B2 (en) 2014-01-17 2017-05-02 Methylgene Inc. Prodrugs of compounds that enhance antifungal activity and compositions of said prodrugs
EP3099333B8 (en) 2014-01-31 2020-12-30 Temple University of the Commonwealth System of Higher Education Bag3 as a target for therapy of heart failure
US20170044232A1 (en) 2014-02-04 2017-02-16 Genentech, Inc. Mutant smoothened and methods of using the same
EP3119789B1 (en) 2014-03-17 2020-04-22 Ionis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
WO2015143245A1 (en) 2014-03-19 2015-09-24 Isis Pharmaceuticals, Inc. Methods for modulating ataxin 2 expression
PL3119888T3 (en) 2014-03-19 2021-12-20 Ionis Pharmaceuticals, Inc. Compositions for modulating ataxin 2 expression
PL3757214T3 (en) 2014-04-01 2022-09-12 Biogen Ma Inc. Compositions for modulating sod-1 expression
US10221416B2 (en) 2014-04-24 2019-03-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
WO2015168172A1 (en) 2014-04-28 2015-11-05 Isis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
HUE052931T2 (en) 2014-05-01 2021-05-28 Ionis Pharmaceuticals Inc Compositions and methods for modulating angiopoietin-like 3 expression
ES2745825T3 (en) 2014-05-01 2020-03-03 Ionis Pharmaceuticals Inc Compositions and methods for modulating expression of complement factor B
KR102149571B1 (en) 2014-05-01 2020-08-31 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating growth hormone receptor expression
ES2908962T3 (en) 2014-06-10 2022-05-04 Univ Erasmus Med Ct Rotterdam Antisense oligonucleotides useful in the treatment of Pompe disease
EP3161159B1 (en) 2014-06-25 2020-08-05 The General Hospital Corporation Targeting human satellite ii (hsatii)
WO2015200901A1 (en) 2014-06-26 2015-12-30 The Trustees Of Columbia University In The City Of New York Inhibition of serotonin expression in gut enteroendocrine cells results in conversion to insulin-positive cells
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
JP6841753B2 (en) 2014-09-15 2021-03-10 ザ チルドレンズ メディカル センター コーポレーション Methods and compositions for increasing somatic cell nuclear transfer (SCNT) efficiency by removing histone H3-lysine trimethylation
WO2016049512A1 (en) 2014-09-26 2016-03-31 University Of Massachusetts Rna-modulating agents
US20170304459A1 (en) 2014-10-10 2017-10-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhalation delivery of conjugated oligonucleotide
US20160101128A1 (en) 2014-10-10 2016-04-14 Idera Pharmaceuticals, Inc. Treatment of cancer using tlr9 agonist with checkpoint inhibitors
CA2968531A1 (en) 2014-11-21 2016-05-26 Northwestern University The sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
US10538763B2 (en) 2015-01-16 2020-01-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of DUX4
AU2016215124B2 (en) 2015-02-06 2020-08-06 The University Of North Carolina At Chapel Hill Optimized human clotting Factor VIII gene expression cassettes and their use
JP6929791B2 (en) 2015-02-09 2021-09-01 デューク ユニバーシティ Compositions and methods for epigenome editing
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
WO2016164463A1 (en) 2015-04-07 2016-10-13 The General Hospital Corporation Methods for reactivating genes on the inactive x chromosome
PL3283500T3 (en) 2015-04-08 2021-05-31 The University Of Chicago Compositions and methods for correcting limb girdle muscular dystrophy type 2c using exon skipping
US10407678B2 (en) 2015-04-16 2019-09-10 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
US10376536B2 (en) 2015-05-11 2019-08-13 Murdoch University Multiple sclerosis treatment
WO2016210241A1 (en) 2015-06-26 2016-12-29 Beth Israel Deaconess Medical Center, Inc. Cancer therapy targeting tetraspanin 33 (tspan33) in myeloid derived suppressor cells
AU2016287499B2 (en) 2015-06-29 2022-08-04 Caris Science, Inc. Therapeutic oligonucleotides
WO2017019918A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
WO2017053781A1 (en) 2015-09-25 2017-03-30 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
WO2017066497A2 (en) 2015-10-13 2017-04-20 Duke University Genome engineering with type i crispr systems in eukaryotic cells
WO2017075670A1 (en) 2015-11-05 2017-05-11 Children's Hospital Los Angeles "mobilizing leukemia cells"
BR122023026882A2 (en) 2015-11-06 2024-01-23 Ionis Pharmaceuticals, Inc USE OF AN OLIGOMERIC COMPOUND
US11001622B2 (en) 2015-11-19 2021-05-11 The Brigham And Women's Hospital, Inc. Method of treating autoimmune disease with lymphocyte antigen CD5-like (CD5L) protein
WO2017096395A1 (en) 2015-12-04 2017-06-08 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
US10993995B2 (en) 2015-12-07 2021-05-04 Erasmus University Medical Center Rotterdam Enzymatic replacement therapy and antisense therapy for pompe disease
JOP20200228A1 (en) 2015-12-21 2017-06-16 Novartis Ag Compositions and methods for decreasing tau expression
CA3006599A1 (en) 2016-01-05 2017-07-13 Ionis Pharmaceuticals, Inc. Methods for reducing lrrk2 expression
EA201891601A1 (en) 2016-01-10 2019-03-29 Неоткс Терапьютикс Лтд. METHODS AND COMPOSITIONS ENHANCED EFFICIENCY BY MEDIATED SUPERANTIGEN IMMUNOTHERAPY OF MALIGNANT TUMORS
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
EP3419665A4 (en) 2016-02-25 2019-10-16 The Brigham and Women's Hospital, Inc. Treatment methods for fibrosis targeting smoc2
US10961271B2 (en) 2016-03-16 2021-03-30 Ionis Pharmaceuticals, Inc. Methods of modulating KEAP1
US10577607B2 (en) 2016-03-16 2020-03-03 Ionis Pharmaceuticals, Inc. Modulation of DYRK1B expression
EP3430137A4 (en) 2016-03-18 2019-11-06 Caris Science, Inc. Oligonucleotide probes and uses thereof
AU2017271579B2 (en) 2016-05-25 2023-10-19 Caris Science, Inc. Oligonucleotide probes and uses thereof
WO2017219017A1 (en) 2016-06-17 2017-12-21 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
CA3030701A1 (en) 2016-07-11 2018-01-18 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
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
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
CA3037046A1 (en) 2016-10-31 2018-05-03 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
EP3548620A4 (en) 2016-12-02 2020-07-22 Cold Spring Harbor Laboratory Modulation of lnc05 expression
EP3577224A4 (en) 2017-02-03 2020-11-25 The University Of Western Australia Novel treatment for neat1 associated disease
WO2018165564A1 (en) 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
CA3058290A1 (en) 2017-04-18 2018-10-25 Universite Libre De Bruxelles Biomarkers and targets for proliferative diseases
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
EP3612232A1 (en) 2017-04-21 2020-02-26 The Broad Institute, Inc. Targeted delivery to beta cells
GB201707501D0 (en) 2017-05-10 2017-06-21 Index Pharmaceuticals Ab New therapy 2
GB201707503D0 (en) 2017-05-10 2017-06-21 Index Pharmaceuticals Ab New Therapy 3
EP3665304A4 (en) 2017-08-11 2021-04-28 Agency for Science, Technology and Research Method for screening splicing variants or events
US11197884B2 (en) 2017-08-18 2021-12-14 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
WO2019051173A1 (en) 2017-09-08 2019-03-14 Ionis Pharmaceuticals, Inc. Modulators of smad7 expression
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
CA3086485A1 (en) 2017-12-21 2019-06-27 Alnylam Pharmaceuticals, Inc. Chirally-enriched double-stranded rna agents
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
JP2021511072A (en) 2018-01-15 2021-05-06 アイオニス・ファーマシューティカルズ・インコーポレイテッドIonis Pharmaceuticals,Inc. DNM2 expression modulator
US11732260B2 (en) 2018-03-02 2023-08-22 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-β precursor protein
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
WO2019183440A1 (en) 2018-03-22 2019-09-26 Ionis Pharmaceuticals, Inc. Methods for modulating fmr1 expression
CA3095545A1 (en) 2018-03-30 2019-10-03 Rheinische Friedrich-Wilhelms-Universitat Bonn Aptamers for targeted activaton of t cell-mediated immunity
KR20200141470A (en) 2018-04-06 2020-12-18 칠드런'즈 메디컬 센터 코포레이션 Composition and method for adjusting somatic cell reprogramming and imprinting
SG11202008660TA (en) 2018-04-11 2020-10-29 Ionis Pharmaceuticals Inc Modulators of ezh2 expression
GB201807312D0 (en) 2018-05-03 2018-06-20 Index Pharmaceuticals Ab Formulation
CN112189053A (en) 2018-05-09 2021-01-05 Ionis制药公司 Compounds and methods for reducing expression of ATXN3
JOP20200280A1 (en) 2018-05-09 2020-11-05 Ionis Pharmaceuticals Inc Compounds and methods for reducing fxi expression
WO2019241648A1 (en) 2018-06-14 2019-12-19 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing stmn2 expression
JP7315594B2 (en) 2018-06-27 2023-07-26 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 Compounds and methods for reducing LRRK2 expression
CN112805004A (en) 2018-07-02 2021-05-14 艾珀特玛治疗公司 Targeted delivery of therapeutic agents to human adipocytes
AR115847A1 (en) 2018-07-25 2021-03-03 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN2
EP3843845A4 (en) 2018-08-29 2022-05-11 University Of Massachusetts Inhibition of protein kinases to treat friedreich ataxia
WO2020048607A1 (en) 2018-09-07 2020-03-12 Université Libre de Bruxelles Srsf2 as rna epigenetic factor
GB201818579D0 (en) 2018-11-14 2018-12-26 Index Pharmaceuticals Ab New therapy
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
CA3119456A1 (en) 2018-11-15 2020-05-22 Ionis Pharmaceuticals, Inc. Modulators of irf5 expression
WO2020160453A1 (en) 2019-01-31 2020-08-06 Ionis Pharmaceuticals, Inc. Modulators of yap1 expression
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
US11279932B2 (en) 2019-02-27 2022-03-22 Ionis Pharmaceuticals, Inc. Modulators of MALAT1 expression
WO2020205463A1 (en) 2019-03-29 2020-10-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating ube3a-ats
WO2020227758A1 (en) 2019-05-14 2020-11-19 Monash University Modulators and modulation of the receptor for advanced glycation end-products rna
US20220213194A1 (en) 2019-05-15 2022-07-07 Neotx Therapeutics Ltd. Cancer treatment
EP3956450A4 (en) 2019-07-26 2022-11-16 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
EP4013767A4 (en) 2019-08-15 2023-10-25 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
GB201912191D0 (en) 2019-08-24 2019-10-09 Index Pharmaceuticals Ab New therapy
EP4041889A1 (en) 2019-10-08 2022-08-17 Luxembourg Institute Of Health (LIH) Inhibitor of dj-1 for use in treating immunoaging
SG10201914033YA (en) 2019-12-31 2021-07-29 Wilmar International Ltd Polypeptides with Lipase Activity and Uses Thereof
MX2022010602A (en) 2020-02-28 2022-09-09 Ionis Pharmaceuticals Inc Compounds and methods for modulating smn2.
CN115484978A (en) 2020-03-05 2022-12-16 尼奥克斯医疗有限公司 Methods and compositions for treating cancer using immune cells
CA3181546A1 (en) 2020-05-01 2021-11-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating atxn1
MX2022016338A (en) 2020-06-29 2023-01-24 Ionis Pharmaceuticals Inc Compounds and methods for modulating plp1.
EP4136092A4 (en) 2020-11-18 2023-10-11 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
GB202018468D0 (en) 2020-11-24 2021-01-06 Index Pharmaceuticals Ab New therapy
AU2022293556A1 (en) 2021-06-18 2024-01-18 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ifnar1 expression
EP4147688A1 (en) 2021-09-09 2023-03-15 InDex Pharmaceuticals AB Enema formulation
WO2023168427A1 (en) 2022-03-03 2023-09-07 Yale University Compositions and methods for delivering therapeutic polynucleotides for exon skipping
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024044352A1 (en) 2022-08-26 2024-02-29 The General Hospital Corporation Methods and compositions for prognosis and treatment of dilated cardiomyopathy and heart failure

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4661450A (en) * 1983-05-03 1987-04-28 Molecular Genetics Research And Development Limited Partnership Molecular cloning of RNA using RNA ligase and synthetic oligonucleotides
EP0260032B1 (en) * 1986-09-08 1994-01-26 Ajinomoto Co., Inc. Compounds for the cleavage at a specific position of RNA, oligomers employed for the formation of said compounds, and starting materials for the synthesis of said oligomers
US5110802A (en) * 1987-07-14 1992-05-05 City Of Hope Oligonucleotide phosphonates and method of inhibiting a human immunodeficiency virus in vitro utilizing said oligonucleotide phosphonates
WO1989005358A1 (en) * 1987-11-30 1989-06-15 University Of Iowa Research Foundation Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
US5098890A (en) * 1988-11-07 1992-03-24 Temple University-Of The Commonwealth System Of Higher Education Antisence oligonucleotides to c-myb proto-oncogene and uses thereof
US5176996A (en) * 1988-12-20 1993-01-05 Baylor College Of Medicine Method for making synthetic oligonucleotides which bind specifically to target sites on duplex DNA molecules, by forming a colinear triplex, the synthetic oligonucleotides and methods of use
US5149798A (en) * 1989-04-06 1992-09-22 Worcester Foundation For Experimental Biology Process for synthesizing oligonucleotides and their analogs adaptable to large scale syntheses
JPH05500799A (en) * 1989-06-05 1993-02-18 ギリアド サイエンシス,インコーポレイテッド Exonuclease-resistant oligonucleotide and method for its preparation
US5220007A (en) * 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
US5149797A (en) * 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides

Also Published As

Publication number Publication date
WO1991012323A1 (en) 1991-08-22
EP0515511B1 (en) 1996-08-14
US5149797A (en) 1992-09-22
DE69121391D1 (en) 1996-09-19
DE69121391T2 (en) 1997-03-06
GR3021278T3 (en) 1997-01-31
DK0515511T3 (en) 1996-11-18
EP0515511A1 (en) 1992-12-02
US5366878A (en) 1994-11-22
US5635377A (en) 1997-06-03
JPH05506355A (en) 1993-09-22
ATE141326T1 (en) 1996-08-15
ES2090314T3 (en) 1996-10-16

Similar Documents

Publication Publication Date Title
CA2076037A1 (en) Method of site-specific alteration of rna and production of encoded polypeptides
US5220007A (en) Method of site-specific alteration of RNA and production of encoded polypeptides
AU740336B2 (en) Heteroduplex mutational vectors and use thereof in bacteria
AU740702B2 (en) Cell-free chimeraplasty and eukaryotic use of heteroduplex mutational vectors
CN104080462B (en) For modifying the composition and method of scheduled target nucleic acid sequence
JP7306696B2 (en) RNA-guided nucleic acid-modifying enzyme and method of use thereof
ES2749108T3 (en) RNA-directed DNA excision using the Cas9-rRNA complex
EP0733059B1 (en) Compounds and methods for site-directed mutations in eukaryotic cells
CA3142883A1 (en) Engineered casx systems
AU2005248371B2 (en) Methods for dynamic vector assembly of DNA cloning vector plasmids
WO2018089664A1 (en) Variant rna-guided polypeptides and methods of use
KR20180081600A (en) Substances and methods for the treatment of ticin-based diarrhea and other ticinopathies
CA3201258A1 (en) Engineered class 2 type v crispr systems
CA3174557A1 (en) Class ii, type v crispr systems
JP2023510352A (en) Compositions and methods for targeting PCSK9
GB2393441A (en) A method for producing a multi-gene recombinant vector
JP2023552379A (en) Compositions and methods for targeting BCL11A
JP2004500018A (en) Oligonucleotide containing antisense sequence stabilized by secondary structure and pharmaceutical composition containing the same
WO2020005980A1 (en) Rna-guided effector proteins and methods of use thereof
CA3219187A1 (en) Class ii, type v crispr systems
KR20220039564A (en) Compositions and methods for use of engineered base editing fusion protein
WO1997043404A2 (en) Ribozyme variants with improved catalytic activity under low magnesium conditions
WO2021071909A1 (en) Compositions and methods for homology-directed recombination
IL303360A (en) Engineered class 2 type v crispr systems

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued