CA2088673A1 - Compounds and methods for inhibiting gene expression - Google Patents

Compounds and methods for inhibiting gene expression

Info

Publication number
CA2088673A1
CA2088673A1 CA002088673A CA2088673A CA2088673A1 CA 2088673 A1 CA2088673 A1 CA 2088673A1 CA 002088673 A CA002088673 A CA 002088673A CA 2088673 A CA2088673 A CA 2088673A CA 2088673 A1 CA2088673 A1 CA 2088673A1
Authority
CA
Canada
Prior art keywords
independently
hydrogen
alkyl
compound
proviso
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
CA002088673A
Other languages
French (fr)
Inventor
Alexander L. Weis
Frederick H. Hausheer
Prasad V. C. Chaturvedula
Daniel J. Delecki
Paul F. Cavanaugh
Patricia S. Moskwa
Fred T. Oakes
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.)
Sanofi SA
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
Priority claimed from US07/562,180 external-priority patent/US5245022A/en
Application filed by Individual filed Critical Individual
Publication of CA2088673A1 publication Critical patent/CA2088673A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids

Abstract

The present invention relates to compounds, compositions and methods for inhibiting gene expression. The compounds of this invention comprise: 1) oligonucleoside sequences of from about 6 to about 200 bases having a three atom internucleoside linkage or 2) oligonucleotide sequences of from about 9 to about 200 bases having a diol at either or both termini.

Description

WO92/n2534PCTtUS91/05531 COMPO~NDS AND ~T~ODS
FOR IN~IBITING G~NE ~P~ESSION
De~criPtion Cro~s ~eference to Relate~ A~Plications 5This application is a continuation-in-part of application U.S. Serial No. 07/562,180, filed August 3, 1990; application U.S. Serial No. 07/582,287, filed September 13, 1990; application V.S. Serial No.
07/582,456, filed September 13, 1990; and application U.S.
Serial No. 07/582,457, filed September 13, 1990.
Technical F~eld The present invention relates to compounds, compositions and methods for inhibiting gene expression.
~ ~~ ~ The compounds of this invention comprise 1) oligonucleoside sequences of from about 6 to about 200 bases having a three atom internucleoside linkage or 2) oligonucleotide sequences of from about 9 to about 200 bases havi~3 a diol at either or both termini.

sacx~round of t~e Inventio~
An antisense compound .Ls a compound that binds to or hybridizes with a nucleotide sequence in a nucleic acid, RNA or DNA, to inhibit the function or synthesis of said nucleic acid. Because of their ability to hybridize with both ~NA and DNA, antisense compounds can interfere with gene expression at the level of transcription, RNA processing or translation.
Antisense molecules can be designed and synthesized to prevent the transcription of specific genes to mRNA by hybridizing with genomic DNA and directly or indirectly in~ibiting the action of RNA
polymerase. An advantage of targeting DNA ic that only small amounts of antisense compounds are needed to achieve a therapeutic effect. Alternatively, antisense compounds can be designed and synthesized to ~ybridize with RNA to inhibit post-transcriptional modification tRNA processing) or protein synthesis (transla~ion) - , ,. . ~ , i : , W092/025~ PCT/US91/0~31 ,~ v ~,J~ 3 - 2 - .

mechanisms. Exemplary target RNAs are messenger RNA
(mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA) and the like. Examples o~ processing and translation mechanisms include splicing of pre-mRNA to remove introns, capping of the 5' terminus of mRNA, hybridization arrest and nuclease mediated mRNA
hydrolysis.
At the pressnt time, however, the development of practical scientific and therapeutic applications of antisense technologies is hampered by a number of technical problems. Klausner, A., Biotechnoloqv, 8:303-304 (l990). Synthetic antisense molecules are susceptible to rapid degradation by nucleases that exist in target cells. The oligonucleoside sequences of antisense DNA or RNA, for example, are destroyed by exonucleases acting at either the 5' or 3' terminus of the nucleic acid. In addition, endonucleases can cleave the DNA or RNA at internal phosphodiester linkages between individual nucleosides. As a result of such cleavago, the effective half-life of administered antisense compounds is very short, necessitating the use of large, frequently administered, dosages.
Another problem is the extremely high cost of producing antisense D~A or RNA using availabl~
semiautomatic DNA synthesizers. It has recently been estimated that the cost of producing one gram of antisense DNA is abdut $100,000. Armstrong, L., Business Week, March 5, l990, page 89.
A further problem relates to the deliYery of antisense agents to desired targets within the body and cell. Antisense agents targeted to genomic DNA must gain acce~s to the nucleus ti.e. the agents mu~t permeate the plasma and nuclear me~brane). The need for increased membrane permeability (increased hydrcphobicity) must be balanced, however, against the . : . . ,. : ::
. . : : , . . . - :
. : . . .- : : :

W092/025~ PCT/US91/0S~l ,~ ) need for aqueous solubility (increased hydrophilicity) in body fluid compartments such as the plasma and cell cytosol.
` A still further problem relates to the stability of antisense agents whether free within the body or hy~ridized to target nucleic acids.
Oligonucleotide sequences such as antisense DNA are susceptible to steric reconfiguration around chiral phosphorous centers.
~ene targeting via antisense agents is the inevitable next step in human therapeutics. Armstrong, supra at 83. The success~ul application of antisense technology to the treatment of disease however, reguires finding solutions to the problems set forth above.
One approach to preparing antisense compounds that are stable, nuclease resistant, inexpensive to produce and which can be delivered to and hybridize with nucleic acid targets throughout the body is to synthesize oligonucleoside saquences with modi~ications in the normal phosphate-sugar bacXbone ~tructure.
In general, two types o$ oligonucleoside se~uences, with modified backbones have been reported.
The first type includes modi~icat:ions to the normal int~rnucleoside phosphodiester linXage. The second type includes replacement of the phosphodiester linkage with non phosphate internucleoside linkagas. Uhlmann, E. and Peyman, A., ~hemical ~eviews, 9(4):544-584 (1990).
Mod~fied phosphodiester linkages that have been report~d to date are phosphorothioates, alkylphosphotriesters, methylphosphonates and alkylphosphoramidat2s.
Phosphorothioate modified phosphodiester linkagas refer to phosphodiester ~onds in which one or more of the bridging oxygen atoms is replaced by sulfur.
Such linka~es, however, are not suitable for use in ' . ,; ~; , . ' .. . :., .

: ,, ~ ; :

W092/02S3~ PCT/US91/05531 -- 4 -- -- _ V ~

antisense compounds. The retention of the chiral phosphorus center results in steric variation of monothioates. Purther, both mono- and dit~ioates lack sequence specific hybridization and both are rapidly cleared from the plasma. The high affinity of phosphorothioates for glass and plastic also makes synthesis of these compounds difficult and inefficient.
Methyl- and ethylphosphotriesters have been prepared by reacting phosphodiester linked oligonucleosides with anhydrous ~ethanol or ethanol.
~iller, P.S. et al., J. Am. Chem. Soc., 93:66S7-6665(1971).
The triester linkage in oligodeoxyribonucleotide ethylphosphotriesters is stable under no~nal physioloqical pH conditions, although it can be hydrolyzed by strong acid or base.
Methylphosphotriesters are less stable than ethyl- and other alXylphosphotriesters at n~sutral pH, owing to the possibility o~ nucleophilic displacement of the triester methyl group by solvent. Oligod~eoxyribonucleotide ,ethylphosphotriesters appear to ]~e complately re~istant to hydrolysis by exonucleases and are not hydrolyzed by nucleases or esterases found in fetal bovine serum or human blood serum. Uhlmann, supra.
The methylphosphonates have several significant shortcomings in terms o~ therapeutic potential which include poor water solu~ility, chiral phosphorous centers, inability to control high yield stereoselective synthesis, rapid plasma clearance and urinary excretion.
Oligodeoxyribonucleoside phosphoramidates have internucleoside bonds containing nitrogen-phosphorus bonds. These nucleic acid analogs can be prepared from phosphoramadite intermediates or ~y oxidation of H-phosphonate intermediates in the presence of a primary W092/~2534 PCT/US91/05531 ~ 5 -c; , or secondary ~mine. Preparation of the H-phosphonate analogs and the oxidation reaction can be readily carried out in a co~mercial DNA synthesizer.
A variety of non-ionic oligonucleoside segyences containing non-phosphate internucleoside linkages such as carbonate, acetate, carbamate and dialkyl- or diarylsilyl- derivatives have been synthesized and reported.
Althouqh the carbonate linkage is resistant to hydrolysis ~y acid, it is rather easily cleav~d with base, and thus special precautions are required for removaL of the~protecting groups at the end of the synthesis. While stable dupl~xes have b~en observed between po:Ly(dA) analogs containing carboxymethyl ~5 internucleotlde linkages and poly(U) analogs, other bases have not been studied. Thu~s, it is not known whether the ~idelity of duplex formation with other bases will ~e perturbed by the carbonate linkage.
Internucleoside carbamates are reported to be more water soluble than other internucleoside bridges.
T~elut~lity o~ carbamate linX~ges is limited, however, since thymine carbamates do not ~orm hybrids with complementary DNA, while cytosine carbamates do not hybridize to guanine oligomers.
The carbamate linkage, like the carbonate linkage, is stable under physiological conditions.
Unlika the carbonates, however, the carba~ate linkage is stable to hydrolysis by bases, a property i.~ich simplifies tha synthesis of oligomers ~ontaining this linkage. The carbamate linkage is resistant to nuclease hydrolysis.
Like the carbonate and acetate linkages, the car~amate linXage does not re~emble the shape of t~e phosphodiester internucleotide bond. However, molecular models suggest that the linkage should allow the W0~)2/n253~ PCT~US9l/05~31 " ~ ~

oligomer to assume conformations which would allow it to form hydrogen-bonded complexes with complementary nucleic acids. There are conflictin~ reports on the stability of duplexes for~ed by carbamate oligomers and complementary nucleic acids. A carbamate-linked oligomer containing six thymidine units does not form complexes with either A(pA)s or dA(pA)5. On the other hand, a carbamate-linked oligomer containing six deoxycytosine units forms stable complexes with d-(pG) 6 and poly(dG).
The internucleoside linkage of dialkyl- or diphenylsilyl oligomer analogs closely resembles the tetrahedral geometry of the normal phosphodiester internucleotide bond. The oligomers are prepared in solution by reacting a suitably protected nucleoside-3~-O-dialkyl- or diphenylsilyl chloride or trifluoromethanesulp~onyl derivative with a 3'-protected nucleoside in anhydrous pyridine. The former can be prepared by reaction of 5~-O-trityl nucleoside with dialkyl- or diphenyldichlorosilane or with the bis(trifluoromethan~sulphonyl)diisopropylsilane.
Because the dialkyl- and diphenylsilyl linkages are sensitive to hydrolysis by acid, care must be taken in choosing protecting groups ~or the synthesis. Nucleoside dimers and hexamers having siloxane internucleoside linkages and a method of synthesizing such polymers have been reported by Ogilvie and Cormier. See, e.n., Ogilvie, K.X. and Cormiar, J.F., Tetrahedron Letters, 26(35):4159-4162 (1985); Cormier J.F. and Ogilvie, X.K., Nucleic Acids Research, 16(10):4583-4594 ~1988).
Although the carbonate, carbamate and silyl linked oligonucleoside sequences have the requisite nuclease resistance to make them attrActive candidates as antisense reagents, their ability to function in t~is :, :,- - :

WO ~2/0253q PCI`/US91/OSS31 r. ,, ~
r . ~

-- 7 ~

capacity has not yet been reported. Further, the abllity of these oligomers to be taken up by cells in culture has not been reported. A potential drawback with these oligomers is their reported low solubility in aqueous solution. It ls not clear whether sufficient concentrations can be obtained or their effective use in biological experiments, although so;ubility could presumably be increased by introduction of hydrophilic groups into the molecules.
The present invention provides oligonucleotide analog compounds, compositions comprising such compounds, intermediatas for preparing such compounds and methods for synthesizing such novel stable, nuclease resistant, target specific, lipid soluble 15 oligonucleotide analogs.

8UmmarY o~ the ~nvention The present invention provides nucleotide analog compounds comprising oligonucleoside seguences of 20 from about 6 to about 200 ~ases having a three atom internucleoside linkage. ~he three atom internualeoside linkage of such oligonu~leoside sequences has the formula:
-D-D-D-25 where each D is independently ~HR, oxygen or NR6, wherein ~ is independently hydrogen, OH, SH or NH2, R6 is hydrogen or Cl-C2 alkyl, with the proviso that only one D is oxygen or NR6, In a pref~rred e~bodiment, the oligonucleoside 30 sequences comprise bases selected from the group consisting of adenine, cytosine, guanine, uracil, thymine and modifications thereof.

..
- .:

' ' , ,:'; `'', ` :
: .. .. :

W092/02~34 PCT/US91105~31 n~; ~7 3 More particularly, compounds of the present invention comprise oligonucleoside sequences of Formula I:

Lw~- ~
, ,_, ,~

~Rl where W is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH or NH2, R6 is hydrogen or C1-C2 alkyl with the proviso that only one D is oxygen or NR's;
each W' is independently W or o Oe each R1 is independently OH, SH, NR~R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl or NHR4 wherein R4 is Cl-Cl2 acyl;
each y is independently H `or OH;
each B is independently adenine, cytosine, gNianine, t~.ymine, uracil or a modification thereof;
j is an integer from l to about 200:
k is o or an integer from l to about 197; and :`
`

,: ~ ~ , . , .:

WO~2/02534 PCTlUS91/0553i 9 _ q is O or an integer from 1 to about 197, with the proviso that the sum of j + k + q is from about 4 to about 200.
The compounds of the present invention comprise oligonucleotide or oligonucleoside sequences optionally having a diol at either or both termini.
Preferred diols are 1,2-diols (glycols).
Representative glycols are polyalkyleneglycols, preferably polyethyleneglycols or polypropyleneglycols.
Preferred glycols are tetraethyleneglycol and hexaethyleneglycol. Suitable diols may also include polyols that have all but two hydroxyls blocked. J
Where the compounds of the present invention are oligonucleoside sequences having a diol at either or both termini, the compounds of the present invention have Formula II:

2 ~

~ ~ II

W ~B

,~: . , , :.
:.

W092/02534 PCT~US91/05~31 f., _ J

where each z is independently R' or O O
Il 11 Rl- ([(CIH)pO]m ~PI~ O)e([(CIH)~ ]n IP 0)~;

where each R1 is independently OH, SH, NHR~ wherein R2 and R3 are independently hydrogen, or C~-C6 alkyl, or NHR4 wherein R4 is Cl~Cl2 acyl:
each Rs is independently hydrogen or Cl-C1. alkyl;
each of W, W', Y, B, j, k, and ~ is as defined above;
each e and f is independently O to 50 with the proviso .
that at least one of e and f be at least l;
each m and n is independently l to 200; and each p is lndependently 2 to 4.
Xn a pre~erred embodiment, the sum of ~ + k q is from about 9 to about 50 baso~s, more preferably from about 12 to about 25 and most preferably from about 15 to about 18. In this embodiment, compounds of this invention comprise oligonucleotid~es of the formula:
O O o Il 11 11 l l R- (t~1H)p]m ~I~ O)C([(lH)pO]n -1- O)~oligo (N)](O -P-[ (CH)pO]n)e(O -P- ~(CH)p]n~
Rl Oe 3S where R is OH, SH, NR2R3 wherein R2 and ~ are independently hydrogen or C1-C6 alkyl, or NHR4 wherein R4 is Cl-Cl2 acyl;
R~ is hydrogen or C1-C12 alkyl;
oligo (N) is a native or modi~ied oligonucleotide sequence of from about 9 to about 2ao bases;
each e and f is independently O to 50, with the proviso that at least one of e and f be at least l;

~ r; ) ~
~,.J

each m and n is independently l to 200; and each p is independently 2 to 4.
In a preferred embodiment, the oligonucleotide contains, in a homopolymer or heteropolymer sequence, S any combination of dA, dC, dG, T.
Where the glycol is polyethyleneglycol, the compounds of this em~odiment comprise oligonucleotides of the formula:
O o R- ~(C~2CHzO)~ -P- o]e[oligo(N)][o -P- (oCH2CH2) n]f -R~
- Oe Oe . ._ .
where R is OH, SH, NR2R~ wherein R2 and R3 are independently hydrogen or C1-C6 alXyl, or NHR~ wherein R~
is C1-C1z aoyl;
oligo N is an oligonucleotide sequence of from about 9 to about 50 bases;
e and f are independently O to 50, with the proviso that at least one of e and f be at least l;
m and n are independently O to 200 with the proviso that at least one of m and n be l to ~00.
T~e oligonucleotides o~' the present invention can include known internucleosid~l lin~ing groups such as phosphodiester, silyl and other well known linking groups providing they contain an effective amount of the -D-D-D- linking groups of the present invention and/or diol terminating groups of the present invention.

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

W~'~2/0253~ PCT/US91/0~31 i~ J _ ;J '~

The present invention is also directed to nucleoside dimers of the formula:

~Y
B
W--<`l ~Y
R

where W is -D-D-D- wherein each ~ is independently CHR, oxygen or NR6 wherein R is independently hydrogen, OH, SH or NH2 ~ R6 is hydrogen or Cl-C2 alkyl, with the proviso that only one D is oxygen or NR6;
each ~ is independently adenine, cytosine, guanine, thymine, uracil or a modi~ication thereof;
R7 is OH, t-butyldimethylsilyloxy or a phosphoramidite and R8 is OH, a protecting group or t-butyldimethylsilyloxy.
The present invention further provides a method of inhibiting nuclease degradation of compounds comprising oligonucleoside sequences. This method comprises attaching a diol to either the 5', the 3' terminus or both termini of said compound. The diols are , : - ~, ~ : ; , . .

W092/0~34 c~ PCT/US91/05~31 ~ G , j '~ _~
~,J`J

attached to the 5' and/or the 3' terminus by reacting the oligonucleotide compounds with an alXoxytrityldiolcyanophosphine, preferably a dimethoxytritylglycolcyanophosphine or a monomethoxytritylglycolcyanophosphine.
The present invention further provides a method of inhibiting nuclease degradation oP native or modified nucleotide compounds comprising preparing oligonucleoside sequences of from about 6 to about 200 lO bases having a three atom internucleoside linkage having the formula -D-D-D- as defined herein.
.
The present invention also provides compositions useful in inhibiting gene expression comprising compounds comprising oligonucleoside 15 sequences of from about 6 to about 200 bases having a three atom internucleoside linkage as defined herein and a physiologically acceptable carrier. The compound may have a diol at either or both termini. Preferred diols are polyethyleneglycols.
The present invention further provides a method of inhibiting gene expression comprising administering to a mammal in need o~ such ~reatment an effective amount of a compound comprising an oligonucleoside sequence from about 6 to about 200 bases 25 having a three atom internucleoside linkage as defined herein. The compounds may h~ve a diol at either or both termini. Preferred diols are polyethalyeneglycols.

Brief Description of the Drawinqs ~igure la depicts a synthetic pathway for preparing a nucleoside aldehyde (Compound I).
Figure lb depicts a synthetic pathway for preparing a phosphonium iodide nucleoside ~Compound II).
Figure 2 depicts a syntpetic pathway ~or 35 preparing nuclaoside dimers connected by a 3 carbon ~ : . . .:,: . :
. : . .

::. : : .: .:: . :
:: .: . ~: . : : , - :- : : ~, ;. ~ :

W0~l2/02534 PCTtUS91/05~31 14 ~

internucleoside lin~age utilizing the aldehyde nucleoside and the phosphonium iodide nucleoside of Figures la and lb respectively.
Figure 3 depicts a synthetic pathway for preparing a thymidine dimer utilizing a thymidine aldehyde and a phosphonium iodidP thymidine (compounds I
and II respectively).
Figure 4 depicts a synthetic pathway for preparing a nucleoside dimer connected by a two carbon-one nitrogen atom internucleoside linkage of the form 3'-C-C-N-5'. Dimers are synthesized by reacting nucleosid~ss that contain aldehydes (CHO) with nucleosides that contain amine functionalities (NH2) undex reductive conditions.
Figure 5 depicts a synthetic pathway ~or preparing a nucleoside dimer connected by a two carbon-one nitrogen atom internucleoside linkage of the form 3l-N-C-C-5'. Dimers are synthesized by reacting nucleosides having aldehyde and amine functionalities under reductive conditions.
Figure 6 depicts a synthetic pathway ~or preparing a thymidine dimer connected by a two carbon-one nitrogen atom internucleoside lin~age of the form 3'-C-C-N-5~. ~imers are synthesized by reacting thymidines that contain aldehydes (CHO) with thymidines that contain amine functionalities (NH2) under reductive conditions.
Figure 7 depicts a synthetic pathway for preparing a thymidine dimer connected by a two carbon-one nitrogen atom internucleoside linkage of the form 3'-N-C-C-5'. Dimers are synthesized by reacting thymidines having aldehyde and amine functionalities under reductive conditions.

: . :

,~

~J J

D~tailed De~criP~on of t~e Invention The compounds of the present invention are generally oligonucleotide or oligonucleoside sequences that are resistant to nuclease degradation.
As used herein, "nucleoside" refers to a combina~ion of a purine or pyrimidin~ base with a five-carbon sugar (pentose).
As used herein, "nucleotide" refers to a phosphoric acid ester of a nucleoside.
As used herein, "oligonucleotide" refers to polynucleotides having only phosphodiester internucleoside linkages, e.g. "native" DNA or RNA.
Exemplary nucleosides are adenosine(A), guanosine(G), cytidine(C), uridine(U), deoxyadenosine (dA), deoxyguanosine(dG), deoxycyt:idine(dC) and thymidine(1').
The compounds of the present invention comprise oligonucleoside sequences of from about 6 to about 200 bases having a phosphodiester or a three atom lnternucleoside linkage. The thr~se atom internucleoside linkag~ ~-D-D-D ) contains l) thr~ae carbon atoms, 2) two carbon atoms and one oxygen atom or 3) two carbon atoms and one nitrogen atom.
$he oligonucleoside sequences are saquences of natiYe or modified nucleosides. As used herein, the phrase "internucleoside linkage" refers to atoms and molecules forming a bridge between the sugar moiety caxbon atom at position 3 of one native or modified nucleoside and the sugar moiety carbon atom at position 5 of an adjacent such nucleoside. The sugar ~oiety may be either a ribose or a deoxyribose moiety or an analog thereof. Thus~ the nucleosides include A, C, G, U, dA, dC, dG, T or modifications thereof as for example 5-bromo or 5-iodouracil, 5-methyl cytosine, isocytosine ~S t2-amino-4-oxopyrimidina~, isoguanine (2-oxo-6-,' ,....

- :
, W092/02534 PCT/US91/~5531 aminopurine), inosine (6-oxopurine), 5-vinyluracil and 5-vinylcytosine.
The three atom internucleoside lin~age has the ~ormula:
-D-D-D-where each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH or NH2, oxygen, R5 is hydrogen or C~-C2 al~yl, with the proviso that only one D is oxygen or NR6.
The compounds of the present invention comprise oligonucleoside se~uences of Formula I:

~ ' ~w . W' ~ ~
~Y

where ~ is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH or NH2, ~6 is hydrogen or Cl-C2 alkyl, with the proviso that only one D is oxygen or NR6;

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

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

.
:.

WO 9~/0'~534 ~ ~ PCr/US91/05531 ~ r ";J ~1 ' ~,, J _, each W' is independently W o- o le each Rl is independently OH, SH, NR~R3 wherein R- and R3 are independently hydrogen or C~-C6 alkyl or N~R4 wherein RJ is C~-CI2 acyl;
each y is independently H or OH;
each B is independently adenine, cytosine, guanine, thymine, uracil or a modification thereof;
j is an integer from 1 to about 200;
k is O or an integer from 1 to about 197; and q is o or an integer from 1 to about 197, wi~h the proviso that the sum of ; + k I q is from about 4 to about 200.
In a preferred embodiment, the sum of j + k +
q is from about 9 to about 50. In a more pre~erred embodiment, the sum of ; + k + q is from about 12 to about 25 and, more preferably from about 15 to about 18.
The compounds of the present invention may have a diol at either or both termini. Pre~erred diols are ~lycols, also known as 1,2-diols, which contain two hydroxyl groups on adjacent carbons. Preferred glycols are polyalkyleneglycols. The term "alkylene" as used herein refers to linear and branched chain radicals having 2 to 4 carbon atoms which may be optionally substituted as herein defined. Representative of such radicals are ethylene, propyle~e, isobutylene, and the like. Preferred polyalkyleneglycols are polyethyleneglycols such as hexaethyleneglycol and tetraethyleneglycol. Suitable diols may also include polyols that have all but two hydroxyls blocked.
The diols are attached to either the 5', the 3' or both termini of the oligonucleosides via phosphodiester linkages. In one embodiment, the diols , .: . . .... . . .

. . . : .:

W092/025~ PCT/US91/05531 - l8 ~ V J ~

are attached to only one terminus of an oligonucleoside sequence.
The terminal diol is linXed to a moiety selected from the group consisting of hydroxyl (OH), sulfhydryl (SH), amino (N~2), alXylamino (NH-alkyl), dialkylamino (N[alkyl].) and amido (NH[acyl]).
Where glycols are present at either or both termini, the compounds of the present invention comprise oligonucleoside seqiuences of Formula II:

~ ~ 1 1 ~ ~ 8 Y

where each Z is independently R' or O O
Il 11 . .
R~- ([(CIH)pO]m -Pl- )~([(ClH)p~O]n-p-o)f;

.. . : , ` : .:
::
; ', . ~ : ' ,: . ;. , , .:

: .......... , : . , -: i ~ . :. ::: .. :: . ::: : :: :: i -where each R~ is independently OH, SH, NHR~R3 wherein R2 and R3 are independently hydrogen or C~-C6 alkyl, or NHR4 wherein R4 is C~-C~2 acyl;
each R5 is independently hydrogen or Cl-C~2 alkyl;
each of W, W', Y, B, j, k, and q is as defined above;
each e and f is independently 0 to 50, with the proviso that at least one of e and f be at least l;
each m and n is independently l to 200; and each p is independently 2 to 4.
In a preferred embodiment, m and n are independently l to 6 and the sum of j + k + q is from ~~~~ ~~ about 9 to about 50. In a more preferred embodiment, ~
the sum of j + k + q is from about 12 to 25, more preferably from about 15 to about 18.
lS In another preferred embodiment, the compounds of the present invention compris~e oligonucleotide sequences of from about 9 to about 200 bases having a diol at either or both termini. In yet another pre~erred embodiment, the compounds of the present invention comprise oligonucleotide sequences of from about 9 to about 200 bases h~ving a (~D-D-D-) linkage of the present invention.
Preferred diols are glycols, also known as l,2-diols, which contain two hydroxyl groups on adjacent carbons. Preferred glycols are polyalkyleneglycols.
The term "alkylene" as used herein refers to linear and branched chain radicals having 2 to 4 carbon atoms which may be optionally substituted as herein defined.
Representative of such radicals are ethylene, propylene, butylene and the like. Pre~erred polyalkyleneglycols are polyethyleneglycols. More preferred are tetraethyleneglycol ar.d hexa2thyleneglyccl.
The diols are attached to either the 5', the 3' or both termini ~f the oligonucleotides via p~osphodiester linkages. In one embodiment, the diols ~ .,: ; .

. . - .
. ~

' ` ;.~, ' :.` , .. ~ : .

WO~2t~2534 PCT/US91/05531 ri r ~
v v rj -" . j are attached to only one terminus of an oligonucleotide sequence.
The terminal diol is linked to a moiety selected from the group consisting of hydroxyl (OH), sulfhydryl ~SH), amino (NH2), alkylamino (NH-alkyl), dialkylamino (N[alXyl]2) and amido (NX[acyl]). As used herein, "alkyl" refers to linear or branched chain radicals having l to 12 carbon atoms which may be optionally substituted as herein defined.
l0 Representative alkyl- and dialkylamino radicals include methyl-, ethyl-, propyl-, butyl-, pentyl-, hexyl-, dimethyl-, diethyl-, dipropyl-, di~utyl-,-dipentyl- and dihexylamines and the like. As used herein, "NH(acyl)"
or "amido" refers to linear or ~ranched chain radicals 15 having 1 to 12 carbon atoms with a termi~al 0-CNH2 group. Representative amido radi.cals ~nclude methanamide, ethanamide, propana~lide, butanamide, pentanamide, hexanamide, heptanan~ide, octanamide, nonanamide, decanamide, undecanamide and dodecanamide.
In one embodiment, the compound~ o~ the pre~ent invention compri~e oligonucleotides o~ the formula:
O o o Il 11 . `11 R ([(CIR)pO]m ~1~ )e(t(cl~)po~n -Pl- o)f[olig~ ~N)~(O -P1-t ~fH)pO~n) ,~ - I-- t(CH~p]m) f R;
R1 Oe 3 S where R is OH, SH, NR2R3 wherein RZ and R3 are independently hydrogen or C1-C6 alkyl, or NHR~ wherein R~
is C~-C12 acyl;
R~ is hydrogen or C1-C12 alkyl;

:.; :

:: :: , . . , - .
- . , .

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

J ~

oligo (N) is a native or modified oligonucleotide sequence of from about g to about 200 bases;
each e and f is independently O to 50;
each m and n is independently 1 to 200; and each p is independently 2 to 4.
The oligonucleotide sequence is preferably a homopolymer or heteropolymer sequence containing any combination of dA, dC, dG, T or analogs thereof.
In a preferred embodiment, m and n are independently 1 to 8 and, more preferably, both m and n are 4. Preferred oligonucleotide sequences contain from about g to about 50 bases, more preferably about 12 to about 25 hases, and most preferably about 15 to about 18 bases.
In a preferred embodiment, the antisense compounds have polyethyalkyleneglycol at both the 5' and 3' termini and have the formula:
Il I O
R ~(CIH)pO]m -Pl- O)~t~(CIH)po]n -1- O)f~oligo (N)](O -X-25 . Il [ (CH) p]n) e~ ~ I ~ ~0 (CH) p]~) t -R
Rl Oe where R is OH, SH, NR2~3 wherein R2 and R3 are independently hydrogen or Cl-C6 alXyl, or N~R~ wherein R4 is Cl-Cl2 acyl:
R1 is hydrogen or Cl~C12 alkyl;
oligo (N) is~a native or modified oligonucleotide sequence of from about 9 to about 200 bases;
each ~ and f is independently 1 to 50;
each m and n is independently 1 to 200; and each p is independently 2 to 4.

:
~ .

~ .-- : .
. ;: : .
:: :
.

W092/025~ PCT/US91/05~31 r l ~

Where the glycol is polyethyleneglycol, the compounds o~ this embodiment comprise oligonucleotides of the formula:
O O
R- t(CH2CH20)~ -P- O~[oligo(N)~0 -P- tOC~2CH2)n], -R:
Oe Oe where R is 0~, SH, NR2Ri wherein R2 and R3 are independently hydrogen or C~-C~ alkyl, or NHR~ wherein R~
iS C~-C~2 acyl;
oligo N is an oligonucleotide sequence of from about g to about 50 bases; and e, f, m and n are each independently 1 to 50.
In a preferred embodiment, the oligonucleotide contains, in a homopolymer or heteropolymer sequence, any combination of dA, dC, dG, T.
In other preferred embodiments, the polyethyleneglycol is tetraethyleneglycol (TEG) and both m and n are 4 or hexaethyleneglyc:ol and both m and n are 6.
The compounds o~ the p~esent invention are u~e~ul as anti3ensQ a~Qnts. AntlqensQ agents hybridize ~5 with a complementary nucleotide ~;equence in a target nucleic acid to inhibit tha translational or transcriptional function of said target nucleic acid.
The target nucleic acid may be either RNA or DNA.
Antisense compounds of the present invention comprise oligonucleoside sequences of from about 6 to about 200 bases having homopolymer or heteropolymer sequences comprising bases selected from the group consisting of adenine (A), cytosine ~C), guanine (G) ` uracil (U), thymine ~T) 2nd modific~tions of these bases. Particular sequences are selected on the basis o~ their desired target. The sequence selected hybridi~es with the targe~ nucleic acid. Exemplary :: ,-: : ;. , . :: :
:: :
. : . ~ ;.; .. , :,;

WO g2/02534 PCr/US91/0553 t~rgets include the MYC oncosena, the RAS oncogene, and viral nucleic acids.
The compounds of the present invention can be prepared by the following procedures:
~. Com~ounds havina a three_carbon internucleoside linXaqe Oligonuclessides connected by a three-carbon internucleoside linkage are synthesi2ed by reacting nucleosides having aldehyde and ylide functionalities at 3' and 6' positions respectively under Wittiq conditions.
The syntheses of a nucleoside aldehyde and a phosphonium iodide nucleoside from commercially available compounds are illustrated in Figure la and lb, respectiva:Ly. The aldehyde ~Compound I from Fiqure la) i5 synthesized ~rom the known 3'-i~llyl-3'-deoxy-5'-0-tert-butyldimethylsilyl-3'-thymid,ine ~Compound A, Figure l). The allyl compound is regioselectively o~idized with a catalytic amount of osmium tetroxide and N-methylmorpholine oxide as a cooxidant. The resultant diol (Compound B, Figure la) iq cle~ved with sodium ~eriodate to give the aldehyde in almost quantitative yield.
~he synthesis of the phosphoniu~ iodide nucleoside (Compound II, Figure lb) follows from a commercially available 5'-tritylated nucleoside (Compound C, Fiqure lb). The tritylatad nucleoside is silylated at the 3' position with tert-butyldimethylsilyl chloride and the trityl group removed under acidic conditions with high effioiency. The resultant primary hydroxyl (Compound E, Figure lb) is oxidi~ed under Swern Gonditions to give the aldehyde (Compound F, Figure lb). The crude aldehyde is "~
immediately reactad with the ylide derived fro~
methyltriphenylphosphonium bromide to give a 4'-vinyl-. , : -. . i : , , "~
4 P~T/US91/05531 ~ ~ n ~
24 _ ~ ~ v ~

4'-deoxy-3'-tert-butyldimethylsilyl nucleoside in good yield. The vinyl compound i-~ regioselectively hydroborated to give a primary alcohol (Compound G, Figure lb) in good yield. The primary alcohol in turn is converted to the corresponding iodide (Compound ~, Figure lb) using triphenylphosphine-iodine in the presence of imidazole in excellent yield. Finally, the iodide is transformed to the desired phosphonium iodide nucleoside using triphenylphosphine in acetonitrile.
A ylide is prepared from the phosphonium iodide nucleoside using potascium tert-butoxide as a base and immediately reacted with the aldehyde to give-a~
Wittig product (Compound 1, Figure 2) in good yield.
The Wittig product is regioselectively hydrogenated with 10% palladium on carbon ~10~ Pd-C) with hydrogen at atmospheric pressure ln quantitative yield to saturate the double bond of the linkage. The saturated compound (Compound 2, Figure 2) ls desilylated with tetrabutylammonium fluoride to gi.ve the diol tCompound 3, Figure 2~. Tha 5'-primary hydlroxyl of the diol is then rQgioselectively protectQd with dimethoxytrityl chloride and the resultant 3'-hydroxyl (Compound 4, Figure 2) is converted to a phosphoramidite (Compound 5, Figure 2) with 2-cyanoethyl-N,N-diisopropylchlorophosphoramidite.
The nucleoside dimers or hiyher oligomers with trialkylsilyloxy protecting groups are conjugat~d to form oligonucleotides of any desired length. Upon completion of chain elongation, the oligomers are deprotected by standard methods. For furt~er chain length extension in a solid phase synthesizer in which the oligomers are connected by phosphate linkages, the terminal 5'- and 3'-hydroxyl groups of the oligomers are appropriately functionalized, raspectively with .

.: . :: . ~.. . . . -; .

W092/025~ PCT/US91/05~31 ~ J~
- 25 ~

tritylating reagents such as dimethoxytritylchloride and phosphoramidite.
B. ~ompounds havinq a two carbon-one ox~gen atom inter~ucleoside linkaqe Oligonucleoside sequences having ~ two carbon-one oxygen atom internucleoside linkage are synthesized by reacting 3'-silylated, 5'-toluenssulfonyl nucleoside with a 5'-protected nucleoside.
A 3'-acetyl-5'-aldehyde nucleoside is prepar~d from a commercially available 3'-acetyl-nucleoside using standard methods well known to those of skill in the -art. The 3'-acetyl-~'-aldehyde nucleoside is then ~~~~ ~ ~~
converted to a 3'-acetyl-5'-carbomethoxymethylene nucleoside using a modified Wittig reaction.
The 5'-methylene side chain is reduced with sodium borohydride in alcohol, preferably isopropanol, followed by deprotection of the l'-acetyl group with sodium methoxide in an alcohol, preferably methanol.
The 3'-hydroxy ~s then protected with a silyl group. In A preferred embodiment the silyl group i5 a t~bu~yldim~thylsilyl group.
The 3'-silyl-5'-carbomethoxymethyl nucleoside is then further reduced to a 3'-0-silyl-5'-deoxy-3'-(2 "-ethanol) derivative of the nucleoside with diisobutyl aluminum hydride (DIB~L) in tetrahydrofuran (THF). The 5'-ethanol group is converted to ~ p-toluenesulfonyl group with p-toluene sulfonyl chloride in pyridine. The exocyclic amino group of the base moiety of the 5'-p-toluenesulfonyl nucleoside is optionally protected by methods well Xnown and readily apparent to those of skill in the art. A preferred protecting group for '.hr exocyclic amino groups of adenine and cytosine is the benzoyl moiety. A preferred protecting group ~or the exocyclic amino group of :
.

.~ ., .. . ~.

W0~.~/02534 PCT/US91/05531 ~ ~ v ~

guanine is the isobutyl moiety. Guanine may also be protacted at the o6 posi.tion.
The 3'-0-silyl-S'-0-p-toluenesulfonyl nucleoside is then reacted with a 5'-protected S nucl~oside to ~orm a ~-0-silyl-S'-protected nucleoside dimer with a two carbon-one oxygen atom internucleoside linkage. The 5'-0-prot~ecting group is preferably a trityl and, more preferably a dimethoxytrityl. The 3'-0-silyl-5'-0-protected nucleoside is optionally protected at the exocyclic amino groups of the nucleoside base moiety.
- The nucleosid,e dimers are`~deprotected and rederivatized at the 3'-carbon atom position with a cyanophosphine reagent, preferably 2-cyanoethoxydiisopropylaminophosphine for use in a phosphoramidite solid p'hase synthesis method of chain elongation. Gait, su~ra.
The nucleosid,e dimers or higher oligomers with trialkylsilyloxy protecting groups are conjugated to form oligonucleosides oE any desired length. Upon completion of chain elol~gation, the oligomers are deprotected by standard ~ethods. For further chain length extension in a solid phase synthesizer in which the oligomers are conne,:ted by phosphate linkages, the ~ terminal 5' and 3' hydroxyl groups of the oligomers are appropriately functionaLized, respectively with tritylating reagents su~:h as di~ethoxytritylchloride and j ~hosphoramidite.
~ C. Compounds havinq a two carbQn-one 30 ~ n~qen atom internucleoside_linkage Oligonucleoside sequences connected by a two c~rbon-cne nitrogen atQ~ internucleoside l~nkage o~" the form C-C-N are synthesi;~ed by reacting nucleosides that j contain aldehydes with nucleosides that con~ain a~ine : ~ - ~ , ", '~ ' ' . ~ ;' ' : . : ' ... , : .
. , ~ .

W092/025~ PC~/US~1/05531 ~. , _ functionalities under reductive conditions as illustrated in Figure 4.
Both the aldehyde and the amine compounds are prepared from commercially available compounds. The 5 aldehyde is prepared from 3'-allyl-3'-deoxy-5'-O tert-butyldimethylsilyl thymidine. The allyl compound is regioselectively oxidized with a catalytic amount of osmium tetroxide in the presence of N-methyl morpholine,N-oxide as a cooxidant to give the diol. The diol is, in turn, oxidized with sodium periodate to give the aldehyde in almost quantitative yield.
The amine compound is synthesi2ed from `'~'~
commercially available nucleosides. In a typical procedure, the primary hydroxyl group of a nucleoside is regioselectively transformed into a tosylate group with p-toluene sulfonyl chloride and then converted into an iodide. The 3'-hydroxy of the iodide intermed'iate is protected with tert-butyldimethylsilyl chloride and the azido group introduced by reactirlg with sodium azide.
The azido functionality is efficiently converted to the required amine by reduction usinsl 10% palladium on carbon under a hydrogen atmosphere or Raney Nickel reduction conditions.
The amine and the aldehyde are coupled (reductive amination) in the presence of sodium cyanoborohydride under buffered conditions. The oligonucleoside dimer with a C-C-N internucleoside linkage is formed in good yield. The oligonucleoside is reacted with trifluoroacetic anhydride-triethylamine, to protect the secondary aliphatic nitrogen. The protected oligonucleoside is desilyla~ed with tetrabutylammonium fluoride and the primary hydroxyl group of the resultant diol is selectively protected with dimethoxytrityl chloride. The remaining secondary hydroxyl is .: , . :
. . .
.. ~
.
.

W O 92/02S34 PC~r/US91/05531 ~ ~ ~! r.

transformed to the required phosphoramidite by reacting with 2-cyanoethyl-N,N-diisopropylchlorophosphoramidite.
Oligonucleosides connected by two car~on-one nitrogen atom internucleoside linkage of the form N-C-C
are synthesized by reacting nucleosides having aldehyde and amine functionalities at 3'- and 51_ positions, respectively, under reductive conditions as illustrated in Figure 5, The amine and the aldehyde components are synthesized from commercially available compnunds. The amine is synthesized from 3-azido-3-deoxy thymidine (AZT). The primary hydroxyl group of AZT is protected with dimethoxytritylchloride and the resultant azide regioselectively transformed to the required amine with 10~ palladium on carbon in the presence of a hydrogen atmospherQ or using Raney Nickel.
The aldehyde is synthelsized from commercially available 5'-O-dimethoxytritylth~ymidine. The tritylated thymidine i5 silylated with tert-butyldimethylsilyl chloride and the trityl group is removed under acidic conditions. The resultant primary hydroxyl group is oxidized under Swern conditlons to give tho aldehyde.
The aldehyde is not isolated but immediately reacted with (carbethoxymethylene)triphenylphosphorane to give the unsaturated ester. The unsaturated ester is regioselectively hydrogenated with 10% palladium on carbon to give a saturated ester in quantitative yield.
The saturated ester in turn is converted to the required aldehyde with diisobutyl aluminum hydride ~DI~AL-~) in a highly selective manner.
The amine and the aldehyde are coupled in the presence o~ sodium cyanoborohydrid2 under buffered reductive amination conditions. The N-C-C
internucleoside linkage is obtained in good yield. The secondary aliphatic nitrogen of the oligonucleoside is ., .. .. ,, - . -. .. ,., : : ~ : - , - ~ - . .- : . . .

-, . . -. . : .. .: , . .

WO')2/02S3'1 PCT/US91/0~531 protected with trifluoroacetic anhydride and triethylamine. The protected dimer or higher oligonucleoside sequence is desilylated and the resultant hydroxyl converted to phosphora~idite with 2-cyanoethyl-N,N-diisopropyl-chlorophosphoramidite.
The nurleoside dimers or higher oligomers with trialkylsiloxyl protecting groups are conjugated to form oligonucleosides of any desired length. Upon completion of chain elongation, the oligomers are desilylated by standard methods. For further chain length extension in a solid phase synthesizer in which the oligom~rs are conne~ted by phosphate linkages, the terminal 5' and 3' hydroxyl groups of the oligomers are appropriately ~unctionalized, respectively with tritylating reagents such as dimethoxytritylchloride and phosphoramidite.
D. ~ompounds havinq a diol at çither or ~oth WherQ desired, diols are attached to either or both termini by a modi~ication of the solid phase phosphor~midi~e method. Q~ cl~o~idq Svn~h~si~: A
Practical ~roach, ed. by M.J. Gait, pages 35-81, IRL
Press, Washington, D.C. tl384).
~n accordance with our modification of the solid phase method, a diol is introduced at one, or both, terminalts) of the oligon~cleotide by a procedure in which the diol is reacted with an alkoxytrityl compound to ~orm a tritylated diol. The diol is preferably a glycol, more preferably, a polyalkyleneglycol. The alkoxytrttyl reagent is preferably monomethoxytrityl chloride or dimethoxytrityl chloride and, most preferably dimethoxytrityl chloride.
The tritylated diols are ~hen reacted with a cyanophosphine reagent to form a trityldiolcyanophosphine compound, which compound is used as a phosphoramidite reagent (hereinafter referred W092/02534 PCT/U~91/05531 J,~

to as a "diol phosphoramidite reagent") in the solid phase synthesis of the compounds of the present invention.
The initial step in solid phase synthesis is attachment of a nucleoside to a solid support, preferably a controlled pore glass (CPG) support. The nucleoside is preferably attached to the CPG via a succinate linkage at the 3'-hydroxyl position of the nucleoside. Other means of attaching nucleosides to solid supports are known and readily apparent to those of skill in the oligonucleotide synthesis art.
Alternatively, in order to introduce a diol ~t the 3' terminal, a diol phosphoramidite reagent can be attached to the solid support prior to addition of the first nucleoside. The diol phosphoramidite reagent i~
attached to the solid support using succinatls or other linkages in a manner analogous to methods used for nucleoside attachment. Means of modifying such methods for use with diol phosphoramidite reagents will be readily apparent to those o~ skill in the art. Any num~er of diols can be placed on the solid support prior to add~tion of the fir~ nucleo~,ide. Preferably ~rom 1 to about 50 diols are used. Whe!re diols are attached only to the 5' terminus, no diols are placed on the solid support.
Following attachment of the first nu~leoside ~r the diol~s) to the solid support, chain elongation occurs via the sequential steps of removing the 5'-hydroxyl protecting group (a functionalized trityl group), activating the 5'-hydroxyl group in the presence of a phosphoramidite reagent, i.e., a 5'-trityl nucleoside, 3'-phosphoramidite, capping the unraactad nucleosides and oxidizing the phosphorous linkage.

::-: : ., ;. . .:

W0~2/02534 P~T/US9t/05531 ,~r;~ 31 -J ~ .
The protecting group at the 5'-hydroxyl position of the attaohed nucleosides is removed with acid, preferably trichloroacetic acid.
Activating reagents that can be used in accordance with this method are well known to those of skill in the art. Preferred activating reagents are tetrazole and activator gold (BecXman Instr. Inc., Palo Alto, CA).
The activation step occurs in the presence of the added nucleoside phosphoramidite reagent or diol phosphoramidite reagent, which latter reagent replaces the nucleoside phosphoramidite reagent of conventional synthetic methods when diol is added to the terminal(s) of the polynucleotide. Unreacted chains are terminated --`
or capped with capping reagents such as acetic anhydride and N-methyl imidazole.
The labile trivalent phosphorus linkage is oxidized, preferably with iodine, to the stable, pentavalent phosphodiester lin~age of the oligonucleotide.
After the desired oligonucleotide chain assembly is complete, the phosphalte protecting groups are removed, the chains are separated ~rom the solid support and the base protecting groups are removed by conventional methods. Gaits, su~ra at 67-70.
T~ose skilled in the art will appre~iate that other means of synthesizing oligonucleotides can be modified in an analogous manner to produce diol-terminated antisense oligonucleotides.
The compounds of the present invention are use~ul in treating mammals with heredit~ry disorders or diseases associated with altered genetic expression mechanisms. At present, attempts are underway to develop antisense therapies for use in treating viral infections such as ~IY, cytomegalovirus, herpes simplex, ` ' , . . ~ ,:
'' ', ~ :

:
' `

W092/025~ PCT/US91/OS~31 ~ ~ ~ ' ;' `3 hepatitis ~, papilloma virus and picorna virus; cancers of the lung, colon, cervix, breast and ovary;
inflammatory diseases; and diseases of the i~mune system such as acquired immunodeficiency syndrome (AIDS), hematological neoplasma and hyperproliferative disorders. Armstrong, supra at 89; Klausner, supra at 303, 304.
Compositions of the present invention useful in inhibiting gene expression comprise physiologically acceptable carriers and 1) compounds comprising oligonucleoside sequences of from about 6 to about 200 bases having an internucleoside linkage of the ~ormula -D-D-D- as defined herein, optionally having a diol at either or both termini or 2) compounds comprisinq oligonucleotide sequences of from about 9 to about 200 bases having a diol at either or both termini.
Compositions of the pr~sent invention useful in inhibiting genQ expression include one or more of the compounds of this invention formulated into compositions together with one or more non-toxic physiologically acceptabl~ carriers, ad~uvants or veh~cles which are collectively ra~erred to herein ~Ig carriers, ~or parenteral injection, for oral administration in solid or liquid form, for rectal or topical administration, and the like.
The compositions can be administered to humans and animals either orally, rectally, parenterally (intravenously, intramuscularly or subcutaneously), intracisternally, intravaginally, intraperitoneally, locally ~powders, ointments or drops), or as a buccal or nasal spray.
Compositions suitable ~or parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders for reconsti~ution into ,. ~ , , ~ , , , :
~ .. . .. . ... .

W~2/0253~ PCTiUS9t/05~31 sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, qlycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispensing r . _ . _ _ . . .
agents. Prevention of the action o~ microorganisms can be ensurecl by various antibacterial and antifungal lS agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride and the like. Prolonged absorption of the in~ectable form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate ~nd gelatin.
~ If desired, and for more effective distribution, the compounds can be incorporated into slow release or targeted delivery systems such as 2~ polymer matrices, liposomes, and microspheres. They may be sterilized, for example, by ~iltration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable mediu~ immediately be~ore use.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
In such solid dosage forms, the active compound is admixed with at least one iner~ customary excipient (or W(~2/025~ PCT/US91/05~31 ~ ,7,~

carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia, (c) humectants, as for example, glycerol, (d) disintegr~ating ag~nts, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary - ammonium compounds, (g) wetting agents, as for example, cetyl alcohol and glycerol monostearate, (h) adsorbents, as for example, kaolin and ben~onite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol~, sodium lauryl sulfate or mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also compriss buffering agents.
Solid compositions of a similar type may also be employed as fillers in ~o~t and hard-~illed gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like.
Solid dosage ~orms such as tablets, dragees, capsules, pills and granules can be prepared with coatings and shells, such as enteric coatings and others well known in this art. They may contain opacifying agents, and can also be of such composition that they ~0 release ~he active compound or compounds in a certain part of the intestinal tract in a delayed manner.
~xamples of embedding oompos~tions which can ~e used are polymeric substances and waxes.

- : . ~, : `: .

W0~2/02534 PCT/US91/05531 The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
Liquid dosage forms ~or oral administration include physiologically acceptable e~ulsions, solutions, ;
suspensions, syrups and elixir~. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubiliz~ng agents and emuls$fiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, l,3-butyleneglycol, . .-dimethylfo:rmamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan or mixtures of these substances, and the like.
Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, ~lavoring and per~umin~ agents~
Suspensions, in addition to the active compounds, may contain suspendin~ agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahy~roxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the lika.
Compositions for rectal administrations are pre~erably suppositories which can be prepared ~y ~ixing the compounds of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body Wo~2/n2s34 PCT/US91/05~31 ~ v - 3~ -temperature and therefore, melt in the rectu~ or vaginal cavity and release the active component.
Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays and inhalants. The act$ve component is admixed under sterile conditions with a physiologically acceptable carrier and any needed preservatives, buffers or propellants as may be required. Ophthalmic formulations, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.
The compounds of the present invention can also be administered in the form of liposomes. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medlum. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present co~po~itions in liposome ~orm can contain, in addition to the lipoxygenase inhibiting compounds o~ the present inven~ion, stabilizers, preservatives, excipients, and the like. The preferred lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and synthetic.
Methods to form liposomes are known in the art. See, for example, ~ethods_in Cell ~ioloov, Ed. by Prescott, Volume XIY, Academic Press, New Yor~, N.Y., p. 33 et seq., (l976).
Actual dosage levels of active ingredient in the compositions of the present invention may be varied so as to obtain an amount of active ingredient that is effective to obtain a desired therapeutic response for a particular co~position and method of administration.
The selected dosage level therefore depends upon t~e '`~ `' , ' ' ' ~
,.,: ,,, :. : :

W092/02534 PCT/US91/05~31 , . . ~
,.,~'j,J~) I
- 37 - .

desired therapeutic effect, on the route of administration, on the desired duration of treatment and other factors.
The total daily dose of the compounds of this invention administered t~ a host in single or d$vid2d doses may ~e in amounts, for example, of from about 1 nanomol to about 5 micromols per kilogram of body weight. Dosage unit compositions may contain such amounts or such submultiples thereof as may be used to make up the daily dose. It will be understood, however, that the specific dose level ~or any particular patient will depend upon a variety~of factors including the ~ody weight, general health, sex, diet, time and route of administration, rates of absorption and excretion, combination with other drugs and the severity o~ the particular disease being treated.
The following examples further illustrate the best mode of carrying out the invention and are not to be construed as limiting of the s;pecification and claims in any way.

EXAMPLE 1: PreP~ration o~ 5~?'-dimeth~xYtri~Yl-3-0'-t-butyldimethyls~Lyl thvmidinç.
Dimethoxytrityl thymidine (5.0 g, 9.2 mmol) and imidaz~le ~1.2 g, 18.4 mmol) were dissolved in 15 ml of anhydrous dimethyl ~ormamide (DMF) and added to tert-butyldimethyl~ilyl chloride (1.7 g, 11.5 mmol).
The reaction mixture was stirred for 4 hours at room temperature, diluted with ethyl acetate and washed with water, saturated sodium chloride and dried with sodium sulfate. A quantitative yield of the title compound was obtained.

:. ''' .. : . : , W092/02534 PCT/US~1/0~31 ~ ~ ., u i 3 EX~MPLE 2: reparation of 3'-Q-t-butyldimethylsilvl thvmidine, 5'-0-dimethoxy-3' 0-t-butyldimethylsilyl thymidine prepared according to the method of Example 1 (O.7 g, 1.1 mmol~ was treated for 1 hour at room temperature with 13 ml of a 3S trichloracetic acid in methylene chloride solution. The reaction mixture was then neutralized with a 5~ (w/v) sodium bicar~onate solution. The organic layer was dried with sodium sul~ate. The title compound was purified by flash chromatography using a 0 to 30~ gradient of ethyl acetate in methylene chloride. The yield of-the-reaction was 85%.

EXAMPLE 3: Preparation of 3'-0-t-butyldilnethylsilyl ~hvmidine ~' aldehvde.
To a well stirred solution of dry ~ethylene chloride at -78-C was added oxalyl chloride (33.0 mmol, 2.88 ml) followed by dropwise addition of DMS0 ~3.12 ml, 4.4 mmol). After 10 minutes, the alcohol (5.6 g, 15.7 mmol), prepa~ed according to the met~od o~ Example 2, in `20.0 ml o~ CH~C12, was add~d dropwise over a period o~ 2 minutes and stirring was continued for 45 minutes. Et3N
(8,1 ml, 58.1 ~mol) was added and stirring continued for another 45 minutes. The reaction mixtur~ was then brought to room temperature and then washed with water ~2 X 10 ml) followed by brine ~10 ml) and dried (Na2SO4). The crude aldehyde was used for the next step.
ExAMæLE 4: Pre~aration of 5'-vinyl-5'-deoxy~3'-t-butyldimethYlsil~l deoxy~hy~idinÇ~
To a solution of methyltriphenyl phosphonium bromide ~O.7 mmol) in dry tetrahydrofuran (THF) at 0~C
was added a solution of sodium bis :" '. "~ -,,', " :

W092/025~ PCT/~S91/0~31 (trimethylsilylamide)(0.6 ~mol~ dropwise. After 30 minutes, a solution of the corresponding 4'-aldehyde in THF was added dropwise under nitrogen. The reaction mixture was stirred for 2 hours, diluted with ethyl acetate, washed with water, ~hen with brine and dried (Na2S0~). The title compound was purified by fl~sh chromatography using 20% ethyl acetate-hexane. The yield was 55-60~.

EXAMPr~ 5: E~paration of 3'-0-t-butYldimethylsilYl-5'-deoxy-5'-hYd~oxvmethyl thymidine, -To a solution of 2M 2-methyl-2-butene (1.6 eq, 1.5 ml, 3 mmol) in 3 ml of anhydrous THF at O-C, 1.6 eqs-of a lM borane-tetrahydrofuran complex t3 ml, 2 mmol) were added slowly under N2.
The solution was stirred for lO minutes followed by the addition of the vinyl thymidine, prepared according to the method o~ Example 4, ~0.7 g, 1.9 mmol) in 5 ml of anhydrous T~F. The reaction mixture was stirred ~or 45 minutes and placed in the re~rigerator for 2 days.
W~rkup was done using a,n aqueous ~olution comprising ~.1 eq of 2M sodium hy~droxide and 3.1 eq of 30% hydrogen peroxide (preferably adding hydrogen peroxide dropwise to the aqueous sodium hydroxlde at O-C
and stirring for 10 minutes). The solution was added slowly through an addition funnel to the reaction mixture at 3'C, stirred for 1 hour, removed from the ice bath, diluted with ethyl acetate, washed with water, satura~ed sodium chloride and dried with sodium sulfate.
The title compound was purified by flash chromatography using a 20-B0% gradient of ethyl acetate in hexane. The yield was 62~.

:,.,., , :: .:
' . :`
. . . :

:. : .
... . . ~i, wo g2/02s3~ Pcr/

EXAMPLE 6: ~Feparation of ~'-iodomethYl-$'-deo~x~
3'-O-t-butvldimet~vlsilYl thvmidine.
To a solution of 3'-0-t-butyldimethylsilyl-5'-deoxy-5'-hydroxYmethyl thymidine prepared according to the method of Example 5 (O.3 g, O.9 mmol) in dry acetonitrile (5 ml) and ether (3.4 ml) were added 3 eq of triphenyl phosphine (0.7 g, 2.8 mmol), 4 eq ~f imidazole (O.3 g, 3.7 mmol) and 2.2 eq of iodine (O.5 g, 2.8 mmol). The reaction mixture was stirred for 45 min and the solvent was evaporated. Ethyl acetate was added to the residue and the--residue washed with water, -- ~
saturated sodium chloride and dried with sodium sulfate.
The title compound was purified by flash chromatography using a 30-50% gradient of ethyl acetate in hexane. The yield was 90S.

EXAMPLE 7: Preparation o~ 3'-0-t-butYldimethvls~lvl-5'-deoxY-5'-~hYmildyl me~hvl ~hosphonium iQ~i~Ç~
~o a stirred solution of 5'-iodomethyl-5'-deoxy-3~-0-t-butyldimethysilyl thymidlne prepared according to the method of Example 6 was added iodide t480 mg, 1 mmol) in dry CH3CN ~5 ml) and triphenylphosphine (1.57 g, 6 mmol) and the mixture re~luxed for 12 hours at 90-C. ~he reaction w~s cooled and the solvent was removed. The title compound was puri~ied by flash chromatography using 5% MeOH in C~2Cl2. The product was obtained in 95-96% yield.
EXAMPLE 8: PreParation of 5'-t-butyldimethYlsi 3~-deoxv-3~ .2~-dihYdroxy-3n-pr thvmidine.
Osmium tetraoxide ~OsO~) (4 drops, 2.5 w/v%) in butanol was added to a stirred mixture of 3' (2"-W092/02534 PCT/US91/05~31 ~ 41 -propenyl)-3'-deoxy-5'-0-t-butyldimethylsilyl thymidine prepared according to the procedure described in ~._or~.
.Chem. 1989, 54:2767-2769 tC~X. ~hu et al.) (183 mg, 0.5 mmol) and 4-methylmorpholine N-oxid~ (53 mg, 0045 mmol) in 5.0 ml anhydrous THF at O-C. The react~on ~ixture was then quenched with 10~ agueous sodium metabisulfite (2.0 ml), stirred for 20 minutes, filtered over a pad of silica and diluted with ethyl acetate (25.0 ml). The organic phase was washed with water (5.0 ml) and brine, and then dried with Na~SO~. The solvent was evaporated and the title compound purified by flash chromatography.

EXAMPLE 9: Pre~aration of 5~-o-t-but~ldimethylsi 3'-deoxY-thymid-3'~1-acetaldehYde.
lS Sodium periodate (214 mg, 1 mmol) was added to a stirred solution of the thymidine diol prepared by the method of Example 2 (200 mg, 0.5 mmol) in THF-H20 (4:1 ratio, 5.0 ml). After 1 hour, khe reaction mi~ture was diluted with ethyl acetate (25 ~1), washed with H20 (2 x 5 ml), brine and dried. The title compou~d was purified by flash chromatography with 70% ethyl acetate in h~xane.

EXAMP~E 10: Pre~aration of thYmidine dimers with a three carbon internucleoslde li~kaqe.
The processes of example lOa-lOe are illustrated in Fi~lre 3.
lOa. T~ a stirred suspension of the phosphonium iodide compound prepared according to the method of Example 7 ~241 mg, 0.32~ ~mol) in dry THF (2.0 ml) was added potassium tert-butoxide (0.62 ml, 1.0 M
solution in THF, 0.62 mmol) at 78-C under nitrogen.
After 20 minutes, the 3~-acetaldehyde compound prepared according to the method of Example 9 (80 mg, 0~22 mmol) was added. After 60 ~inutes, tha reaction mixture was : , .

W092/0253~ PCT~US91/0~31 ~ J ~ '1.3 J -~

diluted with ethyl acetate (30 ml), washed with watar (2 x 5 ml) and brine (5 ml) and dried (Na2S0~). The solvent was evaporated and the ol~fin product (Compound 1) was puri~ied by flash chromato~raphy using 70% ethyl S acetate in hexane. The yield was in the r~ngo of 55-60%.
lOb. 10% Pd-C (20 mg) was added to a stirred szlution of Compound 1 ~109 mg) in methanol ~S.0 ml) at 25'C and at 1 atmospheric pressure of hydrogen. After 4 10 hours, the catalyst was filtered over a pad of celite and the solvent was evaporated. Compound 2 was thus recovered and then purified by flash chromatography in 80% ethyl acetate in hexane.
lOc. About 2.8 equivalents of tetra~utyl 15 ammonium fluoride at O'C were added to a stirred solution of Compound 2 ~350 mg) in 5.0 ml of THF. After 3 hours, the solvent was evaporated and Compound 3 was purified by flash chromatography using 10% methanol in CH2C12, lOd. About 0.05 eguivalents of 4-dimethylamino pyridine, 1.4 equivalents of triethylamine and 1.2 equivalents of 4,4'-dimethoxytrityl chloride were added to a stirred solution of Compound 3 (o.6 mmol) in dry pyridine (4.o 25 ml). After 2 hours, t~e reaction mixture was quenched with 2.0 ml of water and then diluted with 2.0 ml of et~yl acetate. The organic phase was separated, washed with brine and dried. Compound 4 was purified by flash chromatography using 5% methanol in methylene chloride.
lOe. About 2.0 equivalents of diisopropyl ethyl amine and 1.0 ml of dry dichlorometh~ne (CH2Cl2) was added to a stirred solution o~ Compound 4 (0.5 mmo~). After 30 minutes, 0.75 equivalents of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite was 35 added, drop by drop over a period o~ 20 minute~ and . . ,. .. . - .. :
.
, ,.. , ... , , :
. . , , ,, , . , - ,.
:

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

WO~Z/025~ PCT/US91/OS531 stirring continued for another 1 hour. The ~olvent was then evaporated and Compound 5 w~s purified by flash chromatography using ethyl acetate ~containing lS
triethyl amine) under nitrogen atmosphere.
S The processes of steps a-d above re used to make dimers containing three carbon internucleoside linkages in which all three carbons have the formula -CH~-.
Any or all o~ the carbons can be optionally hydroxylated by modifying the process illustrated in Figure 3 as follcws. A drop of 2.5S (w/v) solution of osmium tetraoxide in t-butanol at O-C was added to a ' ~`~'~'' stirred solution of Compound 1 and 4-methyl morpholine N-oxide (~.1 mg) in 0.8 ml of THF. The rsaction mixture was kept at O'C for 24 hours, ~uenchled with an aqueous solution o~ sodium metabisulfite, diluted with ethyl acetate and washed with water and brine.' The solvent was e~aporated and the resulting hydroxylated dimer purified by thin layer chromatography using ethyl acetate a~ an eluent. The hydroxylated dimer is then pxotected and the 5' and ~' ter~inals modi~ied as in steps b-d, above.

EXAMP1E 11: ~re~aration of h~droxYlated three,carbon internu~leoside ~in~aqes.
The thymidine-dimer phosphoramidite compounds produced by steps a-d were used in a modified solld phase phosphoramidite synthetic procedure to make the oligonucleoside sequence~ of Table 1.
The oligodeoxynucleotides were synthesized from the 3' to the 5' terminus.

: . .. :
..: ,: ., .

- ~ . :

W092/02534 PCT/US91/OS~31 4~_ T~le 1 Sequenc ~ç~. Code S' TpTpTpTpTp[TcT]pTpTpTpTpypypT 3' S' TpTpTpTpTpTpTp[TcT]pTpTpypypT 3' 2 5' TpTpTpTpTpTpTpTp[TcT]pypT 3' 3 T= thymidine 1 0 p-- O--P--O
le c= --CH2-C~I~-CH2-Y= tetraethyleneglycol Synthesis then proceeded in accordance with a modifi~d phosphoramidite procedure. The 5'-hydroxyl group of the attached thymidine was reacted with trichloroacetic acid to deprotect: the 5'-hydroxyl group.
Following this deprotection step, the attached thymidine was reacted with the activating a\gent, tetrazole, and a phosphoramidite reagent comprising dimethoxytrityltetraethyleneglycc\lcyanophosphine. The acti~ation step was followed by the capping of unreacted 5'-hydroxyl groups with acetic anhydride and N-methylimidazole. The phosphorous: linkage was then oxidized with iodine in accordance with standard procedures. In sequences l and 2, containing two tetraethyleneglycol (TEG) residues, the deprotecting, activating, capping and oxidizing steps were repeated as described above.
Chain elongation then proceeded via the standard saguential steps o~ deprotection, acti~ation, 3~ c~pping and oxidation with t~e modification t~at a three-carbon linked thymidine dimer, prepared according to the methods of Examples l-9, in the chain was added where desired during an activation step.

: . ~

. : ::: : : :
., . . :~ .
: :::- : - ::: ::- -:
':'~ ":

W~2/02534 PCT/US91/05~31 , .. .
~' J~ - 45 - ~

At the end o~ chain assembly, the thymldine oligomers were removed from the CPG support with concentrated ammonium hydroxide. The solution was then further treated at 55-C for 8 to 15 ~ours to remove all the protecting groups on the exocyclic amines of the bases.

ExAMPm~ 12: Pre~aration o~ 3'-0-acetyl-5'-carbomethoxymethvl-5'-deoxyth~idine About 0.3~ g sodium borohydride was added to a cold (ice bath) stirred, mixture of 3.17 g of 3'-0-acetyl-5'-carbomethoxymethylene-5'-deoxythymidine in 95 ml isopropanol. The mixture was stirred at O~C under nitrogen atmosphere for half an hour, then at room tempQraturQ fo~ an additional ~o~r and one half hours.
The chllled mixture was quenched with 20 ml methanol, followed in 30 minutes ~with 200 ml of distilled water, and then extracted with several portions of ethyl acetate. The combined orqanic extracts were treated with br~ne and dried with anhydrou~ magnesium sulfate. The drying agent was filtered off and the solvent evaporated, yielding the title compound as a residual cllass of 2.7 g.

EXAMPLE 13: Pre~aration of 5'-carbome~hoxyme~hyl~
5'-deoxvth~midine About 10 drops of a 25S (w/v) methanolic solution of sodium methoxide was added to a cold, s~irred solution of 2.~2 g o~ 3'-0-acetyl-6'-carbomethoxymethyl-5~-deoxythymidine, prepared according to the method of Example 12, in about 300 ml of dry (passed throuc3h a bed of neutral alumina) methanol.
The mixtur~ was stirred un~er a nitrogen atmosphere, without replenishing the ice bath, for about 20 hours.
A small amount of cation exchange resin (Bio-Rad AG SOWX8~, was aclded and the ~ixture stirred for 30 .. . ....
. :- .:. . .
:: . . . . .. . . -- ~ .,.

W~2/~253~ PCT/US91/0~31 .~ ~ r, r~ ~ j minutesO The solvent was removed under reduced pressure, yielding a residual glass of 2.1 g, which was treated with warm toluene, and, a~ter cooling, filtered and rinsed out with cyclohexane to yield a crude product as a white solid, 1.64 g.
The title compound was further purified from a trace of starting material by chromatography on silica ~el, eluting with ethyl acetate, then recrystallization from ethyl acetate/hexane to yield white crystals.
EXAMPLE 14: PreParation of 3'-0-t-butvldimethvlsilvl-5'-~2"-hYdroxyethvl!-thvmidine - ~
A~out 19 ml of a lM diisobutylaluminum hydride in tetrahydrofuran (THF) were added to a cold (-40 to -30-C), stirred solution of 1.8~ g of 3'-0-t-butyldimethylsilyl-5~-carboethoxymethyl-5'-deoxythymidine in 40 ml of anhydlrous ~HF, under a nitrogen atmosphere. The reaction temperature was then slowly increased to -20-C.
~he mixture was quench~ed with about 3.5 ml methanol, and the reactlon tempelrature increased to -lO-C. About 18 ml water in 36 ml THF were added ~o the warm mixture and the temperature further increased to 10 C. Most of the T~F was rPmoved, via reduced pressure and the residue diluted with about two volumas of water.
The aqueous phase was extracted several times with ethyl acetate/chloroform. ~he combined extracts were washed with cold 2N hydrochloric acid, and brine, dried with anydrous magnesium sulfate and ~iltered. The sol~ent was removed from ~he filtrate with reduced pressure to yield the title compound (about 1.6 g~.

- . :

:~

W0~2/02534 PCT/US91/05531 EXAMPLE 15: Pre~aration of 3'-O-t-butyldimethylsilvl -5'-r2"-iodoethvl)-5'-deoxYthymidine About 1 g of p-toluenesulfonyl chloride was added to a solution of 1 g of 3'-0-t-butyldimethylsilyl-5'-(2ll-hydroxyethyl)-5'-deoxythymidine, prepared according to the method of Example 14, in 25 to 30 ml of anhydrous pyridine, and the mixture maintained, stoppered, at about 5'C for approximately 19 hours.
The mixture was added to about 200 ml of ice water and extracted several times with ether. The combined organic extracts were washed with cold 2N
- hydrochloric acid, water, and brine. The washed extracts were dried with anhydrous sodium sulfate and filtered. The solvent was removed ~rom the filtrate via reduced pressure to yield a residual glass of 1.24 g of the p-toluenesulfonyl derivative.
About 0.54 g of the p-toluenesulfonyl derivative and 0.38 g sodium iodide were dissolved in 55 ml of dry (molecular sieves, 4A) acetone for three days, followed by the further addition of 0.19 g sodium iodide with stirring for a ~inal day, The reaction mixture w~as filtered and the solvent was evaporated to yield crude product.
The crude product was purified by chromatography on 85 g of silica yel eluting with 25%
ethyl acetate in hexane. The solvent was evaporated to yield 0.4 g of ~he desired 3'-0-t-butyldimethylsilyl-5'(2"-iodoethyl)-5~-deoxythymidine.

EXAMPLE 16: Preparation of 5'-carbomethoxvmethvlene-5~-deoxythY~idine About 10 drops of a 25% sodium methoxide in methanol wer~ added to a stirred solution of 1.5 g of 3'-O-acetyl-5'-carbomethoxymethylene-5'-deoxythymidine in 150 ml of dry methanol (passed through a bed of ; , , : , , ,;
: ` ~ ` ' : : : : , :

: " , WO 92/02534 P(~/US91/05~;31 n n f~ ~/ V

neutral alumina). The mixture was stirred at room temperature under a nitrogen atmosphere for an additional 6 hours.
A small amount of cation exchange resin (Bio-Rad AG-50W-X8) was added to the mixture with stirring for 10 minutes. The solvent was removed under reduced pressure to yield a white solid residue of 1.3 g. The residue was triturated twice with warm toluene, then taken up in hot ethanol, filtered, and chilled to yield the title compound as a white crystalline product, after drying, 0.85 g.

EXAMPLE 17: Pre~aration of 3'-O-t-butYldimeth~lsilYl-5'-(2"-hydroxyethylene~-5'-deoxythymidine A solution of 296 m~ 5~-carbethoxy~ethylene-5'-deoxythymidine was added dropwise under a nitrogen atmosphere to a cold ~ice water bath), stirred solution of 205 mg imidazole and 227 mg t~-butyldimethylsilyl chloride in 1 ml of anhydrous dimethylformamide. After complete addition, the mi~ture W~IS removed ~rom the ice and stirring continued at ambienl: temperature for two hours, then at 35-C for another l:wo hours, and finally at 40-C for half an hour.
The mixture was then quenched with 2 ml of methanol, followed by two to three volumes of water.
The aqueous phase was extracted several times with ethyl acetate. The combined organic extracts were washed with water, saturated bicarbonate solution, and brina, dried with anhydrous magnesium sulfate, and filtered. The solvent was removed from the filtrate via reduced pressure to yield 0.40 g of 3'-O-t-butyldimethylsilylyl-5'-carbomethoxymethylen2-5'-deQxythymidine.
About 4 ml of a lM solution of diisobutylaluminum hydride in tetrahydrofuran was added dropwise to a -30-C to -35~C chilled solution of 0.37 g .
, : .
, . .... : :: :
: : ~

wos2/o2s~ P~T/US91/05531 ,,~ c~
~_ ~ 49 -o~ the 3~-o-t-butyldimethyl5ilyl-5~-carbomethoxymethylene-5'-deoxythymidine dissolved in 10 ml of anhydrous te~rahydrofuran at a temperature below -30-C. After addition was complete, the reaction was stirred under a nitrogen atmosphere for an additional two hours while maintaining the internal temperature in the range of from about -3Q- to about -20-C.
About 0.8 ml o~ methanol was added to the reaction mixture followed by the addition of a solution of 4 ml water in 8 ml of tetrahydrofuran. Most of the more volatile tetrahydro~uran was removed under reduced - --pressure. The aqueous residue was dil~ted-with about twice its volume of water and extracted several tim~s with ethyl acetate. The combined organic extracts were washed with cold lN hydrochloric: acid, and brine, dried with anhydrous magnesium sulfat~! and fil~ered. The filtrate was stripped to yield t:he residual tltle compound, 0.267 g.
A portion of this material was purified to analytical purity by chromatography on silica gel, eluting with 50S ethyl acetate/llexane.

EX~MPLE 18: PreParation of ~hYmidine dimers containinq 2 two carbon - one oxv~en atom ~ 0-C-C-5~) internucleoside Linkaqe 18a. To a stirr~d solution of 5'-0-tritylthymidine is added an equimolar amount of each of a base and 3'-0-t-butyldimethylsilyl-5'-(2n-iodoethyl)-5'-deoxythymidine at 0C. The course of the r~action is monitored'by thin layer chromatography (TL~). After the completion o~ the reaction, the desired dimer is isolate~ and purified ~y ~lash chromatography.
18b. ~o a stirred solution of 3'~0-t-butyldimethylsilyl-5'-(2"-hydroxyethyl)-51-3S deoxythymidine maintained at a te~perature of about -5-C

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

. .. .
: :. . :
.

WO ~ 02534 PCl`/US91/05531 r~ t ~; v is added 2 equivalents of base, and the solvent is evaporated to dryness. The residus is redissolved in DMF, and an equivalent of 5'-dimethoxytrityl-2',3'-cyclothymidine is added. The reaction mixture i~ heated to about 40-C, and the formation of the d~sired dimer is monitored by TLC. After the completion o~ the reaction, the desired dimer is isolated and purified by flash chromatography.

EXAMPLE 19: De~rotection o~ the 3' end of the dimer of Exam~le 18 The 3'-t-butyldimethylsilyl protecting group of the protected dimers is removed by treatment of a THF
solution of the dimer of Example 18 with 2.8 ~equivalents o~ tetrabutylammonium fluoride at O-C. After the completion o~ the reaction (generally about 3 hours), the solvent is evaporated and the desired dimer is isolated and purified by flash chromatography.

EXAMPLE 20: preparation of a functionallzed dimer unit s~l~able for,~utomated sYnthesis The dimer product o~ Example 19 i5 dissolved in dichloromethane, and 2 equiva].ents of diisopropylethyl amine are added. The mixture is stirred for 30 minutes, followe~ by dropwise addition of 0.75 equivalents of 2-cyanoethyl N,N-diisopropylchlorophosphoramidite over a period of about 20 minutes. The stirring is continued ~or another hour, the solvent is evaporated, and the resulting ~unctionalized dimer is isolated and purified on a flash chromatography column using ethyl acetate under an lnert atmosphere.

W092/02534 PCT/US91/05~31 EXAMPLE 21: Preparation of S~-t-~utyldim~thylsilvl-3'-deoxy-3'-rl".2"-dihydroxv-3"-pro~vl)-thymidine.
O~mium tetraoxide (0504) (4 drops, 2.5~ w/v) in ~utanol was added to a stirred mixture of 3'-(2"-propenyl)-3'-deoxy-5'-O-t-butyldimethylsilyl thymidine (183 mg, 0.5 mmol), prepared according to the literature procedure, and 4-methylmorpholine-N-oxide (53 mg, 0.45 mmol) in 5.0 ml dry THF at 0C. The reaction mixture was then quenched with 10% aqueous sodium metabisulfite (2.0 ml), stirred for 20 minutes, filtered over a pad of silica and-~diluted with ethyl acetate (25.0 ml). The organic phase was washed with water (5.0 ml) and brine, and then dried with Na2SO4. The solvent was evaporated and the title compound purified by flash chromatography.

EXAMPLE 22: Preparation o~ 3'~deoxv-thymicl-3-~1-ace~aldehYde-5'-0~-t-butvldimethylsilylthymidine~
Sodium periodate ~214 Ing ~ 1 mmol) wa~ added to a stirred solution o~ the thymidLn~ diol prepared by the method o~ Example 21 ~200 mg, O.!i mmol) in THF-H20 ~4:1 ratio, 5.0 ml). After 1 hour, the reaction mixture was diluted with ethyl ac~tate ~25.0 ml), washed with H20 t2 x 5.0 ml) and brine and dried. The title compound was purified by flash chromatography with 70% ethyl acetate in hexane.

EXAMPLE 23: PreParation of 5'-o (p-toluenesulfonyl) th~idine~
To a stirred solution of thymidine ~20 g, 82.6 mmol) in dry pyridine (200 ml) was added p-toluenesulfonyl chloride (47.2 g, ~47.6 mmol) at O-C
under nitrogen atmosphere. A~ter 3 hours, the reaction mixture was poured onto ice and extracted with et~yl - , : : . . . : ..
.,, ;, ~ . . ., ~;
.: : . , :, : , :
- :~ . . , :
:.................. :. : : . - .:

W092/02s34 PCT/US91/0~531 ~ ~ ~,,~,3 V s -~

acetate. The solvent was evaporated and the product was crystallized from a mixture of ethyl acetate ~nd methanol. The title ~ompound was a whita crystalline solid obtained in 70-75% yield.

EXAMPLE 24: Pre~aration of 5'-iodo-5'-deoxyt~Ymidine.
To a stirred solution of the thymidine ~osylate (10.55 g, 26.9 mmol) prepared according to the method o~ Example 23 in dry acetone (75 ml) was added sodium iodide (10 g, 66.7 mmol) and the mixture refluxed for 16 hours. The solvent was evaporated and diluted with ethyl acetate. The organic phase was washed with water (2 x ~0 ml) and brine (~0 ml) and dried with sodium sulfate. The title compound was crystallized from methanol as a white crystalline solid in 90-95%
yi~ld.

EXAMPT~ 25: PreParation of 3'~0-t-but~ldimç~hY
5'-~odo-5'-deoxvt;hv~idine.
To a stirred solution of the thymidina iodide ~8~0 g, 24.7 mmol) prepared according to the method of Example 24 in dry DMF, wa5 added imidazole (4.2 g, 61.7 mmol). After 5 minutes tert~butyldimethyl silyl chloride (4.47 g, 29.64 mmol) was added and the mixture stirred for 4 hours. The reacti~n mixture was then diluted with ethyl acetate (250 ml), washed with water ~2 x 100 ml) and brine (50 ml) and then dried with sodium sulfate. The title compound was purified ~y flash c~romatography using 60% ethyl acetate in hexane in 92% yield.

EXA~P~ 26: Pre~aration of 3'-O-t-butvldimet~Ylsi 5'-azido-5'-deoxythYmidine.
To a stirred solution of the 3'-0-t-butyldimethylsilyl-5'-iodo-5'-deoxythymidine ~10.8 g, 20 ... . .
, ' :.' ', .
.
- .
:. : - -: ..

Wo~2/n2534 PCT/US91/0~531 r (~
, ~

mmol) prepared according to the meth~d of Example 25 in dry DMF (50 ml) was added sodium azide (3.9 g, SO mmol) and the mixture heated at O-C for 12 hours. Then the reaction mixture was diluted with ethyl acetate (200 ml) washed with water (2 x SO ml) and brine (SO ml) and then dried with sodium sulfate. The title compound was purified by flash chromatography using 50% ethyl acetate in hexance in 90~ yield.
., 0 EXAMPT.F 27: PreParation of 3'-G-t-butyldimethylsil~l-5'-amino-S~-deoxythvmidine.
To a stirred solution of the thymidine azide (5.0 g, 13.1 mmol) prepared according to the method of Example 26, in MeOH was added 200 mg of 10~ Pd-C und~r ni~rogen atmosphere. The nitrogen gas was then evacuated and replaced by hydrogen. The evacuation and replacement procedure was repeated twice and s~irring was continued under 1 atmospheric pressure of hydrogen ~or 12 hours. The hydrogen was removed and the catalyst ~0 was filtQred over a pad of celit~ and the solvent was removed und~r vacuum. ~he crude produc~ was purifiad by flash chromatography using 5-10% MeOH in CH2Cl2 to give the title compound in 85-87% yield.

EXAMPLE 28: Pre~aration of 3'-azido-3'-deoxY-5'-0-dimethoxvltritvl thvmidine.
To a stirred solution of 3'-azido-3'-deoxythymidine (2.67 g, 10 mmol) in dry pyridine (50 ~1) was added 4-dimethylaminopyridine (61 mg, 0.5 mmol), triethylamine (1.9 ml, 14 mmol) and 4,4'-dimethoxytrityl chloride (4.1 g, 12 mmol) in a sequential order. After 3 hours, water (30 ml~ was added and extracted with et~yl acetate (250 ml). The organic phase was separated, washed wit~ brine ~SOml) and dried with sodium sulfate. The title compound was purified by ;

w092/025~ PCT/US91/05~31 ~lash chromatography using 5S methanol in methylene chloride in 80-85% yield.

EXAMPLE 29: Preparation of 3'-amino-3'-deoxy-5'-o-dimethoxytrityl thvmidine.
To a stirred solut~on of the thymidine azide (3.99 g, 40 mmol), prepared according to the method o~
Example 28, in MeOH was added 200 mg of 10% Pd-C under argon atmosphere. The argon gas was remov~d by vacuum and hydrogen was introduced. This procedure was repeated twice and stirring was continued under 1 atmospher,e of hydrogen pressure for 12 hours. Then hydrogen was removed and the catalyst was filtered over a pad of celite and the solvent was removed under vacuum. The crude product was purified by flash chromatography using 5% MeO~ in methylene chloride to give the title compound in 90-93~ yield.
~H NMR t300 MHz, CDCl3) ~ 7.61 (s, 1 H), 7.60-7.21 (m, 1 H) 6.83-6.87 (m, 3 H), 6.85 (t, J~8.5 Hz, 1 H), 3.80 ~5, 6 H), 3.81-3.73(m, 2 H), 3.53-3.49(~, 1 H), 3.38-3.33(m, 1 H), 2.36-2.33(m, 1 H), ` 2.25-2.20 tm, 1 H), l.51(s, 3 H): IR neat YmaX 3020, 2962, 1697, 1605, 1512, 1246, 1030 cm~.

EXA~IPLE 30: Pre~aration of 5-O'-dimethoxytritvl-3-OI-t-butyldimet~Ylsilyl thvmidine.
Dimethoxytrityl thymidine (5.0 g, 9.2 ~mol) and imidazole (1.2 g, 18.4 m~ol) were dissolved in 15 ml of anhydrous dimethyl formamide (DMF) and added to tert-butyldimethylsilyl chloride (1.7 g, 11.5 m~ol). The reaction ~ixture was stirred for 4 hours at room temperature, diluted with ethyl acetate and washed with water, saturated sodium chloride and dried with sodium ~ulfate. A guantitative yield of the title compound was obtained.

t ~ .
. . . : .
:, , . .:, . ,, . . ;

W0')2/02534 P~/US91/0553t ,~ ,~ . ...
' - 55 -EXAMPTE 31: PreDaration o~ 3'-O-t-butyldimethyLsi ~Y
thymidine.
5'-O-dimethoxytrityl-3'-O-t-butyldimethylsilyl thymidine prepared according to the method of Example 30 (0.7 g, 1.1 mmol) was treated for l hour at roo~
temperature with 13 ml of a 3% solution of trichloracetic acid in methylene chloride. The reaction mixture was then neutral~zed with a 5% (w/v) sodium bicarbonate solution. The organic layer was dried with sodium sulEate. The title compound was purified ~y flash chromatography using a 0 to 30% gradient of ethyl acetate in methylene chloride. The yie}d of ~he reaction was 8S%.

EXAM~LE 32: PreDaration of 3'-O-t-butvldim~th~lsilYl-5'-~arbethox~mçthvlene-5'-deoxvthymidin~.
To a well stirred solution of dry m2thylene-chloride at -78'C was added oxalyl chloride ~33.0 mmol, 2.88 ml) ~ollowed by the dropwise addition of DMS0~3.12 ml, 44 mmol). After 10 minutes, the thymidlne alcohol ~5.6 g, 15.7 mmol), prepared according to the method of Example 31, in 20.0 ml of CH2~12 was added dropwise over a period of 2 minutes and stirring continued for 45 minutes. Et3N (8.1 ml, 58.1 ~mol) was added and stirring continued for another 30 minutes. The reaction mixture was then brouqht to -23-C over a period of 30 minutes. Then carbethoxy methylene triphenylphosphorane ~10.94 g, 31.4 mmol) was added and the reaction mixture stirred for 12 hours at room temperature. The reaction mixture was then diluted with water (2 x 125 ml) and brine (50 ml? and dried (Na2SO~. The crude product was purified by flash chromatography using 20% ethyl acetate - hexane ~ 40% ethyl acetate- hexane to give both the : . ....... . :.- r : ~ : . : ,:,- -- . . : ., : . . . :. .

W0~2/02534 PCT/US91/05~31 r~ r, r~
J ~ ~ rj ~ S6 ~

trans and ~is isomers of the title compound in 3:}
ratio. The combin~d yield was about 72-76%.
Data for trans compound. IR (neat) v~ax 3205, 3180~ 2982~ 2964~ 1698~ 1490~ 1274 cm1; 1H NMR(300 MHZ~
S CDCl3) ~ 7~04 (5~ 1 H), 6.87 (dd. J=15.6 and 5.4 Hz, 1 H), 6.23 (t, J= 6.7 Hz, 1 H), 6.03 (dd, J= 15.6 and 1.6 ~Z~ 1 H), 4.33-4.28 (m, 1 H), 4.14 (q~ J= 71 Hz 2 ~) 4.16-4.12 (m, 1 H) 2.28-2.19 (m, 1 H), 2.09-1.98 (m, 1 H~ 1~87 (S~ 3 H), 1.23 10 (t, J37.1 ~z, 3 H), 0.81 (S~ 9 H) o.o1 (s, 6 H): Calcd for C20H32o6N2si; c, 56.58; H, 7.60; N, 6.60; FoundO C~
56.36; H, 7~30;- N,- 6~60~ - - -EXAMpTT~` 33 Preparation of 3'-0-t-butvldimethYlsilYl-5 ~ -ça~ethQxvmethyl-51 -deoX~th~midine .
To a stirred solution of the unsaturated thymidine ester (4 ~ 24 g, 10 ~mol), prepared according to the method of Example 32, in EtOAc was added 200 mg of 10~ Pd-C under nitrogen atmosphere. The nitrogen gas was re~oved by vacuum and ~ydrogen was introduced. This procedure was r~peated twic~ and stirring was continued under 1 atmospheric pr~ssure o~ hydro~en ~or 16 hours.
Then the catalyst was filtered over a pad of cel~te and the solvent was removed under vacuum. The product was crystallized fro~ a mixture of hexane and et~yl acetate.
The ~itla compound was obtained in 95% yield.
IR(neat) vmax 3180, 2925, 2950, 1696, 1486, 1260, 1240 cml; lH NMR (300 MHZ, CDCl3) ~ 7.20 (Bt 1 H), 6.11 (t, 6.6=Hz, 1 H) 4~07 (q~ J=7.1 HZ, 2 H), 4.03 -3.98 ~m, 1 H), 3.73-7.69 (m, 1 H), 2.51-2~32 (~, 2 H), 2.24-2.15 (m, 1 H), 1.18 It, J=7.1 Hz, 3 H), 0.81 (s, 9 H), 0.01 (s, 6 H), Anal. Calcd for C20H~06N2Si: C, 56.31; H, 8.03; N, 6.57 Found: C, 55.91; H, 7.74; N, 6.50.
.

- . . : ~.
:.~ : . .

W092/025~ PCT/US91/05531 EXAMPLE 34: Pre~aration_of 3~-o-t-butyldimethvlsi 5'-deoxv-thv~id-5-Yl-acetaldehYde.
To a stirred solution of the thymidine ester (3.41 g, 8 mmol), prepared according to the method of Exampl~ 33, in 60 ml of dry CH2Cl2 at -78-C was added DiBAL-H tl6.4 ml, 1.0 M solution in h2xane, 16.4 mmol) dropwise over a period of 3 min. After 20 minutes, thP
reaction mixture was diluted with 300 ml of EtOAc and washed with 50 ml of saturated sodium potassium tartrate solution twice. The organic phase was washed with brine ~25ml) and dried ~Na25O4). The title compound was - - purified by flash chromatography using 50~-70% ethyl~
acetate - hexane in 85-87% yield.

EXAMPLE 35: Preparation of a deoxythvmidine dimer h~vinq a 3'-C-C-N-5' internucleoside llnkaae ~Fiqu~e 3) 35a. To a stirred ~olution of the thymidine amine ~rom Example 27 ~1.07 g, 3 ~mol) and the thymidine aldehyde o~ Example 22 (1.38 g, 3.6 ~mol) ~n 50 ml o~
eth~nol and 10 ml of aqueous bu~'er solution ~pH ~ 5.5, NaH,PO~-NaOH) was addQd a solution of NaCNBH3 in THF ~12 mL, l.O M solution in THF, 12 mmol) dropwisa at 5-C over a period of 1 hour. The reaction mixture was stirred for another 4 hours and diluted with 2.50 ml of ethyl acetate. The reaction mixture was was~ed with water ~2 x 40 ml) and brine (25 ml) and dried tNa2SO4). Compound 1 tFigure 6) was purified by flash chromatography by first eluting with ethyl acetata followed by 5-8~ MeOH-C~2Cl2 in 62-64~ yi~ld.
IH NMR ~300 MHz, ~DCl3) ~ 7.60 ~s, 1 H), 7.19 (s, 1 H), 6.18 (t, J-6.6 Hz, 1 H), 6.08 ~t, J=3.9 Hz, 1 H), 4.29-4.23 (m, 1 H), 4.15-3.98 ~m, 1 H~, 3.91-1.85 (m, 1 H), 3.70-3.78 (m, 2 H), 2.95-2.87 (m, 1 H), 2~84~2r66 (m, 3 H), 2.35-2.05 (m, 5 H), :

` - ~.: . -: : ......................... : ; -:. :: . .. ~ -: :i . ~ . , WO~/02534 PCTfUS9ltO~5~1 1.94 (s, 3 H), 1.93 (s, 3 H), 1.80-1.63 (m, 1 H ), 1.55-1.45 (m, 1 H), 0.93 (s, 9 H), 0.69 (s, 9 H), 0.11 (s, 6 H), 0.07 (s, 6 H).
35b. Compound 1 (Figure 6) (166 mg, 0.23 mmol) was added to a stirred ~olution of trifluoroacetic anhydride ~0.32 ml, 2.3 mmol) and triethyla~ine (0.64 ml, 4.6 mmol) in CH2C12 (5.O ml). After 2 hours, the reaction mixture was quenched with aqueous NaHC03 ~5.0 ml) and diluted with EtOAc (25ml). The organic phase was washed with water (2 x 10 ml), brine (5 ml) and dried (Na2SO4). Compound 2 (Figure 6) was purified by flash chromatography using 7% ~eOH-in CH2Cl2 în 91-93%
yield.
35c. To a stirred solution of Compound 2 (164 mg, 0.2 ~nol) in ~F (4.0 ml) was added tetrabutyl-ammonium fluoride (0.8 mmol) at D'C. After ~ hours, the solvent was evaporated and Compound 3 (Fiqure 6) was purified by flash chromatography using 5%-8% MeOH in CH2Cl2 in 90% yield.
35d. To a stirred ~olution o~ Compound 3 (151 mg, 0.26 mmol) in dry pyridine ~ 3.0 ml) was added 4,4-dimethylaminopyridine ~1.6 mg, 0.0128 mmol) and triethylamine (0.057 ml, 0.42 mmol). After 5 minutes, dimethoxytritylchloride tl21 mg, 0.3~8 mmol) was added ~5 and stirring continued. A~ter 2 hours, the reaction mixture was diluted with ethyl acetate (25 ml) and washed with water (2 x 10 ml), brine (5 ml) and dried (Na2SO~). The crude product was purified by ~lash chromatography using 7% MeOH in C~2Cl2 to give Compound 4 (Figure 3) in 85-87% yield.
35e. Dry diisopropyl ethylamine (0.15 ml, ;
0.67 mmol~ was added to Compound 4 (150 mg, 0.168 mmol) followed by dry CH2Cl~ ~0.5 ml). Then the flask was shaken to dissolve the alcohol and 2-cyanoethyl-N,N-diisopropylchlorophosphoramidite ~0.056 mL, 0.25 .;

. ,. . ~, . .

W092/02534 PCT/US91/05~31 'J j~

mmol) was added over a perioA of 20 seconds. After 45 minutes, t~e reaction mixture was quenched with C~30H
tl.0 ml), diluted with EtOAc (50 ml) and Et3N (1.0 ml), washed with 10S aqueous X2CO3 t2 x 5.0 ml), followed by brine (5.0 ml) and dried (Na2SO~). The crude product was purified by flash chromatography using EtOAc to give Compound 5 (Figure 6) in 70-75% yield.

EXAMPLE 36: PreDaration of a deoxythymidine dimer havinq a 3'-N-C-C-5'-internucleoside linka~e !Fi~ure 4).
36a. To a stirred solution of the amine of --~
Example 29 (2.72 g, 5 mmol) and the aldehyde of Example 34 (2.29 g, 6 mmol) in 50 ml of ethanol and 10 ml of 15 aqueous buffer solution (pH - 5.5, NaH2PO~NaOH) was added a solution o~ NaCNBH3 in THF (12ml, 1.0 ~ solution in THF, 12 mmol) dropwise at 5'C over ~ period o~ 1 hour. The reaction mixture was stirred for another 4 hours and then diluted with 250 ml of ethyl acetate.
20 The reaction mixture was washed wit~ water (2 x 60 ml) and brine and driQd ~Na2SO~). Compound 1 (Figure 7) was purifiQd by fl~sh chromatography by first eluting with ethyl acetate followed by 5% MeOH-CH2Cl2. Compound 1 (Figure 7) was obtained in 72-74S yield.
IH NMR (300 MHz, CDCl3) ~ 7.56 (m, 1 H), 7.36-7.34 (m, 2 H), 7.29-7.15 (m, 8 H), 7.03 (s, 1 H), 6.77 (m, 3 H), 6.20 (t, J=6.0 Hz, 1 H), 6.08 (t, J36.7 Hz, 1 H) 4.01-3.97 (m, 2 H), 3.84-3.72 (m, 1 H), 3.72 (s, 6 H), 3.71-3.63 (m, 1 H), 3.48-3.32 30 (m, 2 X), 3.30-3.22 (m, 1 H), 3048-3.32 (m, 2 H), 3.30-3.22 (m, 1 H), 7.52 (m, 2 H), 2.27-2.1~ (m, 3 H), 2.08-1.97 (m, 1 H), 1.83 (s, 3 H), 1.67-1.48 (m, 3 H), 1.43 ts, 3 H), 1-22-1.15 (m, 1 H), 0.82 ~s, 9 H), 0.01 (s, ~

W~9~/02534 PCT/US91/06~31 f~ U ~,~ 'J ;) i i 36b. To a stirred solution of trifluoroacetic anhydride (O.32 ml, 2.3 mmol) and triethylamine (0.64 ml, 4.6 mmol) in CH2Cl2 (5.0 ml) was added Compound 1 (Figure 7) (210 mg, 0.23 mmol). After 2 hours, the reaction was quenched with aqueous NaHCO3 (5.0 ml) and diluted with EtOAc (25 ml). The organic phase was washed with water (2 x 10 ml), brine (5 ml) and dried (Na,SO~). Compound 2 (Figure 7) was purified by flash chromatography using 7S MeOH in CH2C12. Compound 2 was obtained in 89-91% yield.
36c. To a stirred solution of Compound 2 (180 mg, 0.2 mmol) in THF (4.0 ml) was~added tetrabutyla~monium fluoride (0.4 ml, 1.0 M solution in the T~F, 0.4 mmol) at O'C. After 2 hours, the solvent was evaporated and the product was purified by flash chromatography using increasing polarity, 5-8~ MeOH, in CH2Cl2 to give Compound 3 (Figure 7) in 89~ yield.
36d. Dry diisopropyl ethyl amine (0.15 ml, O.67 mmol) was added to Compound 3 (150 mg, 0.168 mmol) followed by thQ addition o~ dry C:H2C11 ~0.5 ml). Then the flask was shaken to dissolva the alcohol and 2--cyanoethyl-N,N-diisopropylchlorophosphora~idite (0.056 ml, 0.25 ~mol) was added over a pariod of 20 seconds.
After 45 minutes the reaotion ~ixture was quenched with CH30H (0.1 ml) and diluted with EtOAc (50 ml) and Et~N
(1.0 ml) and washed with 10~ aqueous K2CO3 (2 x 5.0 ml), and brine (5.0 ml) and dried (Na2SO4). The product was purified by flash chromatography using EtOAc to give Compound 4 in 70-75% yield.
EXAMPLE 37: Svnthesis of deoxYthymidine oliq~ers ~ontaininq a 3'-~-C-C-5' inte_nu~leoside linkaqe.
The thymidine-dimer phosphoramidite compounds produced by steps a-d above were used in a modi~ied . ~ , . . .

.. . . . ..
.

WO~2/0253'l PCT/US91/05~31 solid phase phosphoramidite synthetic procedure to make the oligonucleoside sequences of Table 2.

Table 2 seouence Re. Code 5' TpTpTpTpTpTpTpTp[TnT~pT 3' 4 5' TpTpTpTpTp[TnT]pTpTpTpT 3' 5 5' [TnT]pTpTpTpTpTpTpTpTpT 3' 6 T- thymidine p= --o--P--O--Oe n- 3'-N-C-C-5' The oligodeoxynucleoside ~equenc6~s were synthesized ~rom the 3' to the 5' terminus.
The initial step was the attachment, ~ia a 3'-succina~e linkage, o~ a 5'-dimet~loxytrityl deoxythymidine to a CPG support. The 5'-O-dimQthoxytrityl group o~ the attached thymidine was r~acted with trichloroacetic aci~l ~o deprotect thQ 5'-hydroxyl group.
Chain elongation then proceeded ~ia the standard sequential steps o~ deprotect$on, activation, capping and oxidation with the modification that an -N-C-C- linXed thymidine dimer, prepared a~cording to the methods of Examples 30-37, was added in the chain where desired during an activation step.
At the end of chain assembly, the thymldine oligomers were removed from the CPG support wlth concentrated ammonium hydroxide. The solution was then further treated at 55-C for 8 to 15 hours to remove all the protecting groups on the exocyclic amines o$ the bases.

~ , .
!' , ; ~
:': ' ~ '` " `' ~ ' , . . ' ~ . . '`~ . ''' ' `
. , ' ` ~ .

' ` '; ~ ' W0~2/0253~ PCTtUS91/0~31 ~ r~ 3 ~ rj ~ ;3 EXAMPLE 38: Pre~aration of Tetraethvlene~lycol-terminated ~n~i-RAS onco~ene ~
38a. Preparation of dimethoxytrityltetra-ethyleneglycol (DMTTEG) An excess of tetraethyleneglycol TEG (about 100 ml) was admixed with about 7 ml (5.lg; 40 mmols) of Hunig's base in a round bottom flask. About 3.08g (lO
mmols) of dimethoxytrityl chloride (DMTCl) was added to the TEG admixture and the DMTCl-TEG mixture maintained with constant stirring at room temperature (about 2S~C) for about 8 to 12 hours to ~orm DMTTEG.
~ 38b. Preparation of ~~~~ ~~ ~~~
dimethoxytrityltetraethylene-glycolcyanophosphine (DMTTEGCP).
Six grams of the DMTTEG from step ~a) was admixed with 20 ml of dry dichloromethane. About 6.2 ml of Hunig's base was added to the admixture, followed by the dropwise addition of a chlor~phosphine mixture to form DMTTEGCP. The chlorophosph'Lne mixturs was prepared by dissolving 1.67g of 2-cyanoethyl N,N-diisopropylchlorophosphoramid~te in 5 ml of d~y dichloromethane.
38c. Preparation of T~EG-terminated Anti-RAS
oncogene DNA.
The oligodeoxynucleotides of Table 3 were prepared according to a modified solid phase phosphoramidite method. GAIT, supra. The oligodeoxynucleotides were synthesized from the 3' to the 5' terminus.

' .:' " ', ` ;` :, ~ . ~ . , , W0~2/02534 PCT/US91/05~31 ~ 63 -td TaPle 3 Sequence Ref._ Code 5' X GGA GCT GGT GGC GTA X (A) 3' 7 5' XX GGA GCT GGT GGC GTA XX (A) 3' 8 5' X CCT CGA CCA CCG CAT X (A) 3' 9 5' XX CCT CGA CCA CCG CAT XX rA) 3' lO
5' CCT CGA CCA CCG CAT 3' ll X is TEG
A, C, G ~ T represent the deoxynucleotides adenylic, cytidylic, guanidylic and thymidylic acids, respectively.
Either the nucleoside adenosine (7, 8, 9, lO) or thymidine (ll) was attached to a CPG solid support using a succinate linkage. GAIT, supra. The synthesis o~ ll proceeded in accordance with standard solid phase phosphoramidite procedures. In ~equences 7, 8, 9 and lO, synt~esis proceeded in accorclance with a modi~ied phosphora~idite procedure. The ';' hydroxyl group of the attached adenosine nucleoside wa~; reacted with trichloroacetic acid to deprotec1: the 5' hydroxyl group.
Following this deprotection step 1l the at~ached adenosine nucleoside was reacted with the ~IctiYating agent, tetrazole, and a phosphoramidite reagent comprising D~TTEGCP, prepared by the processes of steps a and b above. The activation step was followed by the capping of unreacted 5' hydroxyl groups with acetic anhydride and N-methylimidazole. The phosphorous lin~age was then oxidized with iodine ~n accordance with standard procedures.
In sequences 8 and lO, containing two TEG
residues, the deprotectin~, activating, capping and oxidizing steps were repe~ted as described above. Chain elongation proceeded via the sequential steps o deprotection, acti~ation, capping and oxid~tion as described above with the modification that the desired ` . :.~ , . ,-W092/n2534 PCT/US91/05531 nucleoside phosphoramidite reagent was substituted for the DMTTEGCP during the activation step. Following attachment of the last desired nucleoside, either one or two ~EG residues were attached at the 5' terminal in a manner analogous to the attachment of TEG at the 3' terminus.
At the end of chain assembly, the DNA strand was removed from the CPG support with concentrated ammonium hydroxide. The solution was then further treated at 55 C for 8 to 15 hours to remove all the protecting groups on the exocyclic amines of the bases.

EXAMPLE 39: Pre~aration of hexaetnvleneqlycol (HEG~-_e,~minated Anti-RAS oncoaene ~A.
Hexaethyleneg~ycol (HEG) terminated anti-RAS
oncogene DNA was prepared accordin~ to the methods of Example 38. HEG was reacted with DMTCl to form DMTHEG.
The DMTHEG was then reacted with a cyanophosphine compoùnd to form DMT~EGCP, which was used in the modified solid phase phosphoramidate synthesis method of Example 38(c) to form HEG-terminated anti-RAS oncogene DNA. The sequences o~ these oligonucleotides are set forth in the following Table 4.
Ta~le 4 sequence 5' X GGA GCT GGT GGC GTA X (A~3' 5' XX GGA GCT GGT GGC GTA XX (A)3' 5' X CCT CGA CCA CCG CAT X (A)3' 5' XX CCT CGA CCA CCG C~T XX (A)3' 5' CCT CGA CCA CCG CAT 3' X is HEG
A, C, G ~ T represent the deoxynucleotides adenylic, cytidylic, guanidylic and thymidylic acids, respectively.

.. , .: , ,~ . :

-WO 92/02534 PCl`/US91/0553 EXAMPT~ 40: ~uclease Resistance of TEG-term1nated ~nti-RAS Oncoqene DNA
The oligonucleotides of Table 4 were dissolved in water. DNA concentrations were then determined by measuring the absorbance of samples at 260 nm (on a Perkin Elmer Lambda 4C Spectrophotom2ter at ambient room temperature) and using calculated extinction coefficients ~method of Cantor and Warsaw, CRC Handbook of BiochemistrY and Molecular 8iolo~v, 3rd. ed. Vol. 1, CRC Press, page 589 (1975)].
The oligonucleotides were incubated for 2 hours at 37~C at a total strand concentration of 6 or 7 ~M in cel]. culture medium containing RPMI 1640: 20 mM N-(2-hydroxyethyl) piperazine-N'-(2-ethanesulfonic acid), pH 7.4: and 10~ fetal calf serum (FCS) (GIBC0 Laboratories, Grand Island, NY). The FCS was h~at inactivatad at 56'C ror 0.5 hour prior to use. Samples were then placed on ice and deproteinized using five extractions with 24:1 chloro~orm~:isoamyl alcohol.
Samples were either stored frozen at -20'C or immediately loaded onto a refrigl3rated ~4'C) WISP
~Waterq) HPLC autoinjector.
Oligonucleotide hydrolysis was quantitated by determining the amount of disappearance of the parent compound. Oligonucleotides (from the reaction mixture) were separated on an LKB Ultrachrome G$i dual pump c~romatography system equipped with a fixed w~velength detector ~260 nm), and recording integrator, uslng a GenPak FAX ~Waters) anion axchange colu~n equilibrated in Buffar A ~lmM EDTA; lS mM sodi~m phosphate, pH 8.5).
Column temperature was maintained at 60-C using a Waters column ov~n. Fifty ~icroliter sample in~ection volumes were used. The oligonucleotides were eluted using a linear gradient of 0~ to 100~ Buffer B (Buffer A

. ;.

WO~/02534 PCT/US91/05531 ! r ~ U V '~

containing 0.5 M NaCl) over 60 ~inutes. Buffer f}ow rate was 1 mL/min.
Following incubation (2 hrs) in the presence of fetal calf serum-associated exonuclease, no degradation of compounds 7 or 10 was observed (Table 5, see % degradation of major peak). During a similar incubation period, 87.0% and 82.1% of 9 and 8, respectively, remained. In comparison, only 24.7% o~
oligomer 11 remained after the same incubation period.
Table S
SAMPLE ID AREA-MAJOR AREA-MAJOR % DEGRADATION
PEAK/O.O MIN PEAK/2.0 HR MAJOR PE~K
7 0.2325 0.3663 0.0 9 0.3744 0.3258 13.0 8 0.2164 0.1~77 17.9 0.3642 0.3697 0.0 11 1.2861 0.3177 75.3 All four TEG-oligomers were resistant to hydrolysis by the FCS-associated exonucleases. The bis-diTEG-oligomers ~7 and 10) appeared to be completely resistant to hydrolysis. TEG-derivatized oligodeoxynucleotides represent significant improvements over unmodified compounds in terms of resistance to exonuclease hydrolysis.

EXAMPLæ 41: Abilitv of TEG-~ntisense Oli~omers to Inhibit P~otein Expression and Growth in ~uman Tum~r ~ell Lines_and_PRA
Sti~ulation o~ Pe~i~heral Blos~
~mphocytes.
It has been demonstrated by others (Heikkila, R.
et 1.~ ~at~re, 328:~45-449, 1987) that unmodified antisense oligonucleotides directed towards the initiation codon region of the c-~yc oncogene oould inhibit the expression of c-myc protein i~ PHA
stimulated peripheral blood lymphocytes (PBL) resulting W0~2/n2534 PCTIUS91/05~31 _ . 1`!'~
~, J - 67 -in a blocX in the progression o~ cells into th S-phase of the cell cycle. C-myc directed antisense DNA wa~
also 6hown to inhibit the growth of ~L-60 human erytholeukemia cells ~n vitrQ (Wickstrom, E.L., et al., Proc. Natl. Acad. Sci. USA, 85:1028-1032, 1988). The sequences shown in Table 6 were prepared and evaluated by the procedures of Example 41a and 41b.

5' AAC GTT GAG GGG CAT 3' 5' XX AAC GTT GAG GGG CAT XX A 3' (X =-TEG)- ~~~

41a. Comparison of the Effect of Modi~ied (with TEG) and Non-Modified C-MYC Anti~sensQ DNA on the Prcgression of PHA St~mulated P~3L Into the S-Phase of the Cell Cycle.
Human PBLIs were stimulated with P~A ~or 4B hours in the presence or absence of the antisense ol~gonucleotide sequences o~ Table 6. ~he percent of the~population of cells ln each treatment group ln the S-phase of the cell cycle as compared to the nontreated control was determined using standard flow cytometric techniques. The results are shown in Table 7.
TA~ 7 ~o OLIGONUCLEOTIDE CONCENTRATION%CONTROL
_ (~M~ S-PHASE

S' AAC GTT GAG GGG CAT 3~ 30 75 + 6 9 + 10 5' XX AAC GTT GAG GGG CAT XX A 3'30 80 + 4 c 6 -~ , .
..
.. .. : :
:, .. . :
.: . ~ . ..

WO 92/0;!534 PCT'/US91/05531 ~ r, The data show that the presence of TEG at both the ~' and 5' termini does not alter the inhibitory effect of the antisense DNA.

41b. Comparison of the Effect of Modified twith TEG) and Non-Modified C-M~C Antisense DNA on C-MXC
Protein Expression in MOLT-4 Human T-Cell LeuXemia Cells.
Asynchronous exponentially growing Molt-4 cells were incubated for 8 hours in the presence or absence of 60 ~M c-myc directed an~isense DNA. The cells were then ~~-~ incubated for 45 minutes in the presence of ~S-methionine and the content of c-myc protein quantitated using radi.oimmunoprecipitation with a c-myc antibody.
The results are displayed in Table 8.

~REDUCTION
OLIGONUCLEOTIDE CONCENTRATION C-MYC
I~M)PROTEIN
NONE
S' AAC GTT GAG GGG CAT 3' ~0 61.0 ~ 2.6 ~5 5' XX AAC GTT GAG GGG CAT XX A 3' 60 67.9 + 0.7 The TEG cDntaining antisense DNA was slightly more potant than the unmodified antisense DNA.
41c. Comparison of the Effect of Modified ~with TE~) and Unmodi~ied C-MYC ~ntisense DNA to Inhibit the Gro~th of Human CCRF-CEM T-Cell Leukemia Cell Growth in Vitro.
Asynchronous exponentially growing CCRF-CEM cells were incubated for 48 hours in the presence or absence of antisense DNA and then cell num~ers determined in each treatment group. The concentration of antisense . ', . . . . .

, W~2/n2534 PCT/US91/OS~31 , J -,_ - 69 -DNA required to inhibit cell growth by 50~ was then determined IIC50). Both of the modified and non-modified antisense DNAs of Table 5 displayed approximately equivalent (IC50) concentrations of 40~M.
These data demonstrate that the presence of TEG at the 3' and 5' termini of antisense DNA does not affect the ability of such antisense DNA to hybridize with and inhibit the function of target nucleic acids.

EXAMPLE 42: Additional Exonuclease Stable Oliaonucleotides.
The exonuclease stable digonucleotides set forth in Table 9 were prepared according to thP methods of Example 38.

.. , . , : , : . - .: , ~L~
5~ XX A-ACG-TTG-AGG-GGC-ATX-XA 3' XX GCC-CGC-CTC-GGT-CCC-CGC-CCX-XA
XX GGG GCG GAG TTA GGG GCG GCG GGX XA
XX GGG-GAG-GAG-GGA-GGG-GAG-GGA-XXA
XX GGG-GAG-GTG-GGT-GGG-GAG-GGT-XXA
AAG GTT GAG GGG CAT XXA
X AA-CGT-TGA-GGG-GCA-TTX-A
1o XX TTC-GCT-TAC-CAG-AGT=XXA
XX GCG-GGA-GGC-TGC-TGG-XXA
XX GGA-GGC-TGC-TGG-AGC-XXA
XX CAA-GTT-CAT-AGG-TGA-TTG-CTC-XXA
AL-CAC-TCC-TTT-AGC-AAG-XXA
AL-GAA-CGA-TTT-CCT-CAC-XXA
XX CTC-ACT-GCC-GCG-CAT-XXA
-- XX GGG~TCT-TCG-GGC-CAT-XXA - -XX GTC-GAC-CGG-T~C-CAT-XXA
XX TGT-AAC-TGC-TAT-AAA-XXA
XX GTT-CCT-CCT-CTT-TAA-XXA
XX TAC-TGC-CTT-ATA-TTC-XXA
XX TAC-TGA-CTT-ATA-TTT-XXA
XX TTT-ATA-TTC-AGT-CAT-XXA
XX TGG-GGA-GGG-TGG-GGA-GGG-TGG-GGA-AGG-XXA
XX CTT-ATA-TTC-CGT-CAT-XXA
XX TAA-CGC-CTA-TTC-TGC-XXA
XX CGT-CTT-ATC-CGC-AAT-XXA
XX TTG-CTC~TCC-TCT-GTC-XXA
XX CTG-TCT-CCT-CTC-GTT-XXA
XX ATC-TAC-TGG-CTC-CAT-XXA
XX TAC-CTC-GGT-CAT-CTA-XXA
XX ACA-CCC-~AT-TCT-G~A-ATG-GXX-A
XX GGT-AAA-GTC-TTA~ACC-CAC-AXX-A
XX TAC-GGG-GAG-TTG-~CAA-XXA

X is TEG
As will be apparent to those skilled in the art, many variations and modlfications may be made without departing ~rom the spirit and scopç of the present invention.

.- - ~
~: ,; , ,. ;` , , ;. . !

:: ', '' '` ' '. . . `'

Claims (24)

WE CLAIM:
1. A compound comprising an oligonucleoside sequence of from about 6 to about 200 bases having a three atom internucleoside linkage of the formula:
-D-D-D-;
where each D is independently CHR, oxygen or NR6 wherein R is independently hydrogen, OH, SH or NH2, wherein R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D
is oxygen or NR6.
2. The compound according to claim 1 which has a diol at either or both termini.
3. The compound according to claim 1 wherein the diol is tetraethyleneglycol or hexaethyleneglycol.
4. A compound comprising an oligonucleoside sequence of the formula:

where W is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH, or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6;
each W' is independently W or ;

each R1 is independently OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl or NHR4 wherein R4 is C1-C12 acyl;
each y is independently H or OH;
each B is independently adenine, cytosine, guanine, thymine, uracil or modifications thereof;
j is an integer from 1 to about 200;
k is 0 or an integer from 1 to about 197; and q is 0 or an integer from 1 to about 197, with the proviso that the sum of j + k + q is from about 4 to about 200.
5. A compound comprising an oligonucleoside sequence of from about 6 to about 200 bases having the formula:

where each z is independently R' or ;

where w is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH
or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6;
each W' is independently W or ;

each R1 is independently OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl or NHR4 wherein R4 is C1-C12 acyl;
each R5 is independently hydrogen or C1-C12 alkyl;
each y is independently H or OH;
each B is independently adenine, cytosine, guanine, thymine, uracil or a modification thereof;
each e and f is independently an integer from 0 to 50, with the proviso that at least one of e and f be at least 1:
j is an integer from 1 to about 200;
k is 0 or an integer from 1 to about 197; and each m and n is independently an integer from 1 to 200;
each p is independently 2 to 4; And q is 0 or an integer from 1 to about 197, with the proviso that the sum of j + k + q is from about 4 to about 200.
6. A nuclease resistant compound comprising an oligonucleotide sequence of from about 9 to about 200 bases having a diol at either or both termini.
7. A compound according to claim 6 wherein the diol is hexaethyleneglycol or tetraethyleneglycol.
8. A compound comprising an oligonucleotide of the formula:

;

where R is OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl, or NHR4 wherein R4 is C1-C12 acyl;
R1 is hydrogen or C1-C12 alkyl;
oligo (N) is native or modified oligonucleotide sequence of from about 9 to about 200 bases;
each e and f is independently 0 to 50, with the proviso that at least one of e and f be at least 1;
each m and n is independently 1 to 200: and each p is independently 2 to 4.
9. A compound comprising an oligonucleotide of the formula:

;

where R is OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl, or NHR4 wherein R4 is C1-C12 acyl;

oligo N is an oligonucleotide sequence of from about 9 to about 50 bases; and e and f are independently 0 to 50, with the proviso that at least one of e and f be at least one; and and n are independently 0 to 200 with the proviso that at least one of m and n be 1 to 200
10. A method of inhibiting nuclease degradation of compounds comprising preparing oligonucleoside sequences of from about 6 to about 200 bases having a three atom internucleoside linkage of the formula:
-D-D-D-;
where each D is independently CHR, oxygen or NR6 wherein R is independently hydrogen, OH, SH or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6.
11. The method according to claim 10 wherein the oligonucleoside sequences have the formula:

where W is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH
or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6;

each W' is independently W or ;

each R1 is independently OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl or NHR4 wherein R4 is C1-C12 acyl;
each y is independently H or OH;
each B is independently adenine, cytosine, guanine, thymine, uracil or modifications thereof;
j is an integer from 1 to about 200:
k is 0 or an integer from 1 to about 197; and q is 0 or an integer from 1 to about 197, with the proviso that the sum of j + k + q is from about 4 to about 200.
12. A method of stabilizing nucleotide or oligonucleoside sequences comprising attaching a diol to either or both termini of said nucleotide or oligonucleoside sequence.
13. A method according to claim 12 wherein a trityldiolcyanophosphine is used to protect the 5' terminus of said compound.
14. A composition useful for inhibiting gene expression comprising a physiologically acceptable carrier and a compound comprising oligonucleoside sequences of from about 6 to about 200 bases having a non-phosphate containing three atom internucleoside linkage of the formula:
-D-D-D-;
where each D is independently CHR, oxygen or NR6 wherein R is independently hydrogen, OH, SH or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6.
15. The composition according to claim 14 wherein the oligonucleoside sequence has the formula:

where W is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH
or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6;

each W' is independently W or ;

each R1 is independently OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl or NHR4 wherein R4 is C1-C12 acyl;
each y is independently H or OH;
each B is independently adenine, cytosine, guanine, thymine, uracil or modifications thereof;
j is an integer from 1 to about 200;
k is 0 or an integer from 1 to about 197; and q is 0 or an integer from 1 to about 197, with the proviso that the sum of j + k + q is from about 4 to about 200.
16. A composition useful for inhibiting gene expression comprising a nuclease resistant compound comprising an oligonucleotide sequence of from about 9 to about 200 bases having a diol at either or both termini and a physiologically acceptable carrier.
17. A composition according to claim 16 wherein the diol is hexaethyleneglycol or tetraethyleneglycol.
18. A composition according to claim 16 wherein the compound comprises an oligonucleotide of the formula:

;

where R is OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl, or NHR4 wherein R4 is C1-C12 acyl:
R1 is hydrogen or C1-C12 alkyl;
oligo (N) is native or modified oligonucleotide sequence of from about 9 to about 200 bases;
each e and f is independently 0 to 50, with the proviso that at least one of e and f be at least 1;
each m and n is independently 1 to 200; and each p is independently 2 to 4.
19. A method of inhibiting gene expression in a mammal in need of such treatment comprising administering to said mammal an effective amount of a compound comprising an oligonucleoside sequence of from about 6 to about 200 bases having a three atom internucleoside linkage of the formula:
-D-D-D-;
where each D is independently CHR, oxygen or NR6 wherein R is independently hydrogen, OH, SH or NH2, wherein R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D
is oxygen or NR6.
20. The method according to claim 19 wherein the oligonucleoside sequence has the formula:

where W is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH
or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6;
each W' is independently W or ;

each R1 is independently OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl or NHR4 wherein R4 is C1-C12 acyl;
each y is independently H or OH;
each B is independently adenine, cytosine, guanine, thymine, uracil or modifications thereof;
j is an integer from 1 to about 200;
X is 0 or an integer from 1 to about 197; and q is 0 or an integer from 1 to about 197, with the proviso that the sum of j + k + q is from about 4 to about 200.
21. The method according to claim 20 wherein the compound has a diol at either or both termini.
22. A method according to claim 19 wherein the compound comprises an oligonucleotide of the formula ;

where R is OH, SH, NR2R3 wherein R2 and R3 are independently hydrogen or C1-C6 alkyl, or NHR4 wherein R4 is C1-C12 acyl;
R1 is hydrogen or C1-C12 alkyl;
oligo (N) is native or modified oligonucleotide sequence of from about 9 to about 200 bases;
each e and f is independently 0 to 50, with the proviso that at least one of e and f are at least 1;
each m and n is independently 1 to 200; and each p is independently 2 to 4.
23. A compound comprising a nucleoside dimer of the formula:

where W is -D-D-D- wherein each D is independently CHR, oxygen or NR6, wherein R is independently hydrogen, OH, SH
or NH2, R6 is hydrogen or C1-C2 alkyl, with the proviso that only one D is oxygen or NR6;
each B is independently adenine, cytosine, guanine, thymine, uracil or a modification thereof;
R7 is OH, t-butyldimethylsilyloxy or a phosphoramidite;
and R8 is OH, a protecting group or t-butyldimethylsilyloxy.
24. A compound of the formula:
5' Xn CCT CGA CCA CCG CAT Xn (A) 3';
where X is tetraethyleneglycol; and n is 1 or 2.
CA002088673A 1990-08-03 1991-08-02 Compounds and methods for inhibiting gene expression Abandoned CA2088673A1 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US07/562,180 US5245022A (en) 1990-08-03 1990-08-03 Exonuclease resistant terminally substituted oligonucleotides
US562,180 1990-08-03
US58245690A 1990-09-13 1990-09-13
US58245790A 1990-09-13 1990-09-13
US58228790A 1990-09-13 1990-09-13
US582,287 1990-09-13
US582,456 1990-09-13
US582,457 1990-09-13
US68278491A 1991-04-09 1991-04-09
US682,784 1991-04-09

Publications (1)

Publication Number Publication Date
CA2088673A1 true CA2088673A1 (en) 1992-02-04

Family

ID=27541908

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002088673A Abandoned CA2088673A1 (en) 1990-08-03 1991-08-02 Compounds and methods for inhibiting gene expression

Country Status (20)

Country Link
US (1) US5677439A (en)
EP (1) EP0541722B1 (en)
JP (1) JPH06502300A (en)
KR (1) KR100211552B1 (en)
AT (1) ATE131827T1 (en)
AU (2) AU667459B2 (en)
BR (1) BR9106729A (en)
CA (1) CA2088673A1 (en)
DE (1) DE69115702T2 (en)
DK (1) DK0541722T3 (en)
ES (1) ES2083593T3 (en)
FI (1) FI930455A (en)
GR (1) GR3018881T3 (en)
HU (2) HU217036B (en)
IL (3) IL99066A (en)
MY (1) MY107332A (en)
NZ (1) NZ239247A (en)
PT (1) PT98562B (en)
TW (1) TW222641B (en)
WO (1) WO1992002534A2 (en)

Families Citing this family (784)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5914396A (en) * 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US5378825A (en) * 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5610289A (en) * 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5783682A (en) 1990-07-27 1998-07-21 Isis Pharmaceuticals, Inc. Oligonucleotide mimics having nitrogen-containing linkages
US6087482A (en) * 1990-07-27 2000-07-11 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5792844A (en) * 1990-07-27 1998-08-11 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent nitrogen atoms
US5623070A (en) * 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5677437A (en) * 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5618704A (en) * 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5386023A (en) * 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
PT98562B (en) * 1990-08-03 1999-01-29 Sanofi Sa PROCESS FOR THE PREPARATION OF COMPOSITIONS THAT UNDERSEAD SEEDS OF NUCLEO-SIDS WITH NEAR 6 TO NEAR 200 NUCLEASE-RESISTANT BASES
US5965722A (en) * 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
US6030954A (en) * 1991-09-05 2000-02-29 University Of Connecticut Targeted delivery of poly- or oligonucleotides to cells
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
US5817781A (en) * 1992-06-01 1998-10-06 Gilead Sciences, Inc. Modified internucleoside linkages (II)
EP0690726B1 (en) * 1993-01-07 2001-11-14 Thomas Jefferson University Antisense inhibition of c-myc to modulate the proliferation of smooth muscle cells
GB9304618D0 (en) * 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
WO1994022893A1 (en) * 1993-03-30 1994-10-13 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent nitrogen atoms
CA2159632A1 (en) * 1993-03-31 1994-10-13 Ashis Kumar Saha Novel 5'-substituted nucleosides and oligomers produced therefrom
EP0701564A1 (en) * 1993-03-31 1996-03-20 Sanofi Bifunctional nucleosides, oligomers thereof, and methods of making and using the same
CA2170869C (en) 1993-09-03 1999-09-14 Phillip Dan Cook Amine-derivatized nucleosides and oligonucleosides
US5886177A (en) * 1994-01-11 1999-03-23 Isis Pharmaceuticals, Inc. Phosphate linked oligomers
US6448373B1 (en) 1994-01-11 2002-09-10 Isis Pharmaceuticals, Inc. Phosphate linked oligomers formed of monomeric diols and processes for preparing same
WO1995020597A1 (en) * 1994-01-26 1995-08-03 Ciba-Geigy Ag Modified oligonucleotides
EP1260586A3 (en) * 1994-02-23 2004-04-28 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting the expression of disease related genes
FR2733500B1 (en) * 1995-04-28 1997-07-25 Centre Nat Rech Scient NOVEL ANTISENS DIRECTED AGAINST RAS, PREPARATION AND USES
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20040161844A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Sugar and backbone-surrogate-containing oligomeric compounds and compositions for use in gene modulation
US20030044941A1 (en) 1996-06-06 2003-03-06 Crooke Stanley T. Human RNase III and compositions and uses thereof
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US6716625B1 (en) 1997-04-16 2004-04-06 Claude Selitrennikoff Histidine kinases of Aspergillus and other fungal species, related compositions, and methods of use
DE69834038D1 (en) 1997-07-01 2006-05-18 Isis Pharmaceutical Inc COMPOSITIONS AND METHOD FOR THE ADMINISTRATION OF OLIGONUCLEOTIDES OVER THE DISHES
AU770896C (en) 1998-09-29 2006-09-07 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
US20040186071A1 (en) 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
EP1080226A4 (en) 1998-05-21 2004-04-21 Isis Pharmaceuticals Inc Compositions and methods for topical delivery of oligonucleotides
CA2329130A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6867294B1 (en) * 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6225293B1 (en) 1998-09-02 2001-05-01 Isis Pharmaceuticals, Inc. Methods and compounds for tracking the biodistribution of macromolecule-carrier combinations
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
US6300320B1 (en) 1999-01-05 2001-10-09 Isis Pharmaceuticals, Inc. Modulation of c-jun using inhibitors of protein kinase C
US6127124A (en) 1999-01-20 2000-10-03 Isis Pharmaceuticals, Inc. Fluorescence based nuclease assay
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US7534605B2 (en) 1999-06-08 2009-05-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem CD44 polypeptides, polynucleotides encoding same, antibodies directed thereagainst and method of using same for diagnosing and treating inflammatory diseases
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US20030190644A1 (en) 1999-10-13 2003-10-09 Andreas Braun Methods for generating databases and databases for identifying polymorphic genetic markers
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
EP2202241A3 (en) * 2000-04-13 2010-07-28 Thomas N. Wight Therapeutic compounds and methods for modulating V3, a versican isoform
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
US6686188B2 (en) 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
US8568766B2 (en) 2000-08-24 2013-10-29 Gattadahalli M. Anantharamaiah Peptides and peptide mimetics to treat pathologies associated with eye disease
DE10046184A1 (en) * 2000-09-18 2002-04-04 November Ag Molekulare Medizin Universally applicable method for parallel detection of nucleic acids, by forming a primer extension product that includes single-stranded segment for specific hybridization
WO2002030465A2 (en) 2000-10-12 2002-04-18 University Of Rochester Compositions that inhibit proliferation of cancer cells
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US6573051B2 (en) * 2001-03-09 2003-06-03 Molecular Staging, Inc. Open circle probes with intramolecular stem structures
ES2328796T3 (en) 2001-03-14 2009-11-18 Myriad Genetics, Inc. TSG101-GAG INTERACTION AND USE OF THE SAME.
KR100788092B1 (en) 2001-06-20 2007-12-21 제넨테크, 인크. Compositions and Methods for the Diagnosis and Treatment of Tumor
US7785610B2 (en) * 2001-06-21 2010-08-31 Dynavax Technologies Corporation Chimeric immunomodulatory compounds and methods of using the same—III
CA2790034A1 (en) 2001-06-21 2003-01-03 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
DK2423335T3 (en) 2001-06-21 2014-08-18 Dynavax Tech Corp CHEMICAL IMMUNAL MODULATOR RELATIONS AND PROCEDURES FOR USE THEREOF
CA2452458A1 (en) * 2001-07-03 2003-01-16 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. 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
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
CA2455424A1 (en) * 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
US20040096880A1 (en) * 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
DE60238143D1 (en) 2001-09-18 2010-12-09 Genentech Inc COMPOSITIONS AND METHODS FOR THE DIAGNOSIS OF TUMORS
US7070933B2 (en) * 2001-09-28 2006-07-04 Gen-Probe Incorporated Inversion probes
NZ585001A (en) 2001-10-09 2011-08-26 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expression
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
MXPA04006554A (en) 2002-01-02 2005-03-31 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
AU2003203079B9 (en) * 2002-02-04 2009-01-15 Oncothyreon Inc. Immunostimulatory, covalently lipidated oligonucleotides
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
EP2011886A3 (en) 2002-04-16 2009-02-11 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7176181B2 (en) * 2002-05-21 2007-02-13 Yeda Research And Development Co. Ltd. Compositions and methods of using galectin-8 as an inhibitor of tumor cell growth
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
US20050287648A1 (en) 2002-08-05 2005-12-29 University Of Rochester Protein Transducing Domain/Deaminase Chimeric Proteins, Related Compounds, and Uses Thereof
CA2498777C (en) 2002-09-13 2015-01-13 Replicor, Inc. Non-sequence complementary antiviral oligonucleotides
EP2233494A3 (en) * 2002-09-20 2013-05-29 Yale University Riboswitches, methods for their use, and compositions for use with riboswitches
EP2272958A1 (en) 2002-09-26 2011-01-12 ISIS Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
CA2504720C (en) 2002-11-05 2013-12-24 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
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
US8604183B2 (en) 2002-11-05 2013-12-10 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
CA2505801A1 (en) 2002-11-13 2004-05-27 Rosanne Crooke Antisense modulation of apolipoprotein b expression
SI1569695T1 (en) 2002-11-13 2013-08-30 Genzyme Corporation Antisense modulation of apolipoprotein b expression
US20060009378A1 (en) * 2002-11-14 2006-01-12 Itshak Golan Novel galectin sequences and compositions and methods utilizing same for treating or diagnosing arthritis and other chronic inflammatory diseases
AU2003298650B2 (en) 2002-11-15 2010-03-11 Musc Foundation For Research Development Complement receptor 2 targeted complement modulators
WO2004046330A2 (en) 2002-11-15 2004-06-03 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
WO2004047749A2 (en) 2002-11-21 2004-06-10 University Of Utah Research Foundation Purinergic modulation of smell
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US20040110698A1 (en) * 2002-12-10 2004-06-10 Kimron Veterinary Institute Oligonucleotides and methods using same for treating cox-ll associated diseases
US20040115643A1 (en) * 2002-12-12 2004-06-17 Lizardi Paul M. Thermodynamic equilibrium extension of primers
US20040121338A1 (en) * 2002-12-19 2004-06-24 Alsmadi Osama A. Real-time detection of rolling circle amplification products
US9487823B2 (en) 2002-12-20 2016-11-08 Qiagen Gmbh Nucleic acid amplification
CA2510587A1 (en) 2002-12-20 2004-07-15 Qiagen Gmbh Nucleic acid amplification
EP1597366B1 (en) 2003-02-11 2012-11-21 Antisense Therapeutics Ltd Modulation of insulin like growth factor i receptor expression
WO2004076621A2 (en) 2003-02-27 2004-09-10 Yeda Research And Development Co. Ltd. Compositions of nucleic acids for treating and detecting influenza virus
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US8043834B2 (en) 2003-03-31 2011-10-25 Qiagen Gmbh Universal reagents for rolling circle amplification and methods of use
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
EP2241572A3 (en) 2003-06-03 2011-04-06 Eli Lilly And Company Modulation of survivin expression
US20040248103A1 (en) * 2003-06-04 2004-12-09 Feaver William John Proximity-mediated rolling circle amplification
ES2905724T3 (en) 2003-06-13 2022-04-11 Alnylam Europe Ag Double-stranded ribonucleic acid with high efficacy in an organism
US7683036B2 (en) 2003-07-31 2010-03-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
CA2538252C (en) 2003-09-18 2014-02-25 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
EP2256201A3 (en) 2003-09-18 2012-07-04 Isis Pharmaceuticals, Inc. Modulation of eIF4E expression
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
EP2412725A3 (en) 2003-11-17 2012-04-25 Genentech, Inc. Antibodies against CD79b for the treatment of tumor of hematopoeitic origin
WO2006054296A2 (en) 2004-11-17 2006-05-26 Spectrum Dynamics Llc Methods of detecting prostate cancer
JP2007520222A (en) 2004-01-20 2007-07-26 アイシス ファーマシューティカルズ インコーポレイテッド Regulation of glucocorticoid receptor expression
US8778900B2 (en) 2004-01-22 2014-07-15 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP1 expression
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US7842459B2 (en) 2004-01-27 2010-11-30 Compugen Ltd. Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
WO2005089268A2 (en) 2004-03-15 2005-09-29 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of rna by rnase h
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
AU2005230684B2 (en) 2004-04-05 2011-10-06 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
JP4584987B2 (en) 2004-04-30 2010-11-24 アルニラム ファーマスーティカルズ インコーポレイテッド Oligonucleotides containing C5-modified pyrimidines
CA2564251C (en) 2004-05-21 2018-04-10 The Uab Research Foundation Variable lymphocyte receptors, related polypeptides and nucleic acids, and uses thereof
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
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
PL1809303T3 (en) 2004-09-23 2019-11-29 Arc Medical Devices Inc Pharmaceutical compositions and methods relating to inhibiting fibrous adhesions or inflammatory disease using low sulphate fucans
WO2006050999A2 (en) * 2004-11-15 2006-05-18 Obe Therapy Biotechnology S.A.S Methods of reducing body fat
EP1859040A2 (en) * 2005-02-25 2007-11-28 Isis Pharmaceuticals, Inc. Compositions and their uses directed to il-4r alpha
JP2008535796A (en) 2005-03-10 2008-09-04 ジェネンテック・インコーポレーテッド Methods and compositions for regulating vascular integrity
US8309303B2 (en) 2005-04-01 2012-11-13 Qiagen Gmbh Reverse transcription and amplification of RNA with simultaneous degradation of DNA
WO2007008300A2 (en) 2005-05-31 2007-01-18 ECOLE POLYTECHNIQUE FéDéRALE DE LAUSANNE Triblock copolymers for cytoplasmic delivery of gene-based drugs
WO2006133022A2 (en) 2005-06-03 2006-12-14 The Johns Hopkins University Compositions and methods for decreasing microrna expression for the treatment of neoplasia
WO2006138145A1 (en) 2005-06-14 2006-12-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
CN101273130B (en) * 2005-07-07 2012-05-30 耶路撒冷希伯来大学伊森姆研究发展公司 Nucleic acid agents for downregulating H19, and methods of using same
DK2239327T3 (en) 2005-08-11 2015-05-18 Synthetic Genomics Inc A method for in vitro recombination
CA2617693A1 (en) 2005-08-17 2007-02-22 Medexis S.A. Composition and method for determination of ck19 expression
EP2338992A3 (en) 2005-08-29 2011-10-12 Regulus Therapeutics, Inc Antisense compounds having enhanced anti-microRNA activity
WO2007027775A2 (en) 2005-08-29 2007-03-08 Isis Pharmaceuticals, Inc. Methods for use in modulating mir-122a
EP1762627A1 (en) 2005-09-09 2007-03-14 Qiagen GmbH Method for the activation of a nucleic acid for performing a polymerase reaction
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for diagnosis of preeclamsia
US8080534B2 (en) 2005-10-14 2011-12-20 Phigenix, Inc Targeting PAX2 for the treatment of breast cancer
AU2006304321B2 (en) 2005-10-14 2012-10-04 Musc Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
EP1941059A4 (en) 2005-10-28 2010-11-03 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of huntingtin gene
US20100069461A1 (en) 2005-11-09 2010-03-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor v leiden mutant gene
WO2007062380A2 (en) 2005-11-21 2007-05-31 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
EP1969143A4 (en) * 2005-12-20 2009-07-22 Isis Pharmaceuticals Inc Double stranded nucleic acid molecules targeted to il-4 receptor alpha
JP2009524411A (en) * 2005-12-21 2009-07-02 イェール ユニバーシティー Methods and compositions related to the regulation of riboswitches
EP1976567B1 (en) 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
CA2637267A1 (en) 2006-01-16 2007-07-19 Compugen Ltd. Novel nucleotide and amino acid sequences, and methods of use thereof for diagnosis
DK2314594T3 (en) 2006-01-27 2014-10-27 Isis Pharmaceuticals Inc 6-modified bicyclic nucleic acid analogues
AU2007211082B2 (en) 2006-01-27 2012-09-27 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for the use in modulation of microRNAs
WO2007115168A2 (en) 2006-03-31 2007-10-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 gene
WO2007125173A2 (en) 2006-05-03 2007-11-08 Baltic Technology Development, Ltd. Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease
CA3044969A1 (en) 2006-05-05 2007-12-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gene expression
CA2651453C (en) 2006-05-11 2014-10-14 Isis Pharmaceuticals, Inc. 5'-modified bicyclic nucleic acid analogs
NZ572666A (en) 2006-05-11 2010-11-26 Alnylam Pharmaceuticals Inc Compositions comprising double stranded rna and methods for inhibiting expression of the pcsk9 gene
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
EP2023937B1 (en) 2006-05-19 2011-10-12 Alnylam Pharmaceuticals Inc. Rnai modulation of aha and therapeutic uses thereof
WO2007137220A2 (en) 2006-05-22 2007-11-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of ikk-b gene
WO2008011473A2 (en) 2006-07-19 2008-01-24 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hbxip
US20100137440A1 (en) * 2006-09-11 2010-06-03 Yale University Lysine riboswitches, structure-based compound design with lysine riboswitches, and methods and compositions for use of and with lysine riboswitches
US20090209478A1 (en) 2006-09-21 2009-08-20 Tomoko Nakayama Compositions and methods for inhibiting expression of the hamp gene
AU2007303205A1 (en) 2006-10-03 2008-04-10 Tekmira Pharmaceuticals Corporation Lipid containing formulations
US20100166743A1 (en) 2006-10-06 2010-07-01 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
WO2008136852A2 (en) 2006-11-01 2008-11-13 University Of Rochester Methods and compositions related to the structure and function of apobec3g
CA2670563A1 (en) * 2006-11-27 2008-06-05 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
BRPI0720038A2 (en) 2006-12-11 2013-12-24 Univ Utah Res Found METHODS TO INHIBIT VASCULAR PERMEABILITY IN TISSUE, TO TRIAL OR EVALUATE AN AGENT THAT INHIBITS VASCULAR PERMEABILITY, TO TREAT OR PREVENT RESPIRATORY ANGUS, SYNDROME TO PREMATULAR DIABETARY RATE, INDIVIDUALS WITH REPULSIVE OR MIMETIC SUGGESTIONS AND TO PROMOTE ANGIOGENESIS IN A FABRIC, ISOLATED POLYPEPTIDE, ISOLATED NUCLEIC ACID, AND, VECTOR
EA200900782A1 (en) * 2006-12-14 2009-12-30 Новартис Аг COMPOSITIONS AND METHODS INTENDED FOR THE TREATMENT OF MUSCULAR AND CARDIOVASCULAR DISORDERS
WO2008094370A2 (en) 2006-12-22 2008-08-07 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
CA2675967A1 (en) * 2007-01-16 2008-07-24 Yissum Research Development Company Of The Hebrew University Of Jerusale M Nucleic acid constructs and methods for specific silencing of h19
US20100196403A1 (en) * 2007-01-29 2010-08-05 Jacob Hochman Antibody conjugates for circumventing multi-drug resistance
US8507200B2 (en) 2007-02-09 2013-08-13 Northwestern University Particles for detecting intracellular targets
US20100167290A1 (en) * 2007-02-27 2010-07-01 Robert Elghanian Molecule attachment to nanoparticles
EP2471925A1 (en) 2007-03-22 2012-07-04 Yale University Methods and compositions related to riboswitches that control alternative splicing
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
WO2008121604A2 (en) 2007-03-29 2008-10-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the ebola
WO2008141275A1 (en) 2007-05-11 2008-11-20 The Johns Hopkins University Biomarkers for melanoma
US20100286082A1 (en) 2007-05-29 2010-11-11 Yale University Riboswitches and methods and compositions for use of and with riboswitches
CA2687684A1 (en) * 2007-05-29 2008-12-11 Yale University Methods and compositions related to riboswitches that control alternative splicing and rna processing
WO2008151049A2 (en) 2007-05-30 2008-12-11 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
EP2170917B1 (en) 2007-05-30 2012-06-27 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
US7807372B2 (en) * 2007-06-04 2010-10-05 Northwestern University Screening sequence selectivity of oligonucleotide-binding molecules using nanoparticle based colorimetric assay
WO2008154401A2 (en) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
CA2692579C (en) 2007-07-05 2016-05-03 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
EP2164966A2 (en) 2007-07-05 2010-03-24 Novartis Ag Dsrna for treating viral infection
KR101654007B1 (en) 2007-08-15 2016-09-05 아이오니스 파마수티컬즈, 인코포레이티드 Tetrahydropyran nucleic acid analogs
AU2008296487A1 (en) 2007-08-28 2009-03-12 The Uab Research Foundation Synthetic apolipoprotein E mimicking polypeptides and methods of use
DK2682400T5 (en) 2007-08-28 2017-11-27 Uab Research Foundation Synthetic apolipoprotein E mimic polypeptides and methods of use
PT2769729T (en) 2007-09-04 2019-05-08 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2009039466A1 (en) 2007-09-20 2009-03-26 Vanderbilt University Free solution measurement of molecular interactions by backscattering interferometry
WO2009039442A1 (en) 2007-09-21 2009-03-26 California Institute Of Technology Nfia in glial fate determination, glioma therapy and astrocytoma treatment
CA2700953A1 (en) 2007-10-02 2009-04-09 Amgen Inc. Increasing erythropoietin using nucleic acids hybridizable to micro-rna and precursors thereof
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
USRE47320E1 (en) 2007-11-20 2019-03-26 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
EP2245159A2 (en) 2007-12-10 2010-11-03 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor vii gene
EP3100718B1 (en) 2008-01-02 2019-11-27 Arbutus Biopharma Corporation Improved compositions and methods for the delivery of nucleic acids
EP2265627A2 (en) 2008-02-07 2010-12-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
WO2009111658A2 (en) 2008-03-05 2009-09-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 and vegf genes
WO2009117589A1 (en) 2008-03-21 2009-09-24 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucleosides and methods for their use
EP2982753B1 (en) 2008-04-18 2018-06-06 Baxter International Inc. Microsphere-based composition for preventing and/or reversing new-onset autoimmune diabetes
EP2280995A2 (en) * 2008-04-29 2011-02-09 Wyeth LLC Methods for treating inflammation
US8341614B2 (en) * 2008-06-06 2012-12-25 Apple Inc. Memory management for closures
EP3081648A1 (en) 2008-08-25 2016-10-19 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
US8318693B2 (en) 2008-09-02 2012-11-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mutant EGFR gene
EP2356129B1 (en) 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Substituted alpha-l-bicyclic nucleosides
WO2010036696A1 (en) 2008-09-24 2010-04-01 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acid analogs
EP3109321B1 (en) 2008-09-25 2019-05-01 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
US9139554B2 (en) 2008-10-09 2015-09-22 Tekmira Pharmaceuticals Corporation Amino lipids and methods for the delivery of nucleic acids
AU2009305636A1 (en) 2008-10-15 2010-04-22 Ionis Pharmaceuticals, Inc. Modulation of Factor 11 expression
NO2937418T3 (en) 2008-10-20 2018-03-17
CA2741294C (en) 2008-10-24 2018-04-24 Isis Pharmaceuticals, Inc. 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
US8987435B2 (en) 2008-10-24 2015-03-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
MX2011005429A (en) 2008-11-24 2011-06-21 Univ Northwestern Polyvalent rna-nanoparticle compositions.
WO2010061393A1 (en) 2008-11-30 2010-06-03 Compugen Ltd. He4 variant nucleotide and amino acid sequences, and methods of use thereof
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
RU2746478C2 (en) 2008-12-04 2021-04-14 КьюРНА, Инк. Treatment of tumors of diseases related to the genom-suppressor by therapy of natural transcript inhibition in anti-significant orientation regarding this gene
EP2370580B1 (en) 2008-12-04 2019-09-11 CuRNA, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
EP2633854B1 (en) 2008-12-05 2015-09-16 Yeda Research And Development Co. Ltd. miRNA-9 or miRNA-9* for use in treating ALS
CA2746514C (en) 2008-12-10 2018-11-27 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
EP2376633A1 (en) * 2008-12-17 2011-10-19 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
EP3243504A1 (en) 2009-01-29 2017-11-15 Arbutus Biopharma Corporation Improved lipid formulation
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8536320B2 (en) 2009-02-06 2013-09-17 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
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)
PT2396038E (en) 2009-02-12 2016-02-19 Curna Inc Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
WO2010099341A1 (en) 2009-02-26 2010-09-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mig-12 gene
EP2403863B1 (en) 2009-03-02 2013-08-28 Alnylam Pharmaceuticals Inc. Nucleic acid chemical modifications
CA2754749C (en) 2009-03-04 2019-04-30 Opko Curna, Llc Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt1
NZ594995A (en) 2009-03-12 2013-06-28 Alnylam Pharmaceuticals Inc LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF HUMAN KINESIN FAMILY MEMBER 11 (Eg5) AND VASCULAR ENDOTHELIAL GROWTH FACTOR (VEGF) GENES
ES2656290T3 (en) 2009-03-16 2018-02-26 Curna, Inc. Treatment of diseases related to nuclear factor (derived from erythroid 2) similar to 2 (NRF2) by inhibition of natural antisense transcript to NRF2
EP2408920B1 (en) 2009-03-17 2017-03-08 CuRNA, Inc. Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
WO2010120420A1 (en) 2009-04-15 2010-10-21 Northwestern University Delivery of oligonucleotide-functionalized nanoparticles
EP3248618A1 (en) 2009-04-22 2017-11-29 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
CA2760589C (en) 2009-05-01 2019-08-20 Joseph Collard Treatment of hemoglobin (hbf/hbg) related diseases by inhibition of natural antisense transcript to hbf/hbg
NZ621981A (en) 2009-05-05 2015-09-25 Tekmira Pharmaceuticals Corp Lipid compositions
CA3045126A1 (en) 2009-05-05 2010-11-11 Arbutus Biopharma Corporation Methods of delivering oligonucleotides to immune cells
KR101722541B1 (en) 2009-05-06 2017-04-04 큐알엔에이, 인크. Treatment of tristetraproline(ttp) related diseases by inhibition of natural antisense transcript to ttp
CN103223177B (en) 2009-05-06 2016-08-10 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
EP2430161A1 (en) 2009-05-15 2012-03-21 Yale University Gemm riboswitches, structure-based compound design with gemm riboswitches, and methods and compositions for use of and with gemm riboswitches
CA2762369C (en) 2009-05-18 2021-12-28 Joseph Collard Treatment of reprogramming factor related diseases by inhibition of natural antisense transcript to a reprogramming factor
EP2432882B1 (en) 2009-05-22 2019-12-25 CuRNA, Inc. TREATMENT OF TRANSCRIPTION FACTOR E3 (TFE3) and INSULIN RECEPTOR SUBSTRATE 2 (IRS2) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO TFE3
CA2764683A1 (en) 2009-05-28 2010-12-02 Joseph Collard Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
HUE056773T2 (en) 2009-06-10 2022-03-28 Arbutus Biopharma Corp Improved lipid formulation
EP2443237B1 (en) 2009-06-16 2017-02-22 CuRNA, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
JP6128846B2 (en) 2009-06-16 2017-05-17 クルナ・インコーポレーテッド Treatment of PON1 gene-related diseases by suppression of natural antisense transcripts against paraoxonase (PON1)
CN102597238B (en) 2009-06-24 2016-06-29 库尔纳公司 The relevant disease of TNFR2 is treated by suppressing for the natural antisense transcript of tumor necrosis factor receptor 2 (TNFR2)
CN102482672B (en) 2009-06-26 2016-11-09 库尔纳公司 By suppressing the natural antisense transcript treatment Down syndrome gene-associated diseases of Down syndrome gene
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
WO2011005861A1 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
CN102762731B (en) 2009-08-05 2018-06-22 库尔纳公司 By inhibiting to treat insulin gene (INS) relevant disease for the natural antisense transcript of insulin gene (INS)
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
AP3574A (en) 2009-08-14 2016-02-08 Alnylam Pharmaceuticals Inc Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
US20120157324A1 (en) 2009-08-17 2012-06-21 Yale University Methylation biomarkers and methods of use
JP5964232B2 (en) 2009-08-25 2016-08-03 カッパーアールエヌエー,インコーポレイテッド Treatment of IQGAP-related diseases by inhibition of natural antisense transcripts against 'IQ motif-containing GTPase-activating protein' (IQGAP)
JP5887270B2 (en) 2009-09-02 2016-03-16 ジェネンテック, インコーポレイテッド Mutant SMOOTHENED AND METHOD OF USING THE SAME
EP2488646B1 (en) 2009-10-14 2017-12-06 Yissum Research Development Company of the Hebrew University of Jerusalem Ltd. Compositions for controlling varroa mites in bees
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
RU2539772C2 (en) 2009-10-22 2015-01-27 Дженентек, Инк. Methods and compositions for hepsin modulation of macrophage-stimulating protein
JP6147502B2 (en) 2009-10-27 2017-06-14 スウィフト バイオサイエンシーズ, インコーポレイテッド Polynucleotide primers and probes
EP2494075B1 (en) 2009-10-30 2018-04-04 Northwestern University Templated nanoconjugates
AU2010315867A1 (en) 2009-11-03 2012-06-21 University Of Virginia Patent Foundation Versatile, visible method for detecting polymeric analytes
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
KR101944119B1 (en) 2009-11-13 2019-01-30 사렙타 쎄러퓨틱스 인코퍼레이티드 Antisense antiviral compound and method for treating influenza viral infection
CA2781469A1 (en) 2009-11-23 2011-05-26 Swift Biosciences, Inc. Devices to extend single stranded target molecules
CN103755809B (en) 2009-11-30 2016-06-01 霍夫曼-拉罗奇有限公司 The antibody of the tumour of SLC34A2 (TAT211=SEQID2) is expressed in treatment and diagnosis
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
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.
US8940708B2 (en) 2009-12-23 2015-01-27 Curna, Inc. Treatment of hepatocyte growth factor (HGF) related diseases by inhibition of natural antisense transcript to HGF
CA2785177C (en) 2009-12-29 2019-09-24 Curna, Inc. Treatment of tumor protein 63 (p63) related diseases by inhibition of natural antisense transcript to p63
CN102782134B (en) 2009-12-29 2017-11-24 库尔纳公司 NRF1 relevant diseases are treated by suppressing the natural antisense transcript of the core breathing factor 1 (NRF1)
WO2011082409A2 (en) 2010-01-04 2011-07-07 Curna, Inc. Treatment of interferon regulatory factor 8 (irf8) related diseases by inhibition of natural antisense transcript to irf8
WO2011085066A2 (en) 2010-01-06 2011-07-14 Curna, Inc. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
EP2524039B1 (en) 2010-01-11 2017-11-29 CuRNA, Inc. Treatment of sex hormone binding globulin (shbg) related diseases by inhibition of natural antisense transcript to shbg
US8779118B2 (en) 2010-01-11 2014-07-15 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
US20120321647A1 (en) 2010-01-12 2012-12-20 Yale University Structured rna motifs and compounds and methods for their use
RU2611192C2 (en) 2010-01-25 2017-02-21 Курна, Инк. TREATMENT OF RNase H1 RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO RNase H1
WO2011097407A1 (en) 2010-02-04 2011-08-11 Ico Therapeutics Inc. Dosing regimens for treating and preventing ocular disorders using c-raf antisense
US8962586B2 (en) 2010-02-22 2015-02-24 Curna, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcript to PYCR1
WO2011105900A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-alpha (c8-alpha) and uses thereof
WO2011105902A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-beta (c8-beta) and uses thereof
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
AR080243A1 (en) 2010-02-23 2012-03-21 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSIS AND TUMOR TREATMENT
WO2011112516A1 (en) 2010-03-08 2011-09-15 Ico Therapeutics Inc. Treating and preventing hepatitis c virus infection using c-raf kinase antisense oligonucleotides
CA2790211C (en) 2010-03-08 2020-06-09 Monsanto Technology Llc Methods and compositions for delivering polynucleotides into plants
US9068185B2 (en) 2010-03-12 2015-06-30 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
US20130101512A1 (en) 2010-03-12 2013-04-25 Chad A. Mirkin Crosslinked polynucleotide structure
EP2544703A4 (en) 2010-03-12 2013-09-18 Brigham & Womens Hospital 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
WO2011120046A2 (en) 2010-03-26 2011-09-29 Swift Biosciences, Inc. Methods and compositions for isolating polynucleotides
AU2011235276B2 (en) 2010-03-29 2015-09-03 Alnylam Pharmaceuticals, Inc. SiRNA therapy for transthyretin (TTR) related ocular amyloidosis
WO2011123621A2 (en) 2010-04-01 2011-10-06 Alnylam Pharmaceuticals Inc. 2' and 5' modified monomers and oligonucleotides
TWI644675B (en) 2010-04-09 2018-12-21 可娜公司 Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
WO2011133695A2 (en) 2010-04-20 2011-10-27 Swift Biosciences, Inc. Materials and methods for nucleic acid fractionation by solid phase entrapment and enzyme-mediated detachment
US9725479B2 (en) 2010-04-22 2017-08-08 Ionis Pharmaceuticals, Inc. 5′-end derivatives
WO2011139699A2 (en) 2010-04-28 2011-11-10 Isis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
US8993738B2 (en) 2010-04-28 2015-03-31 Isis Pharmaceuticals, Inc. Modified nucleosides, analogs thereof and oligomeric compounds prepared therefrom
BR112012027547B1 (en) 2010-04-29 2022-06-14 Ionis Pharmaceuticals, Inc SINGLE STRIP MODIFIED OLIGONUCLEOTIDE, COMPOSITION, AND ITS USES TO TREAT TRANSTHIRRETIN AYLOIDOSIS, REDUCE ITS SYMPTOMS, AND TO REDUCE TRANSTHIRRETIN MRNA OR PROTEIN EXPRESSION
SG185027A1 (en) 2010-05-03 2012-11-29 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
KR101915115B1 (en) 2010-05-03 2018-11-05 큐알엔에이, 인크. Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CA3090304A1 (en) 2010-05-13 2011-11-17 Sarepta Therapeutics, Inc. Antisense modulation of interleukins 17 and 23 signaling
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
US20130203045A1 (en) 2010-05-26 2013-08-08 University Of Virginia Patent Foundation Method for detecting nucleic acids based on aggregate formation
US8895528B2 (en) 2010-05-26 2014-11-25 Curna, Inc. Treatment of atonal homolog 1 (ATOH1) related diseases by inhibition of natural antisense transcript to ATOH1
EP2576579B1 (en) 2010-06-02 2018-08-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
WO2011156278A1 (en) 2010-06-07 2011-12-15 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
WO2011156713A1 (en) 2010-06-11 2011-12-15 Vanderbilt University Multiplexed interferometric detection system and method
US9168297B2 (en) 2010-06-23 2015-10-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (NRG-1)
EP3202415B1 (en) 2010-06-30 2021-12-08 Compugen Ltd. Fusion protein comprising a c1orf32 polypeptide and one or more domains of an immunoglobulin heavy chain constant region for treatment of multiple sclerosis, rheumatoid arthritis and other autoimmune disorders.
US20130143955A1 (en) 2010-08-09 2013-06-06 Yale University Cyclic di-GMP-II Riboswitches, Motifs, and Compounds, and Methods for Their Use
WO2012038956A1 (en) 2010-09-20 2012-03-29 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method of treating neurodegenerative diseases
EP2625197B1 (en) 2010-10-05 2016-06-29 Genentech, Inc. Mutant smoothened and methods of using the same
CN103210086B (en) 2010-10-06 2017-06-09 库尔纳公司 NEU4 relevant diseases are treated by suppressing the natural antisense transcript of sialidase 4 (NEU4)
CN103517990A (en) 2010-10-07 2014-01-15 通用医疗公司 Biomarkers of cancer
WO2012052872A2 (en) 2010-10-17 2012-04-26 Yeda Research And Development Co. Ltd. Methods and compositions for the treatment of insulin-associated medical conditions
EP2630241B1 (en) 2010-10-22 2018-10-17 CuRNA, Inc. Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
JP6073795B2 (en) 2010-10-27 2017-02-01 カッパーアールエヌエー,インコーポレイテッド Treatment of IFRD1-related diseases by inhibition of natural antisense transcripts to interferon-related developmental regulator 1 (IFRD1)
JP2012111933A (en) * 2010-11-02 2012-06-14 Nitto Denko Corp Thermoplastic silicone resin
CN110123830A (en) 2010-11-09 2019-08-16 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting the lipid of the expression of Eg5 and VEGF gene to prepare
EP2638163B1 (en) 2010-11-12 2017-05-17 The General Hospital Corporation Polycomb-associated non-coding rnas
AU2011329777B2 (en) 2010-11-17 2016-06-09 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
KR102010598B1 (en) 2010-11-23 2019-08-13 큐알엔에이, 인크. Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
EP2649182A4 (en) 2010-12-10 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for increasing erythropoietin (epo) production
US9045749B2 (en) 2011-01-14 2015-06-02 The General Hospital Corporation Methods targeting miR-128 for regulating cholesterol/lipid metabolism
EP2670404B1 (en) 2011-02-02 2018-08-29 The Trustees of Princeton University Sirtuin modulators as virus production modulators
KR101697396B1 (en) 2011-02-02 2017-01-17 엑스칼리아드 파마슈티컬즈, 인코포레이티드 Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
US9562853B2 (en) 2011-02-22 2017-02-07 Vanderbilt University Nonaqueous backscattering interferometric methods
MX360349B (en) 2011-03-29 2018-10-30 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene.
WO2012138453A1 (en) 2011-04-03 2012-10-11 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
EP2697256A1 (en) 2011-04-15 2014-02-19 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
WO2012149154A1 (en) 2011-04-26 2012-11-01 Swift Biosciences, Inc. Polynucleotide primers and probes
WO2012151289A2 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system to detect aggregate formation on a substrate
WO2012151268A1 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system for high throughput optical and label free detection of analytes
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
CA2838588C (en) 2011-06-09 2021-09-14 Curna, Inc. Treatment of frataxin (fxn) related diseases by inhibition of natural antisense transcript to fxn
EP2723756B1 (en) 2011-06-21 2020-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP2723351B1 (en) 2011-06-21 2018-02-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
SG10201800715PA (en) 2011-06-21 2018-02-27 Alnylam Pharmaceuticals Inc Angiopoietin-like 3 (angptl3) irna compostions and methods of use thereof
US20140235693A1 (en) 2011-06-23 2014-08-21 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
AU2012277376B2 (en) 2011-06-30 2016-11-24 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
US20140328811A1 (en) 2011-08-01 2014-11-06 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
EP2739736B1 (en) 2011-08-04 2017-04-19 Yeda Research and Development Co. Ltd. Mir-135 and compositions comprising same for the treatment of serotonin associated medical conditions
US10760086B2 (en) 2011-09-13 2020-09-01 Monsanto Technology Llc Methods and compositions for weed control
US9840715B1 (en) 2011-09-13 2017-12-12 Monsanto Technology Llc Methods and compositions for delaying senescence and improving disease tolerance and yield in plants
US10829828B2 (en) 2011-09-13 2020-11-10 Monsanto Technology Llc Methods and compositions for weed control
US10806146B2 (en) 2011-09-13 2020-10-20 Monsanto Technology Llc Methods and compositions for weed control
MX343072B (en) 2011-09-13 2016-10-21 Monsanto Technology Llc Methods and compositions for weed control.
BR112014005975A8 (en) 2011-09-13 2017-09-12 Monsanto Technology Llc PLANT CONTROL METHOD, METHOD OF REDUCING EXPRESSION OF A PDS GENE IN A PLANT, MICROBIAL EXPRESSION CASSETTE, METHOD OF MAKING A POLYNUCLEOTIDE, METHOD OF IDENTIFICATION OF POLYNUCLEOTIDES, AND COMPOSITIONS FOR WEED CONTROL
EP2755467B1 (en) 2011-09-13 2017-07-19 Monsanto Technology LLC Methods and compositions for weed control
MX343071B (en) 2011-09-13 2016-10-21 Monsanto Technology Llc Methods and compositions for weed control.
CA2847698C (en) 2011-09-14 2020-09-01 Northwestern University Nanoconjugates able to cross the blood-brain barrier
US9920326B1 (en) 2011-09-14 2018-03-20 Monsanto Technology Llc Methods and compositions for increasing invertase activity in plants
JP6129844B2 (en) 2011-09-14 2017-05-17 ラナ セラピューティクス インコーポレイテッド Multimeric oligonucleotide compounds
US9580713B2 (en) 2011-09-17 2017-02-28 Yale University Fluoride-responsive riboswitches, fluoride transporters, and methods of use
AU2012322788B2 (en) 2011-10-11 2018-01-04 The Brigham And Women's Hospital, Inc. Micrornas in neurodegenerative disorders
KR102102862B1 (en) 2011-10-14 2020-04-22 제넨테크, 인크. ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
US9145559B2 (en) 2011-10-27 2015-09-29 Yeda Research And Development Co. Ltd. Methods of treating cancer
DK2790736T3 (en) 2011-12-12 2018-05-07 Oncoimmunin Inc In vivo delivery of oligonucleotides
CA2860731C (en) 2012-01-10 2023-02-28 M. Mahmood Hussain Method of treating hyperlipidemia and atherosclerosis with mir-30c
MX2014009289A (en) 2012-02-01 2015-09-08 Compugen Ltd C10rf32 antibodies, and uses thereof for treatment of cancer.
SG11201404608WA (en) 2012-02-13 2014-09-26 Gamida Cell Ltd Mesenchymal stem cells conditioned medium and methods of generating and using the same
EP2844745A2 (en) 2012-02-22 2015-03-11 Brainstem Biotec Ltd. Generation of neural stem cells and motor neurons
EP3401393B1 (en) 2012-02-22 2020-02-19 Exostem Biotec Ltd Micrornas for the generation of astrocytes
WO2013138536A1 (en) 2012-03-13 2013-09-19 Swift Biosciences, Inc. Methods and compositions for size-controlled homopolymer tailing of substrate polynucleotides by a nucleic acid polymerase
EP2639238A1 (en) 2012-03-15 2013-09-18 Universität Bern Tricyclic nucleosides and oligomeric compounds prepared therefrom
KR20140136488A (en) 2012-03-15 2014-11-28 큐알엔에이, 인크. Treatment of brain derived neurotrophic factor(bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US9221864B2 (en) 2012-04-09 2015-12-29 Isis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
AU2013248981B2 (en) 2012-04-20 2018-11-29 Aptamir Therapeutics, Inc. Mirna modulators of thermogenesis
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
US9273949B2 (en) 2012-05-11 2016-03-01 Vanderbilt University Backscattering interferometric methods
SG11201407483YA (en) 2012-05-16 2014-12-30 Rana Therapeutics Inc Compositions and methods for modulating smn gene family expression
KR102028784B1 (en) 2012-05-16 2019-10-04 트랜슬레이트 바이오 인코포레이티드 Compositions and methods for modulating gene expression
AU2013264742B2 (en) 2012-05-24 2018-10-04 A.B. Seeds Ltd. Compositions and methods for silencing gene 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
US20140038182A1 (en) 2012-07-17 2014-02-06 Dna Logix, Inc. Cooperative primers, probes, and applications thereof
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
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
KR102237882B1 (en) 2012-08-15 2021-04-07 아이오니스 파마수티컬즈, 인코포레이티드 Method of preparing oligomeric compounds using modified capping protocols
CA2880869A1 (en) 2012-08-20 2014-02-27 The Regents Of The University Of California Polynucleotides having bioreversible groups
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
CN104870647A (en) 2012-10-18 2015-08-26 孟山都技术公司 Methods and compositions for plant pest control
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
US10683505B2 (en) 2013-01-01 2020-06-16 Monsanto Technology Llc Methods of introducing dsRNA to plant seeds for modulating gene expression
WO2014106837A2 (en) 2013-01-01 2014-07-10 A. B. Seeds Ltd. ISOLATED dsRNA MOLECULES AND METHODS OF USING SAME FOR SILENCING TARGET MOLECULES OF INTEREST
EP2945652B1 (en) 2013-01-18 2021-07-07 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
US10000767B2 (en) 2013-01-28 2018-06-19 Monsanto Technology Llc Methods and compositions for plant pest control
US9701708B2 (en) 2013-01-31 2017-07-11 Ionis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified coupling protocols
WO2014130922A1 (en) 2013-02-25 2014-08-28 Trustees Of Boston University Compositions and methods for treating fungal infections
CA2905027A1 (en) 2013-03-13 2014-10-09 Monsanto Technology Llc Methods and compositions for weed control
AR095233A1 (en) 2013-03-13 2015-09-30 Monsanto Technology Llc METHODS AND COMPOSITIONS FOR WEED CONTROL
DK2970970T3 (en) 2013-03-14 2019-02-11 Andes Biotechnologies Global Inc Antisense oligonucleotides for the treatment of cancer stem cells
WO2014153206A2 (en) 2013-03-14 2014-09-25 Andes Biotechnologies S.A. Methods for detecting and treating multiple myeloma
KR20230162998A (en) 2013-03-14 2023-11-29 알닐람 파마슈티칼스 인코포레이티드 Complement component c5 irna compositions and methods of use thereof
US20140283211A1 (en) 2013-03-14 2014-09-18 Monsanto Technology Llc Methods and Compositions for Plant Pest Control
US10568328B2 (en) 2013-03-15 2020-02-25 Monsanto Technology Llc Methods and compositions for weed control
CN115261411A (en) 2013-04-04 2022-11-01 哈佛学院校长同事会 Therapeutic uses of genome editing with CRISPR/Cas systems
EP3633039A1 (en) 2013-05-01 2020-04-08 Ionis Pharmaceuticals, Inc. Compositions and methods
HUE038146T2 (en) 2013-05-22 2018-09-28 Alnylam Pharmaceuticals Inc Serpina1 irna compositions and methods of use thereof
KR102365809B1 (en) 2013-05-22 2022-02-23 알닐람 파마슈티칼스 인코포레이티드 Tmprss6 compositions and methods of use thereof
US20160113911A1 (en) 2013-06-06 2016-04-28 The General Hospital Corporation Methods and compositions for the treatment of cancer
WO2014197938A1 (en) 2013-06-13 2014-12-18 Antisense Therapeutics Ltd Combination therapy
US9850496B2 (en) 2013-07-19 2017-12-26 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
EP3030663B1 (en) 2013-07-19 2019-09-04 Monsanto Technology LLC Compositions and methods for controlling leptinotarsa
AU2014292926B2 (en) 2013-07-25 2020-03-05 Exicure Operating Company Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
HUE048710T2 (en) 2013-08-08 2020-08-28 Scripps Research Inst A method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
EP3052626A1 (en) 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
PE20161130A1 (en) 2013-10-04 2016-11-25 Icahn School Med Mount Sinai COMPOSITIONS AND METHODS TO INHIBIT THE EXPRESSION OF THE ALAS1 GENE
WO2015054451A1 (en) 2013-10-09 2015-04-16 The United States Of America As Represented By The Secretary Department Of Health And Human Services Detection of hepatitis delta virus (hdv) for the diagnosis and treatment of sjögren's syndrome and lymphoma
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
US9758546B2 (en) 2013-10-21 2017-09-12 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
JP2016536303A (en) 2013-10-21 2016-11-24 ザ ジェネラル ホスピタル コーポレイション Peripheral circulating tumor cell clusters and methods for cancer treatment
US9540642B2 (en) 2013-11-04 2017-01-10 The United States Of America, As Represented By The Secretary Of Agriculture Compositions and methods for controlling arthropod parasite and pest infestations
WO2015066708A1 (en) 2013-11-04 2015-05-07 Northwestern University Quantification and spatio-temporal tracking of a target using a spherical nucleic acid (sna)
WO2015073922A2 (en) 2013-11-15 2015-05-21 Northwestern University Inhibition of oxidative stress in atrial fibrillation
CA2932122C (en) 2013-12-03 2022-04-19 Northwestern University Liposomal particles, methods of making same and uses thereof
US10385388B2 (en) 2013-12-06 2019-08-20 Swift Biosciences, Inc. Cleavable competitor polynucleotides
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
EP3572513A1 (en) 2013-12-09 2019-11-27 Baylor College of Medicine Hippo and dystrophin complex signaling in cardiomyocyte renewal
UA119253C2 (en) 2013-12-10 2019-05-27 Біолоджикс, Інк. Compositions and methods for virus control in varroa mite and bees
AU2014362262B2 (en) 2013-12-12 2021-05-13 Alnylam Pharmaceuticals, Inc. Complement component iRNA compositions and methods of use thereof
CA2934344A1 (en) 2013-12-20 2015-06-25 David T. TING Methods and assays relating to circulating tumor cells
AU2015206585A1 (en) 2014-01-15 2016-07-21 Monsanto Technology Llc Methods and compositions for weed control using EPSPS polynucleotides
CA2937539A1 (en) 2014-02-04 2015-08-13 Genentech, Inc. Mutant smoothened and methods of using the same
US10125365B2 (en) 2014-02-05 2018-11-13 Yeda Research And Development Co. Ltd. Micro-RNAs and compositions comprising same for the treatment and diagnosis of serotonin-, adrenalin-, noradrenalin-, glutamate-, and corticotropin-releasing hormone- associated medical conditions
CA2938857A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2015142910A1 (en) 2014-03-17 2015-09-24 Isis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
CA2942340A1 (en) 2014-03-19 2015-09-24 Ionis Pharmaceuticals, Inc. Compositions for modulating ataxin 2 expression
US10006027B2 (en) 2014-03-19 2018-06-26 Ionis Pharmaceuticals, Inc. Methods for modulating Ataxin 2 expression
EP3420809A1 (en) 2014-04-01 2019-01-02 Monsanto Technology LLC Compositions and methods for controlling insect pests
SG10201906520RA (en) 2014-04-01 2019-09-27 Biogen Ma Inc Compositions for modulating sod-1 expression
WO2015157555A2 (en) 2014-04-09 2015-10-15 The Scripps Research Institute Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
WO2015164693A1 (en) 2014-04-24 2015-10-29 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
PE20170010A1 (en) 2014-05-01 2017-03-04 Ionis Pharmaceuticals Inc COMPOSITIONS AND METHODS TO MODULATE THE EXPRESSION OF THE COMPLEMENT FACTOR B
US10098959B2 (en) 2014-05-01 2018-10-16 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
WO2015175510A1 (en) 2014-05-12 2015-11-19 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
JP6811094B2 (en) 2014-05-22 2021-01-13 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Angiotensinogen (AGT) iRNA composition and its use
US10456445B2 (en) 2014-06-02 2019-10-29 Children's Medical Center Corporation Methods and compositions for immunomodulation
TR201908550T4 (en) 2014-06-04 2019-07-22 Exicure Inc Multivalent delivery of immune modulators by liposomal spherical nucleic acids for prophylactic or therapeutic applications.
CN106661580B (en) 2014-06-10 2022-02-15 鹿特丹伊拉斯谟大学医疗中心 Antisense oligonucleotides for treating pompe disease
WO2015200223A1 (en) 2014-06-23 2015-12-30 Monsanto Technology Llc Compositions and methods for regulating gene expression via rna interference
EP3161138A4 (en) 2014-06-25 2017-12-06 Monsanto Technology LLC Methods and compositions for delivering nucleic acids to plant cells and regulating gene expression
WO2015200697A1 (en) 2014-06-25 2015-12-30 The General Hospital Corporation Targeting human satellite ii (hsatii)
BR112017000710B1 (en) 2014-07-15 2024-02-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem ISOLATED POLYPEPTIDE, COMPOSITION OF MATTER, AND, USE OF THE ISOLATED POLYPEPTIDE OR COMPOSITION OF MATTER
US9951327B1 (en) 2014-07-17 2018-04-24 Integrated Dna Technologies, Inc. Efficient and rapid method for assembling and cloning double-stranded DNA fragments
WO2016018887A1 (en) 2014-07-29 2016-02-04 Monsanto Technology Llc Compositions and methods for controlling insect pests
MX2017001432A (en) 2014-07-31 2017-05-09 Uab Res Found Apoe mimetic peptides and higher potency to clear plasma cholesterol.
EP3183007B1 (en) 2014-08-19 2020-06-17 Northwestern University Protein/oligonucleotide core-shell nanoparticle therapeutics
WO2016030899A1 (en) 2014-08-28 2016-03-03 Yeda Research And Development Co. Ltd. Methods of treating amyotrophic lateral scleroses
EP3185957B1 (en) 2014-08-29 2022-06-01 Alnylam Pharmaceuticals, Inc. Patisiran for use in treating transthyretin mediated amyloidosis
JP2017526367A (en) 2014-08-29 2017-09-14 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 the treatment of cancer
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
WO2016040589A1 (en) 2014-09-12 2016-03-17 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
EP3198012B1 (en) 2014-09-26 2019-09-04 University of Massachusetts Rna-modulating agents
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CN113846101A (en) 2014-11-17 2021-12-28 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
CN107532162A (en) 2014-12-12 2018-01-02 托德·M·伍尔夫 For the composition and method using oligonucleotides editor's cell amplifying nucleic acid
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
US10793855B2 (en) 2015-01-06 2020-10-06 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
US10538763B2 (en) 2015-01-16 2020-01-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of DUX4
US10968449B2 (en) 2015-01-22 2021-04-06 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
WO2016118812A1 (en) 2015-01-23 2016-07-28 Vanderbilt University A robust interferometer and methods of using same
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
WO2016130806A2 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
US11421229B2 (en) 2015-02-20 2022-08-23 Baylor College Of Medicine p63 inactivation for the treatment of heart failure
US10450342B2 (en) 2015-02-23 2019-10-22 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
US20180200387A1 (en) 2015-02-23 2018-07-19 Crispr Therapeutics Ag Materials and methods for treatment of human genetic diseases including hemoglobinopathies
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
WO2016142948A1 (en) 2015-03-11 2016-09-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Decoy oligonucleotides for the treatment of diseases
CA2981068C (en) 2015-03-26 2021-12-14 Women & Infants Hospital Of Rhode Island Therapy for malignant disease comprising the inhibition of human epididymal secretory protein e4 and immune checkpoint inhibitors
WO2016157175A1 (en) 2015-03-27 2016-10-06 Yeda Research And Development Co. Ltd. Methods of treating motor neuron diseases
BR112017020750A2 (en) 2015-03-27 2018-06-26 Harvard College modified t-cells and methods of producing and using them
WO2016164463A1 (en) 2015-04-07 2016-10-13 The General Hospital Corporation Methods for reactivating genes on the inactive x chromosome
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
BR112017023754B1 (en) 2015-05-04 2022-08-09 Beeologics, Inc. INSECTICIDE COMPOSITIONS, AND USE OF A NUCLEIC ACID
AU2016257150B2 (en) 2015-05-06 2022-03-31 Benitec IP Holdings Inc. Reagents for treatment of hepatitis B virus (HBV) infection and use thereof
EP3294280A1 (en) 2015-05-11 2018-03-21 Yeda Research and Development Co., Ltd. Citrin inhibitors for the treatment of cancer
UY36703A (en) 2015-06-02 2016-12-30 Monsanto Technology Llc COMPOSITIONS AND METHODS FOR THE ADMINISTRATION OF A POLINUCLEOTIDE ON A PLANT
EP3302030A4 (en) 2015-06-03 2019-04-24 Monsanto Technology LLC Methods and compositions for introducing nucleic acids into plants
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
EP3311165B1 (en) 2015-06-19 2020-12-09 University Of Rochester Septin proteins as novel biomarkers for detection and treatment of müllerian cancers
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
CA2990852A1 (en) 2015-06-26 2016-12-29 Beth Israel Deaconess Medical Center, Inc. Cancer therapy targeting tetraspanin 33 (tspan33) in myeloid derived suppressor cells
CA2991045A1 (en) 2015-06-29 2017-01-05 Caris Science, Inc. Therapeutic oligonucleotides binding c1q
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
WO2017019918A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
WO2017021963A1 (en) 2015-08-03 2017-02-09 Biokine Therapeutics Ltd. Cxcr4 binding agents for treatment of diseases
WO2017021961A1 (en) 2015-08-04 2017-02-09 Yeda Research And Development Co. Ltd. Methods of screening for riboswitches and attenuators
SG10202007937SA (en) 2015-09-02 2020-09-29 Alnylam Pharmaceuticals Inc PROGRAMMED CELL DEATH 1 LIGAND 1 (PD-L1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3353297A1 (en) 2015-09-24 2018-08-01 Crispr Therapeutics AG Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
EP3353303B1 (en) 2015-09-25 2023-08-02 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
AU2016344609B2 (en) 2015-10-28 2022-05-12 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of duchenne muscular dystrophy
IL295097A (en) 2015-10-30 2022-09-01 Genentech Inc Anti-htra1 antibodies and methods of use thereof
AU2016349954B2 (en) 2015-11-05 2022-08-25 Antisense Therapeutics Ltd Mobilizing leukemia cells
MX2018005332A (en) 2015-11-06 2018-11-09 Crispr Therapeutics Ag Materials and methods for treatment of glycogen storage disease type 1a.
WO2017081686A1 (en) 2015-11-10 2017-05-18 B. G. Negev Technologies And Applications Ltd., At Ben-Gurion University Means and methods for reducing tumorigenicity of cancer stem cells
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
US20170145394A1 (en) 2015-11-23 2017-05-25 The Regents Of The University Of California Tracking and manipulating cellular rna via nuclear delivery of crispr/cas9
WO2017093804A2 (en) 2015-12-01 2017-06-08 Crispr Therapeutics Ag Materials and methods for treatment of alpha-1 antitrypsin deficiency
EP3389670A4 (en) 2015-12-04 2020-01-08 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
WO2017106767A1 (en) 2015-12-18 2017-06-22 The Scripps Research Institute Production of unnatural nucleotides using a crispr/cas9 system
BR112018012894A2 (en) 2015-12-23 2018-12-04 Crispr Therapeutics Ag Materials and Methods for Treatment of Amyotrophic Lateral Sclerosis and / or Frontotemporal Lobular Degeneration
CA3006599A1 (en) 2016-01-05 2017-07-13 Ionis Pharmaceuticals, Inc. Methods for reducing lrrk2 expression
US10627396B2 (en) 2016-01-29 2020-04-21 Vanderbilt University Free-solution response function interferometry
US20190038771A1 (en) 2016-02-02 2019-02-07 Crispr Therapeutics Ag Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
JP2019509721A (en) 2016-02-04 2019-04-11 キュリス,インコーポレイテッド Mutant smoothened and method of using the same
EP3416689B1 (en) 2016-02-18 2023-01-18 CRISPR Therapeutics AG Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
WO2017147087A1 (en) 2016-02-25 2017-08-31 The Brigham And Women's Hospital, Inc. Treatment methods for fibrosis targeting smoc2
WO2017152182A1 (en) 2016-03-04 2017-09-08 Rhode Island Hospital Targeting microrna for cancer treatment
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
EP3429632B1 (en) 2016-03-16 2023-01-04 CRISPR Therapeutics AG Materials and methods for treatment of hereditary haemochromatosis
CA3013799A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Methods of modulating keap1
CN109715802A (en) 2016-03-18 2019-05-03 卡里斯科学公司 Oligonucleotide probe and application thereof
AU2017250017B2 (en) 2016-04-14 2022-12-22 Benitec IP Holdings Inc. Reagents for treatment of oculopharyngeal muscular dystrophy (OPMD) and use thereof
SG10202010261YA (en) 2016-04-18 2020-11-27 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
WO2017191503A1 (en) 2016-05-05 2017-11-09 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
JP2019518028A (en) 2016-06-10 2019-06-27 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Complement component C5i RNA composition and its use for treating paroxysmal nocturnal hemoglobinuria (PNH)
US11236339B2 (en) 2016-06-17 2022-02-01 Ionis Pharmaceuticals, Inc. Modulation of GYS1 expression
WO2017223528A1 (en) 2016-06-24 2017-12-28 The Scripps Research Institute Novel nucleoside triphosphate transporter and uses thereof
US11427838B2 (en) 2016-06-29 2022-08-30 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of myotonic dystrophy type 1 (DM1) and other related disorders
WO2018002762A1 (en) 2016-06-29 2018-01-04 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis (als) and other related disorders
US11564997B2 (en) 2016-06-29 2023-01-31 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
US11801313B2 (en) 2016-07-06 2023-10-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of pain related disorders
CA3029141A1 (en) 2016-07-06 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of pain related disorders
WO2018007871A1 (en) 2016-07-08 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of transthyretin amyloidosis
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
US20190247436A1 (en) 2016-07-21 2019-08-15 Maxcyte, Inc. Methods and compositions for modifying genomic dna
WO2018020323A2 (en) 2016-07-25 2018-02-01 Crispr Therapeutics Ag Materials and methods for treatment of fatty acid disorders
NL2017295B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Antisense oligomeric compound for Pompe disease
NL2017294B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Natural cryptic exon removal by pairs of antisense oligonucleotides.
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
HUE059718T2 (en) 2016-09-02 2022-12-28 Dicerna Pharmaceuticals Inc 4'-phosphate analogs and oligonucleotides comprising the same
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
TW202313978A (en) 2016-11-23 2023-04-01 美商阿尼拉製藥公司 Serpina1 irna compositions and methods of use thereof
EP3330276A1 (en) 2016-11-30 2018-06-06 Universität Bern Novel bicyclic nucleosides and oligomers prepared therefrom
US11033570B2 (en) 2016-12-02 2021-06-15 Cold Spring Harbor Laboratory Modulation of Lnc05 expression
WO2018111834A1 (en) 2016-12-13 2018-06-21 Seattle Children's Hospital (dba Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
EP3555292A1 (en) 2016-12-16 2019-10-23 Alnylam Pharmaceuticals, Inc. Methods for treating or preventing ttr-associated diseases using transthyretin (ttr) irna compositions
IL301053A (en) 2017-01-23 2023-05-01 Regeneron Pharma Hsd17b13 variants and uses thereof
WO2018154439A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 1 (sca1) and other spinocerebellar ataxia type 1 protein (atxn1) gene related conditions or disorders
EP3585899A1 (en) 2017-02-22 2020-01-01 CRISPR Therapeutics AG Materials and methods for treatment of primary hyperoxaluria type 1 (ph1) and other alanine-glyoxylate aminotransferase (agxt) gene related conditions or disorders
WO2018154418A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
CA3054031A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Compositions and methods for gene editing
US20200216857A1 (en) 2017-02-22 2020-07-09 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
WO2018165564A1 (en) 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
WO2018183969A1 (en) 2017-03-30 2018-10-04 California Institute Of Technology Barcoded rapid assay platform for efficient analysis of candidate molecules and methods of making and using the platform
TWI801377B (en) 2017-04-18 2023-05-11 美商阿尼拉製藥公司 Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
US11872239B2 (en) 2017-04-20 2024-01-16 Synthena Ag Modified oligomeric compounds comprising tricyclo-DNA nucleosides and uses thereof
WO2018193428A1 (en) 2017-04-20 2018-10-25 Synthena Ag Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
AU2018256435A1 (en) 2017-04-20 2019-11-07 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
WO2018195486A1 (en) 2017-04-21 2018-10-25 The Broad Institute, Inc. Targeted delivery to beta cells
JP7398279B2 (en) 2017-05-10 2023-12-14 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Targeted editing of cellular RNA by CRISPR/CAS9 nuclear delivery
EP3621981A2 (en) 2017-05-12 2020-03-18 CRISPR Therapeutics AG Materials and methods for engineering cells and uses thereof in immuno-oncology
SG11202000167SA (en) 2017-07-11 2020-02-27 Synthorx Inc Incorporation of unnatural nucleotides and methods thereof
CA3069909A1 (en) 2017-07-13 2019-02-14 Northwestern University General and direct method for preparing oligonucleotide-functionalized metal-organic framework nanoparticles
AU2018301477A1 (en) 2017-07-13 2020-02-27 Alnylam Pharmaceuticals Inc. Lactate dehydrogenase a (LDHA) iRNA compositions and methods of use thereof
CA3071016A1 (en) 2017-08-03 2019-02-07 Synthorx, Inc. Cytokine conjugates for the treatment of autoimmune diseases
WO2019036613A1 (en) 2017-08-18 2019-02-21 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
KR20200089656A (en) 2017-09-19 2020-07-27 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for treating transthyretin (TTR) mediated amyloidosis
CA3079172A1 (en) 2017-10-17 2019-04-25 Crispr Therapeutics Ag Compositions and methods for gene editing for hemophilia a
EP3701029A1 (en) 2017-10-26 2020-09-02 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
US11866701B2 (en) 2017-11-01 2024-01-09 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
MA50578A (en) 2017-11-09 2021-09-15 Vertex Pharma CRISPR / CAS SYSTEMS FOR THE TREATMENT OF DMD
EP3710587A1 (en) 2017-11-16 2020-09-23 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
EP3714054A1 (en) 2017-11-20 2020-09-30 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
US10662425B2 (en) 2017-11-21 2020-05-26 Crispr Therapeutics Ag Materials and methods for treatment of autosomal dominant retinitis pigmentosa
US20200384033A1 (en) 2017-12-05 2020-12-10 Vertex Pharmaceuticals Incorporated Crispr-cas9 modified cd34+ human hematopoietic stem and progenitor cells and uses thereof
CN111801417A (en) 2017-12-14 2020-10-20 克里斯珀医疗股份公司 Novel RNA-programmable endonuclease systems and their use in genome editing and other applications
KR20200110655A (en) 2017-12-18 2020-09-24 알닐람 파마슈티칼스 인코포레이티드 High mobility group box-1 (HMGB1) IRNA composition and method of use thereof
EP3728595A1 (en) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Materials and methods for treatment of usher syndrome type 2a and/or non-syndromic autosomal recessive retinitis pigmentosa (arrp)
CA3084632A1 (en) 2017-12-21 2019-06-27 Crispr Therapeutics Ag Materials and methods for treatment of usher syndrome type 2a
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
CA3088180A1 (en) 2018-01-12 2019-07-18 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting transferrin
MA51645A (en) 2018-01-15 2020-11-25 Ionis Pharmaceuticals Inc DNM2 EXPRESSION MODULATORS
WO2019142135A1 (en) 2018-01-19 2019-07-25 Synthena Ag Tricyclo-dna nucleoside precursors and processes for preparing the same
US20190233816A1 (en) 2018-01-26 2019-08-01 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
MA51788A (en) 2018-02-05 2020-12-16 Vertex Pharma SUBSTANCES AND METHODS FOR TREATING HEMOGLOBINOPATHIES
US11566236B2 (en) 2018-02-05 2023-01-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2019155465A1 (en) 2018-02-08 2019-08-15 Yeda Research And Development Co. Ltd. Methods of identifying and using agents for treating diseases associated with intestinal barrier dysfunction
US20210130824A1 (en) 2018-02-16 2021-05-06 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting fibrinogen-alpha
AR114445A1 (en) 2018-02-26 2020-09-09 Synthorx Inc IL-15 CONJUGATES, AND THEIR USES
US11732260B2 (en) 2018-03-02 2023-08-22 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-β precursor protein
WO2019169219A1 (en) 2018-03-02 2019-09-06 Ionis Pharmaceuticals, Inc. Modulators of irf4 expression
EP3762395A1 (en) 2018-03-07 2021-01-13 Sanofi Nucleotide precursors, nucleotide analogs and oligomeric compounds containing the same
WO2019183150A1 (en) 2018-03-19 2019-09-26 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and uses thereof
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
EP3772928A4 (en) 2018-04-06 2021-12-29 Children's Medical Center Corporation Compositions and methods for somatic cell reprogramming and modulating imprinting
US11365416B2 (en) 2018-04-11 2022-06-21 Ionis Pharmaceuticals, Inc. Modulators of EZH2 expression
WO2019204668A1 (en) 2018-04-18 2019-10-24 Casebia Therapeutics Limited Liability Partnership Compositions and methods for knockdown of apo(a) by gene editing for treatment of cardiovascular disease
BR112020020957B1 (en) 2018-05-09 2022-05-10 Ionis Pharmaceuticals, Inc Oligomeric compounds, population and pharmaceutical composition thereof and their uses
PE20212131A1 (en) 2018-05-09 2021-11-05 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN3
TW202016304A (en) 2018-05-14 2020-05-01 美商阿尼拉製藥公司 Angiotensinogen (agt) irna compositions and methods of use thereof
JP2021526823A (en) 2018-06-14 2021-10-11 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. Compounds and methods for increasing STMN2 expression
AU2019294838A1 (en) 2018-06-27 2021-01-07 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing LRRK2 expression
BR112020025824A2 (en) 2018-06-28 2021-03-23 Crispr Therapeutics Ag compositions and methods for genome editing by inserting donor polynucleotides
US11078486B2 (en) 2018-07-25 2021-08-03 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ATXN2 expression
AR114551A1 (en) 2018-08-13 2020-09-16 Alnylam Pharmaceuticals Inc COMPOSITIONS OF hdRNA AGENTS AGAINST HEPATITIS B VIRUS (HBV) AND METHODS FOR THEIR USE
US20210348162A1 (en) 2018-08-16 2021-11-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US20210292766A1 (en) 2018-08-29 2021-09-23 University Of Massachusetts Inhibition of Protein Kinases to Treat Friedreich Ataxia
EP3849584A4 (en) 2018-09-14 2022-06-22 Northwestern University Programming protein polymerization with dna
AU2019344776A1 (en) 2018-09-18 2021-01-21 Alnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA compositions and methods of use thereof
WO2020081843A1 (en) 2018-10-17 2020-04-23 Casebia Therapeutics Limited Liability Partnership Compositions and methods for delivering transgenes
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
MX2021005758A (en) 2018-11-15 2021-09-21 Ionis Pharmaceuticals Inc Modulators of irf5 expression.
IL263184A (en) 2018-11-21 2020-05-31 Yarden Yosef Method of treating cancer and compositions for same
US20210332495A1 (en) 2018-12-06 2021-10-28 Northwestern University Protein Crystal Engineering Through DNA Hybridization Interactions
CA3123617A1 (en) 2018-12-20 2020-06-25 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of kcnt1 related disorders
EA202191736A1 (en) 2018-12-20 2021-10-01 Вир Байотекнолоджи, Инк. HBV COMBINATION THERAPY
CN113557023A (en) 2019-01-16 2021-10-26 建新公司 Serpinc1 iRNA compositions and methods of use thereof
US11214803B2 (en) 2019-01-31 2022-01-04 Ionis Pharmaceuticals, Inc. Modulators of YAP1 expression
MA54952A (en) 2019-02-06 2022-05-11 Synthorx Inc IL-2 CONJUGATES AND METHODS OF USING THE SAME
MA54951A (en) 2019-02-15 2021-12-22 Bayer Healthcare Llc GENE EDITING FOR HEMOPHILIA A WITH ENHANCED FACTOR VIII EXPRESSION
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
WO2020170240A1 (en) 2019-02-21 2020-08-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reduction drug-induced nephrotoxicity
WO2020176771A1 (en) 2019-02-27 2020-09-03 Ionis Pharmaceuticals, Inc. Modulators of malat1 expression
SG11202109741VA (en) 2019-03-12 2021-10-28 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
BR112021018739A2 (en) 2019-03-29 2022-05-03 Dicerna Pharmaceuticals Inc Compositions and methods for treating kras-associated diseases or disorders
CN113728104B (en) 2019-03-29 2023-10-27 Ionis制药公司 Compounds and methods for modulating UBE3A-ATS
BR112021021686A2 (en) 2019-05-03 2022-03-22 Dicerna Pharmaceuticals Inc Double-stranded nucleic acid inhibitor molecules with short sense strands
EP3966327A1 (en) 2019-05-08 2022-03-16 Vertex Pharmaceuticals Incorporated Crispr/cas all-in-two vector systems for treatment of dmd
CN114126628A (en) 2019-05-13 2022-03-01 维尔生物科技有限公司 Compositions and methods for treating Hepatitis B Virus (HBV) infection
EP3983543A4 (en) 2019-06-14 2023-05-03 The Scripps Research Institute Reagents and methods for replication, transcription, and translation in semi-synthetic organisms
WO2020254872A2 (en) 2019-06-17 2020-12-24 Crispr Therapeutics Ag Methods and compositions for improved homology directed repair
WO2021021673A1 (en) 2019-07-26 2021-02-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
WO2021022109A1 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. SERPIN FAMILY F MEMBER 2 (SERPINF2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4013870A1 (en) 2019-08-13 2022-06-22 Alnylam Pharmaceuticals, Inc. Small ribosomal protein subunit 25 (rps25) irna agent compositions and methods of use thereof
WO2021030778A1 (en) 2019-08-15 2021-02-18 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
MX2022001776A (en) 2019-08-15 2022-03-17 Synthorx Inc Immuno oncology combination therapies with il-2 conjugates.
AU2020337869A1 (en) 2019-08-23 2022-03-03 Synthorx, Inc. IL-15 conjugates and uses thereof
US20220290152A1 (en) 2019-09-03 2022-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
CA3150082A1 (en) 2019-09-10 2021-03-18 Jerod PTACIN Il-2 conjugates and methods of use to treat autoimmune diseases
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021076828A1 (en) 2019-10-18 2021-04-22 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
WO2021081026A1 (en) 2019-10-22 2021-04-29 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
US20230040920A1 (en) 2019-11-01 2023-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
JP2023500661A (en) 2019-11-01 2023-01-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
JP2022554272A (en) 2019-11-04 2022-12-28 シンソークス, インコーポレイテッド Interleukin 10 conjugates and uses thereof
MX2022005692A (en) 2019-11-13 2022-06-08 Alnylam Pharmaceuticals Inc Methods and compositions for treating an angiotensinogen- (agt-) associated disorder.
US20230056569A1 (en) 2019-11-22 2023-02-23 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
CA3159501A1 (en) 2019-11-27 2021-06-03 Brian Joseph CAFFERTY Methods of synthesizing rna molecules
IL293824A (en) 2019-12-13 2022-08-01 Alnylam Pharmaceuticals Inc Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
TW202138559A (en) 2019-12-16 2021-10-16 美商阿尼拉製藥公司 Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2021122944A1 (en) 2019-12-18 2021-06-24 Alia Therapeutics Srl Compositions and methods for treating retinitis pigmentosa
AU2021207504A1 (en) 2020-01-15 2022-08-04 Dicerna Pharmaceuticals, Inc. 4'-O-methylene phosphonate nucleic acids and analogues thereof
US20230057461A1 (en) 2020-01-27 2023-02-23 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Rab13 and net1 antisense oligonucleotides to treat metastatic cancer
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
IL295445A (en) 2020-02-10 2022-10-01 Alnylam Pharmaceuticals Inc Compositions and methods for silencing vegf-a expression
WO2021167841A1 (en) 2020-02-18 2021-08-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
KR20220148230A (en) 2020-02-28 2022-11-04 아이오니스 파마수티컬즈, 인코포레이티드 Compounds and methods for modulating SMN2
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
CA3174725A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
JP2023519274A (en) 2020-03-26 2023-05-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド CORONAVIRUS iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US20230190785A1 (en) 2020-03-30 2023-06-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajc15 gene expression
CA3179411A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
MX2022012561A (en) 2020-04-07 2022-11-07 Alnylam Pharmaceuticals Inc Compositions and methods for silencing scn9a expression.
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
MX2022013525A (en) 2020-04-27 2023-01-24 Alnylam Pharmaceuticals Inc Apolipoprotein e (apoe) irna agent compositions and methods of use thereof.
BR112022021136A2 (en) 2020-04-30 2022-11-29 Alnylam Pharmaceuticals Inc COMPLEMENT FACTOR B IRNA COMPOSITIONS (CFB) AND METHODS OF USE THEREOF
JP2023524065A (en) 2020-05-01 2023-06-08 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 modulating ATXN1
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
CA3162416C (en) 2020-05-15 2023-07-04 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
WO2021231698A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
EP4150087A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
WO2021231685A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150076A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
EP4153746A1 (en) 2020-05-21 2023-03-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
US20230203484A1 (en) 2020-05-22 2023-06-29 Wave Life Sciences Ltd. Double stranded oligonucleotide compositions and methods relating thereto
US11408000B2 (en) 2020-06-03 2022-08-09 Triplet Therapeutics, Inc. Oligonucleotides for the treatment of nucleotide repeat expansion disorders associated with MSH3 activity
EP4161552A1 (en) 2020-06-05 2023-04-12 The Broad Institute, Inc. Compositions and methods for treating neoplasia
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
IL299063A (en) 2020-06-18 2023-02-01 Alnylam Pharmaceuticals Inc XANTHINE DEHYDROGENASE (XDH) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021262840A1 (en) 2020-06-24 2021-12-30 Vir Biotechnology, Inc. Engineered hepatitis b virus neutralizing antibodies and uses thereof
EP4171648A1 (en) 2020-06-25 2023-05-03 Synthorx, Inc. Immuno oncology combination therapy with il-2 conjugates and anti-egfr antibodies
MX2022016338A (en) 2020-06-29 2023-01-24 Ionis Pharmaceuticals Inc Compounds and methods for modulating plp1.
EP4192505A1 (en) 2020-08-04 2023-06-14 Dicerna Pharmaceuticals, Inc. Systemic delivery of oligonucleotides
WO2022066847A1 (en) 2020-09-24 2022-03-31 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
WO2022070107A1 (en) 2020-09-30 2022-04-07 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis
EP3978608A1 (en) 2020-10-05 2022-04-06 SQY Therapeutics Oligomeric compound for dystrophin rescue in dmd patients throughout skipping of exon-51
EP4225917A1 (en) 2020-10-05 2023-08-16 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
KR20230084203A (en) 2020-10-09 2023-06-12 신톡스, 인크. Immuno-oncology Combination Therapy Using IL-2 Conjugates and Pembrolizumab
WO2022076859A1 (en) 2020-10-09 2022-04-14 Synthorx, Inc. Immuno oncology therapies with il-2 conjugates
WO2022079719A1 (en) 2020-10-15 2022-04-21 Yeda Research And Development Co. Ltd. Method of treating myeloid malignancies
WO2022084331A2 (en) 2020-10-20 2022-04-28 Sanofi Novel ligands for asialoglycoprotein receptor
CA3198823A1 (en) 2020-10-21 2022-04-28 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
WO2022103999A1 (en) 2020-11-13 2022-05-19 Alnylam Pharmaceuticals, Inc. COAGULATION FACTOR V (F5) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
JP2023549563A (en) 2020-11-18 2023-11-27 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 modulating angiotensinogen expression
EP4256053A1 (en) 2020-12-01 2023-10-11 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
JP2024501288A (en) 2020-12-23 2024-01-11 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー Compositions of modified TREM and uses thereof
EP4274896A1 (en) 2021-01-05 2023-11-15 Alnylam Pharmaceuticals, Inc. Complement component 9 (c9) irna compositions and methods of use thereof
WO2022174101A1 (en) 2021-02-12 2022-08-18 Synthorx, Inc. Skin cancer combination therapy with il-2 conjugates and cemiplimab
KR20230146048A (en) 2021-02-12 2023-10-18 알닐람 파마슈티칼스 인코포레이티드 Superoxide dismutase 1 (SOD1) IRNA compositions and methods of using them to treat or prevent superoxide dismutase 1- (SOD1-)-related neurodegenerative diseases
TW202302148A (en) 2021-02-12 2023-01-16 美商欣爍克斯公司 Lung cancer combination therapy with il-2 conjugates and an anti-pd-1 antibody or antigen-binding fragment thereof
EP4298220A1 (en) 2021-02-25 2024-01-03 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods of use thereof
BR112023016645A2 (en) 2021-02-26 2023-11-14 Alnylam Pharmaceuticals Inc KETOHEXOKINASE (KHK) IRNA COMPOSITIONS AND METHODS OF USE THEREOF
BR112023017737A2 (en) 2021-03-04 2023-10-03 Alnylam Pharmaceuticals Inc ANGIOPOIETIN-SIMILAR IRNA COMPOSITIONS 3 (ANGPTL3) AND METHODS OF USE THEREOF
WO2022192038A1 (en) 2021-03-12 2022-09-15 Northwestern University Antiviral vaccines using spherical nucleic acids
EP4305169A1 (en) 2021-03-12 2024-01-17 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
IL307239A (en) 2021-03-29 2023-11-01 Alnylam Pharmaceuticals Inc Huntingtin (htt) irna agent compositions and methods of use thereof
EP4314293A1 (en) 2021-04-01 2024-02-07 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
IL307926A (en) 2021-04-26 2023-12-01 Alnylam Pharmaceuticals Inc Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
EP4330396A1 (en) 2021-04-29 2024-03-06 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022235537A1 (en) 2021-05-03 2022-11-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (ttr) mediated amyloidosis
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
EP4341405A1 (en) 2021-05-20 2024-03-27 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
WO2022256395A1 (en) 2021-06-02 2022-12-08 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
TW202313117A (en) 2021-06-03 2023-04-01 美商欣爍克斯公司 Head and neck cancer combination therapy comprising an il-2 conjugate and cetuximab
KR20240017911A (en) 2021-06-04 2024-02-08 알닐람 파마슈티칼스 인코포레이티드 Human chromosome 9 open reading frame 72 (C9orf72) iRNA preparation composition and method of using the same
AR126070A1 (en) 2021-06-08 2023-09-06 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR TREATING OR PREVENTING STARGARDT DISEASE AND/OR DISORDERS ASSOCIATED WITH RETINOL BORDER PROTEIN 4 (RBP4)
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
KR20240021218A (en) 2021-06-11 2024-02-16 바이엘 악티엔게젤샤프트 Novel type V RNA programmable endonuclease system
BR112023026050A2 (en) 2021-06-18 2024-03-05 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE IFNAR1 EXPRESSION
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
KR20240026203A (en) 2021-06-30 2024-02-27 알닐람 파마슈티칼스 인코포레이티드 Methods and compositions for treating angiotensinogen (AGT)-related disorders
WO2023285431A1 (en) 2021-07-12 2023-01-19 Alia Therapeutics Srl Compositions and methods for allele specific treatment of retinitis pigmentosa
TW202333748A (en) 2021-07-19 2023-09-01 美商艾拉倫製藥股份有限公司 Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
WO2023003995A1 (en) 2021-07-23 2023-01-26 Alnylam Pharmaceuticals, Inc. Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
AU2022323090A1 (en) 2021-08-03 2024-02-01 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
TW202337474A (en) 2021-08-04 2023-10-01 美商艾拉倫製藥股份有限公司 Irna compositions and methods for silencing angiotensinogen (agt)
CA3228733A1 (en) 2021-08-13 2023-02-16 Alnylam Pharmaceuticals, Inc. Factor xii (f12) irna compositions and methods of use thereof
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
CA3232420A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
CA3234835A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
AU2022378567A1 (en) 2021-10-29 2024-04-11 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2023076450A2 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023118349A1 (en) 2021-12-21 2023-06-29 Alia Therapeutics Srl Type ii cas proteins and applications thereof
WO2023122750A1 (en) 2021-12-23 2023-06-29 Synthorx, Inc. Cancer combination therapy with il-2 conjugates and cetuximab
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023177866A1 (en) 2022-03-18 2023-09-21 Dicerna Pharmaceuticals, Inc. Decarboxylative acetoxylation using mn(ii) or mn(iii) reagent for synthesis of 4'-acetoxy- nucleoside and use thereof for synthesis of corresponding 4'-(dimethoxyphosphoryl)methoxy- nucleotide
WO2023194359A1 (en) 2022-04-04 2023-10-12 Alia Therapeutics Srl Compositions and methods for treatment of usher syndrome type 2a
WO2024039776A2 (en) 2022-08-18 2024-02-22 Alnylam Pharmaceuticals, Inc. Universal non-targeting sirna compositions and methods of use thereof
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
WO2024056880A2 (en) 2022-09-16 2024-03-21 Alia Therapeutics Srl Enqp type ii cas proteins and applications thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5927900A (en) * 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
US4904582A (en) * 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
EP0348458B1 (en) * 1987-11-30 1997-04-09 University Of Iowa Research Foundation Dna 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
AU5192190A (en) * 1989-02-24 1990-09-26 City Of Hope Heterologous block oligomers
DE69027443T2 (en) * 1989-10-24 1998-03-19 Gilead Sciences Inc OLIGONUCLEOTID ANALOGUE WITH NEW BINDINGS
US5138045A (en) * 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5489677A (en) * 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5223618A (en) * 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5378825A (en) * 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5245022A (en) * 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
PT98562B (en) * 1990-08-03 1999-01-29 Sanofi Sa PROCESS FOR THE PREPARATION OF COMPOSITIONS THAT UNDERSEAD SEEDS OF NUCLEO-SIDS WITH NEAR 6 TO NEAR 200 NUCLEASE-RESISTANT BASES
US5214134A (en) * 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5389023A (en) * 1993-03-22 1995-02-14 Mcintyre; Jonothon M. W. Body surfing board

Also Published As

Publication number Publication date
TW222641B (en) 1994-04-21
EP0541722B1 (en) 1995-12-20
HUT67834A (en) 1995-05-29
US5677439A (en) 1997-10-14
ES2083593T3 (en) 1996-04-16
NZ239247A (en) 1993-11-25
HU9300282D0 (en) 1993-04-28
KR100211552B1 (en) 1999-08-02
PT98562B (en) 1999-01-29
HU217036B (en) 1999-11-29
AU8521791A (en) 1992-03-02
GR3018881T3 (en) 1996-05-31
PT98562A (en) 1992-06-30
HU211668A9 (en) 1995-12-28
FI930455A (en) 1993-03-24
DE69115702T2 (en) 1996-06-13
AU692532B2 (en) 1998-06-11
BR9106729A (en) 1993-07-20
DK0541722T3 (en) 1996-04-22
ATE131827T1 (en) 1996-01-15
EP0541722A1 (en) 1993-05-19
FI930455A0 (en) 1993-02-02
JPH06502300A (en) 1994-03-17
IL99066A (en) 1996-01-31
WO1992002534A2 (en) 1992-02-20
WO1992002534A3 (en) 1992-06-11
IL99066A0 (en) 1992-07-15
AU2008195A (en) 1995-10-12
AU667459B2 (en) 1996-03-28
IL113519A (en) 1997-11-20
DE69115702D1 (en) 1996-02-01
MY107332A (en) 1995-11-30
KR930702372A (en) 1993-09-08
IL121421A0 (en) 1998-01-04
IL113519A0 (en) 1995-07-31

Similar Documents

Publication Publication Date Title
AU667459B2 (en) Compounds and methods for inhibiting gene expression
IE74705B1 (en) Compounds and methods for inhibiting gene expression
EP1212339B1 (en) 2&#39;-o-acetamido modified monomers and oligomers
EP0691977B1 (en) Oligonucleotides with amide linkages replacing phosphodiester linkages
RU2079508C1 (en) Method of nucleoside linkage by 3&#39;--5&#39;-internucleotide silyl unit
US5596091A (en) Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
EP0691979A1 (en) Novel 5&#39;-substituted nucleosides and oligomers produced therefrom
CA2080640A1 (en) Bicyclic nucleosides, oligonucleotides, process for their preparation and intermediates
US5892024A (en) Bifunctional nucleosides, oligomers thereof, and methods of making and using the same
CA2210031C (en) Solid phase synthesis of oligonucleotides
WO1995024413A1 (en) Compositions and methods for use in the synthesis of oligonucleotides
US5674856A (en) Modified oligodeoxyribonucleoditides
US6414135B1 (en) C3′-methylene hydrogen phosphonate monomers and related compounds
US6974865B2 (en) C3′ -methylene hydrogen phosphonate oligomers and related compounds
RU2131436C1 (en) Nuclease-resistant oligonucleosides, method of preparing thereof, and nuclease-resistant nucleoside dimer
Jeong et al. Synthesis and hybridization property of sugar and phosphate linkage modified oligonucleotides
WO1998049183A1 (en) Base protecting groups and process for oligonucleotide synthesis
FR2612930A1 (en) alpha Oligonucleotide probes

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued