CA1339935C - Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes - Google Patents

Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes

Info

Publication number
CA1339935C
CA1339935C CA000582338A CA582338A CA1339935C CA 1339935 C CA1339935 C CA 1339935C CA 000582338 A CA000582338 A CA 000582338A CA 582338 A CA582338 A CA 582338A CA 1339935 C CA1339935 C CA 1339935C
Authority
CA
Canada
Prior art keywords
oligodeoxynucleotide
linkage
modified
internucleotide phosphodiester
mrna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA000582338A
Other languages
French (fr)
Inventor
Joseph A. Walder
Roxanne Y. Walder
Paul S. Eder
John M. Dagle
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.)
University of Iowa Research Foundation UIRF
Original Assignee
University of Iowa Research Foundation UIRF
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22425523&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA1339935(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by University of Iowa Research Foundation UIRF filed Critical University of Iowa Research Foundation UIRF
Application granted granted Critical
Publication of CA1339935C publication Critical patent/CA1339935C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6848Nucleic acid amplification reactions characterised by the means for preventing contamination or increasing the specificity or sensitivity of an amplification reaction
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/311Phosphotriesters
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3517Marker; Tag
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3521Methyl
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3527Other alkyl chain
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Abstract

The use of oligodeoxynucleotides modified at the 3'-terminal internucleotide link as therapeutic agents by a method of hybridizing the modified oligonucleotide to a complementary sequence within a targeted mRNA and cleaving the mRNA within the RNA-DNA helix by the enzyme RNaseH to block the expression of the corresponding gene.

Description

133~93S

GRANT REFEI~ENCE
The invention described herein was made in part in the course of work under grants from the tJational Insti-tutes of Health, Grant Nos. Hl-33555 and AM-25295.

BACKGROUND OF TEIE INVENTION
It is well known that nucleic acids, i.e., deoxyribo-nucleic acids (DNA) and ribonucleic acids (RNA) are essential building blocks in living cells. These compounds are high molecular weight polymers which are made up of many "nucleotide" units, each such nucleotide unit being composed of a base (a purine or a pyrimidine), a sugar ~which is either ribose or deoxyribose) and a molecule of phosphoric acid. DNA contains deoxyribose as the sugar moiety and the bases adenine, guanine, cytosine, and thymine (which may be represented as A, G, C and T, respectively). RNA contains ribose instead of deoxyribose and uracil (U) in place of thymine.
The nucleotide units in DNA and RNA are assembled in de~inite linear sequences which determine specific biological functions. In the normal mammalian cell, D~A replicates itself, and also serves as the template ~or the synthesis of RNA molecules whose nucleotide sequences carry the inforlnation encoded by the ~NA.
RNA molecules serve several different functions within the cell. Messenger ~NA (mRNA) directs protein synthesis.
According to the well known ~atson-Crick model, DNA
molecules consist of two polynucleotide strands coiled about a common axis. The resulting double helix is held together by hydrogen bonds between complementary base pairs in each strand. ~ydrogen bonds are formed 'k 1339'~3~

only between adenine (A) and thymine (T) and between guanine (G) and cytosine (C). ~lence within the double helix, adenine (A) and thymine (T) are viewed as comple-mentary bases which bind to each other as A-T. The same is true for guanine (G) and cytosine (C), as G-C.
In a single polynucleotide strand, any sequence of nucleotides is possible. ~lowever, once the order of bases within one strand of a DNA molecule is specified, the exact sequence of the other strand is simultaneously determined due to the indicated base pairing. Accordingly, each strand of a DNA molecule is the complement of the other. In the process of DNA replication, the two strands act as templates for the synthesis of two new chains with complementary nucleotide sequences. The net result is the production of two new DNA molecules each containing one of the original strands plus a newly synthesized strand that is complementary to it.
This process is referred to as semiconservative repli-cation and allows the genetic information encoded with-in the DNA, specified by the nucleotide sequence, to be transmitted from one generation to the next.
Collectively, the genetic information of an organ-ism is termed the genome. The genome of bacteria and of all higher species is composed of DNA. The genome of viruses may be either DNA or ~NA. In any case, the genome of any particular species, whether a virus, bacteria, or higher organism has a characteristic nucleo-tide sequence wllich, stated simply, can he viewed as the "fingerprint" of that species.
In the process of transcription, DNA is copied, or transcribed, into RNA. T~le RNA molecule that is syn-thesized is single stranded. Its nucleotide sequence ~3t33~ 3rj is complementary to a segment of one cf the two strands of the DNA molecule, and hence it is an exact copy of the opposite strand except that thymine is replaced with uracil. The length of DNA that is copied to form a single RNA molecule represents one gene. Ihe human genome contains approximately 50,000 genes.
Messenger RNAs (mRNAs) carry the coding information necessary for protein synthesis. The sequence of nucleo-tides in the mRNA molecule specifies the arrangement of amino acids in the protein whose synthesis will be directed by that mRNA. Each set of three nucleotides, a unit called a codon, specifies a particular amino acid. The process by which mRNAs direct protein synthesis is termed translation. Translation of mRNAs occurs on ribosomes within the cytoplasm of the cell.
The flow of information frorn gene to protein can thus be represented by the following scheme:

Gene > mRNA > Protein For each protein made by an organism there exists a corresponding gene.
It is generally known in the field of molecular biology that oligodeoxyribonucleotides, short single-stranded DNA fragments, havin~ a nucleotide sequence complementary to a portion of a specified mRNA may be used to block the expression of the corresponding gene.
This occurs by hybridization ~binding) of the oligo-nucleotide to the mRNA, according to the rules of base pairing described above, which then prevents translation of the m~NA. It has now been discovered that the inhibition of translation observed is due to cleavage of the mRNA

1339~35 by the enzyme RNaseH at the site of the RNA-DNA double helix formed with the oligonucleotide, ~3ybridization of the oligonucleotide to the mRNA is generally not sufficient in itsel~ to significantly inhibit translation. Evidently an oligonucleotide bound to mRNA can be stripped off in the process of translation. An important corollary of this result is that for any modified oligonucleotide to efficiently block expression of a gene, the hybrid formed with the selected mRNA must be recognized by RNase~ and the mRNA
cleaved by the enzyme.
The process oE hybrid-arrested translation outlined herein has obvious implications for the use of oli~onucleotides as therapeutic agents. By selectively blocking the expression of a particular gene essential for the replication of a certain virus or bacteria an oligonucleotide could serve as an antimicrobial agent.
Similarly, an oligonucleoti~e targeted against a gene responsible or required for the uncontrolled prolifera-tion of a cancer cell would be useful as an anticancer agent. There are also applications in the treatment of genetic diseases such as sickle cell disease and thal-assemia for which no adequate treatment currently exists.
Any gene can be targeted by this approach. The problern of drug specificity, a major hurdle in the development of conventional chemotherapeutic a~ents, is immedia-tely solved by choosing the appropriate oligonucleotide sequence complementary to the selected mRr~A. This cir-cumvents toxicity resulting frorn a lack of selectivity of the drug, a serious limitation of all existing anti-viral and anticancer agents.

' ' ;41 1~3~ 3~

In addition to the use of oligonucleotides to block the express-on o~ selected genes for the treatment of diseases in man, other applications are also recognized.
Additional applications include but are not limited to _ ~ ~"
the use of such oligonucleotides in veterinary medicine, ~ D f~.
as pesticides or fungicides, and in industrial or agricul-tural processes in which it is desirable to inhibit the expression of a particular gene by an organism utilized in that process.
A major problem limiting the utility of oligonucleo-tides as therapeutic agents is the rapid degradation of the oligonucleotide in blood and within cells. Enzymes which degrade DNA or RNA are termed nucleases. Such enzymes hydrolyze the phosphodiester bonds joining the nucleotides within a DNA or R~A chain, thereby cleaving the molecule into smaller fragments.
In the past there has been some progress made in the development of oligonucleotide analo~s that are resistant to nuclease degradation, but the use of such derivatives to block the expression of specifically targeted genes has met with limited success. See, for example, Ts'o et al. U.S. Patent 4,469,863 issued Septem-ber 4, 1984; Miller et al. U.S Patent 4,507,433 issued March 26, 1985; and Miller et al. U.S. Patent 4,511,713 issued April 16, 1985. Each of the above patents have in common the objective of blocking the expression of selected genes, but the approach taken has been less than successful judging fr~m the high concentrations of oligonucleotide required and a corresponding lack of selectivity. The work described in each of the three 1~ ~9i3 ~

prior patents mentioned involves the u5e of oligonucleo-tides in which all of the phosphate groups have been modified in the form of methylphosphonates:
HO o~ B1 0 I - Cl~3-P~~
O ~ 2 o ~ B7 Oil Orl Normal Nucleotide Methylphosphonate Phosphodiester Diester B, and B2 are nucleic acid bases (either A, G, C or T).
In particular, U.S. Patent 4,469,863 involves inter alia, methylphosphonate modification of oligonucleotides.
U.S. Patent 4,507,433 involves a process for synthesiz-ing deoxyribonucleotide rnethylpllosphonates on polysty-rene supports; and U.S. Patent 4,511,713 involves deter-mining the base sequence of a nucleic acid and hybridiz-,ng to it an appropriately synthesized oligonucleotide methylphosphonate to interfere with its function.
The low level of activity observed usin~3 these rully modified methylphosphonate analogs led us to suspect that they would not form effective substrates for RNase~l when hybridized with mRNAs. Results present-ed in Example 2 show this to be the case, In an efFort to design new oligonucleotide analogs 1~3933S

o~ greater use as therapeutic agents, we undertook a study of the pathways by which oligonucleotides are degraded within blood and within cells. We have dis-covered that the sole pathway of degradation in blood (Example 3) and the predominant pathway in cells (Example 4) is via sequential de~radation from the 3'-end to the 5'-end of the DNA chain in which one nucleotide is removed at a time. This pathway of degradation is illustrated. These results provided for the first time a rational basis for the design of oligo-nucleotide analogs modified so as to inhibit their degradation without interfering with their ability to form substrates for RNaseli.
It has now been discovered that oligonucleotides modified at only the 3'-most internucleotide link are markedly protected from degradation within blood and within cells (Examples 5 and 6). Moreover, we have found that such derivatives have normal hybridization properties and do form substrates with mRNAs that are recognized and cleaved by RNaseH, thereby preventing expression oE the targeted gene (Example 7).
- The accomplishlnent oE the inhibition of expression of selected genes by oligonucleotides that are resist-ant to degradation, and that are, therefore, more effect-ive when used therapeutically, is the primary objective of this invention.
Other objectives oE the present invention include the preparation of oligonucleotides modified at the 3'-phosphodiester linkage and the formulation of the modified oli~onucleotide in a manner suitable for therapeutic 1 ~ 3 9 r~ 3 ~i use. It is also apparent that DN~ or RNA molecules so modified would be useful as hybridization probes for diagnostic applications, in which case, the modification of the 3'-internucleotide link would inhibit degrada-tion of the probe by nucleases which may be present within the test sample.

B~IEF DESCRIPTION OF TIIE DRAWINGS
Figure 1 is a schematic illustration to show how the expression of a selected gene is blocked in accord-ance with the process of this invention by hybridiza-tion of an oligonucleotide to its complementary sequence within the mRNA and cleavage of the mRNA by the enzyme RNaseH at the site of the RNA-DNA double helix.
Figure 2 illustrates the degradation of a single stranded DNA molecule by the action of a nuclease that removes sequentially one nucleotide at a time from the 3'- to the 5'-end of the chain.
Figure 3 is a drawing of an autoradio~ram demon-strating that the exclusive pathway of degradation of DNA fragments in hurnan blood is by exonucleolytic cleavage from the 3'- to the 5'-end of the DNA chain. The oligo-nucleotide utilized in this experiment, MA-15, has the sequence: 5'- GAGCACCATGGTTTC. In lanes 1-5, the oligo-nucleotide was radiolabeled at the 5'-end of the mole-cule with 32p. In lanes 6-10, the 32P-label is at the 3'-terminal internucleotide link. The labeled oligo-nucleotide was incubated in human blood at 37~C. Ali-quots were removed at the times shown and analyzed by polyacrylamide gel electrophoresis. The labeled bands ~339~35 --1 o--within the gel were visualized by autoradiography.
Figure 4 is a drawing o~ an autoradiogram demon-strating that an oligonucleotide in which only the 3'-most internucleotide link is modified is completely resistant to degradation in human blood. The oligo-nucleotide is a derivative of MA-15 in which the 3'-terminal internucleotide link is a 2,2,2-trichloro-1,1-dimethylethyl phosphotriester. Lane 1 is the oligonucleo-tide prior to incubation. Lanes 2-5 are aliquots taken after incubation of the oligonucleotide in blood for 15 minutes, 1 hour, 4 hours, and 18 hours, respectively.
The decrease in intensity of the original band is due simply to removal of the 5'-end label rather than de-gradation of the oligonucleotide. Pi is inorganic phosphate.
Figure 5 is a drawing of an autoradiogram of a Northern blot demonstratinq that oligonucleotides m~di-fied at the 3'- internucleotide link form substrates with mRNAs that are acted upon by RNaseH resulting in cleavage of the mRNA at the site of the RNA-DNA double helix. Lane 1 is mouse globin mRNA alone. Lane 2 is mouse globin mRNA incubated with RNasell in the absence of oligonucleotide. Lane 3 is (~lobin m~NA hybridlzed with MA-l5 and cleaved with RNasel~. In lane 4, the substrate for nNasel~ is ~ormed with the derivative of MA-l5 in which the 3'- terminal internucleotide link is a tri-chlorodimethylethyl phosphotriester. In lane 5, the oligonucleotide used was further modified with naphthylisocyanate at the 5'-0~l group of the molecule.
Samples were electrophoresed on a l.5S agarose gel.
After electrophoresis the gel was blotted onto (*)Gene A ( * ) Trade Mark 1~

(*) Screen Plus (DuPont), and bands were localized using P labeled MA-15 as a probe for a-globin mRNA.

SUMMARY OE' THE INVENTION
The in~ention comprises the method, means and composi-tion which together enable the use of oligonucleotides that are modified at the 3'-terminal phosphodiester linkage, and are thereby rendered resistant to degradation within cells and body ~luids, to selectively block the expression of a particular gene. The method involves the hybridization of the modified oligonucleotide to the corresponding mRNA to fonn a substrate fully capable of being recognized by the enzyme RNasel~, followed by the cleavage of the mRNA at the site of the RNA-DNA
double helix such that the expression of the targeted gene is blocked. The invention further details the use of DNA and RNA molecules modified at the 3'-terminal phosphodiester linkage as nucleic acid probes for diagnostic applications.

DETAILE~ DESCRIPTION O~ TI~E INVENTION
In the broadest terms, the objectives of the pre-sent invention are accomplished by modifying oligonucleo-tides at just the 3'-terminal phosphodiester linkage.
When this is accomplished it has been found that the oligonucleotide is markedly resistant to degradation by nucleases within cells and within blood. Equally important, such modified oligonucleotides hybridize normally with complementary nucleic acid sequences and do form substrates with mRNAs that are recognized and cleaved by RNaseH.

(*)Trade Mark 1~39935 As a result of the cleave of the mRNA, expression of the corresponding gene is selectively blocked as is desired in various therapeutic applications.
Recent advances in molecular biology employing recombinant DNA techniques have led to new diagnostic and therapeutic strategies. In DNA diagnostics, a DNA
or RNA probe is used to detect the presence of a comple-mentary nucleic acid (DNA or RNA) sequence. In this process the probe hybridizes to its complementary se-quence if it is ~resent within the sample. The target se~uence may be a DNA or RNA molecule of a bacteria or virus, or an altered gene leading to a genetic abnor-mality such as sickle cell disease. Frequently, the sample to be analyzed is immobilized on a solid support such as nitrocellulose or a nylon membrane. The probe carries a label, e.g., a radioactive, fluorescent, or enzyme marker to permit its detection. The field of DNA
diagnostics is currently under very active inYeStigation~
having applications in the diagnosis of infectious diseases, cancer, and genetic disorders. E~owever, there are as yet no products in widespread clinical use.
Existing metl-ods, although extremely useful as research tools, lack adequate sensitivity for many ~ractical applications, and are often too complicated to be carried out routinely and reliably in a clinical laboratory.
In DNA therapeutics, an oligonucleotide is used to block the expression o~ a specifically tar~eted gene hy hybridizing to a complementary sequence within the corresponding mRNA and preventing the mR~A from being translated. In this manner, synthesis of the protein 133~33~

encoded by the gene is blocked. Such is the basic con-cept employed in each of the above three mentioned prior patents.
However, recent studies which formed the essence of the present invention have shown that blocking of the expression of the targeted gene occurs not simply by the hybridization of the oligonucleotide to the mRNA, but requires that the RNA-DNA double helix so formed serve as a substrate for the enzyme RNase~l. RNaseH, a ubiquitous enzyme required for DNA replication, then digests the m~NA at the RNA-DNA duplex. Only the mRNA
is cleaved. The oligonucleotide is released intact and can react repeatedly with other mRNA molecules as indicated by the dashed line in Figure l. The effect of each molecule of the oli~onucleotide is thereby amplified, increasing the efficiency by which the expression of the targeted ~ene is blocked.
RNaseH acts only on RNA-DNA double helices. RNA-RNA duplexes do not serve as substrates for the enzyme.
Therefore, oligodeoxyribonucleotides (short single-stranded DNA fragments) are preferred as therapeutic agents, rather than ~NA molecules. Either RNA or DNA
molecules can be used as probes for diagnostic purposes.
One problem faced in both DNA diagnostics and DNA
therapeutics is the degradation of the DNA or RNA mole-cule used either as a probe or tllerapeutic agent by nydrolytic enzymes, termed nucleases, present in body fluids and tissues. This problem is of far greater concern in the case of DNA therapeutics. Oligonucleo-tides are degraded in blood, and even more rapidly 1~3!~93~

within cells, the rate of degradation being suffici-ently fast to completely abrogate the effect of the drug. In preparing clinical samples for DNA diagnostic tests, efforts are made to remove contalninating nuc-leases. However, low levels of activity may remain. In certain instances, particularly when applied to genetic tests, the detection method is based on specific cleav-age of the probe when it is hybridized to the target sequence. Non-specific degradation of the probe can give rise to an increased level of background signal, obscuring detection o~ the target-dependent cleavage of the probe.
Nucleases catalyze the degradation of DNA or RNA
strands by hydrolysis of the phosphodiester bonds which join the nucleotide units within the chain, thereby cleaving the molecule into smaller fragments. DNA or RNA molecules useful as hybridization probes range from approximately lS nucleotide5 in length to over several thousand nucleotides. DNA fragments useful as therapeutic agents range from about 10 to about 75 nucleotides in - length, preferably from 15 to 35 nucleotides. Thus, any DNA or RNA molecule used as a probe or therapeutic asent would have many potential sites at which cleav-eage by nucleases may occur.
It is ~enerally known in the field of molecular biolo~y that phosphodiester linkages normally subject to enzymatic cleavage by nucleases can be protected by modifying the phosphate ~roup. Earlier efforts, such as the three mentioned patents, have focused on deri-vatives in which all of the phosphate groups within the X

~3933~

strand are modified, or on derivatives in which the phosphate groups are extensively modified in a random fashion following the synthesis of the oligonucleotide (Tullis, R.H., P.C.T. W083/01451, 1983). Such modifica-tions, however, often interfere with hyhridization of the DNA to its target sequence, and consequently limits its utility for both diagnostic and therapeutic applica-tions. Moreover, it has been found that D~A fragments in which the phosphate groups are extensively modified do not form substrates for RNaseH (Example 2). This abrogates their use as therapeutic agents.
While not wishing to be bound by any theory, the important discoveries underlying the present invention are: (1) that the mechanism by which oli~onucleotides used as therapeutic a~ents block the expression of specifically targeted genes is to hybridize to the corresponding mRNA, forming a substrate for RNaseH
which then cleaves the m~NA; and (2) that the exclusive pathway of degradation of oligonucleotides in blood, and the predominant pathway within cells, is via sequential degradation from the 3'- to the 5'- end of the chain as outlined in Figure 2. It has thus proven possible to greatly increase the stability of DNA fragrnents by modification of only the first internucleotide link from the 3'- end of the DNA molecule. ~hen this is done, cleavage of the first nucleotide unit is blocked, and no further degradation can proceed. A variety of substitutions at the 3'- terminal internucleotide link, as explained below, will serve this purpose, the position of the modification being the important factor. As demonstrated in Example 7, such modified derivatives . ~

133933~

have normal hybridization properties and do form sub-strates with mRNAs that are recognized and cleaved by ~NaseH. Such modified oligonucleotides, thus, are useful for both diagnostic and therapeutic applications.
Oligonucleotides, althou~h highly negatively charged molecules, do enter cells at a ~inite rate. This was first suggested by a study carried out by Zamecnik and Stephenson (Proceedings of the National Academy of Sciences USA (1978) 75, 280-2~4 and 285-28~), in which an oligonucleotide ~omplementary to the 5'- and 3'-repeat sequences of Rous sarcoma virus were found to inhihit ' viral replication and the synthesis of viral proteins in chick embryo fibroblasts. In these studies, the mechanism o~ action of the oligonucleotide was not established, nor was the possiblity that the effects observed were due to degradation prod~ucts of the oligo-nucleotide ruled out. Nonetheless, ihis study, with more recent work, Heikkila et al., Nature (1987) 328, 445-449, indicates that oligonucleotides do enter cells.
Zamecnik and Stephenson also showed that the activity of the oligonucleotide could be substantially increased by modifying the 3'-OEI and 5'-OEI groups with large apolar substituents, i.e., phenyl and naphthyl g~oups.
These large apolar groups enhance the binding of the oligonucleotide to the cell membrane and transport into the cell. The introduction of these groups may also have inhibited the degradation of the oligonucleotide by e~onucleases. E~owever, only by directly modifying the phosphodiester group is it possible to precluAe cleavage of the internucleotide link within a DNA or RNA strand. Neither the degradation of the native 133~335 oligonucleotide nor the modified~ derivatives wer~ studied in this work. Even the most recent study of'the degradation of oligonucleotides is completely silent as to the pathway by which degradation occurs ~Wickstrom, E.
(1986) Journal o~ Biochemical and Biophysical Methods 3, 97-102).
Thus, while there has been some study of the degrada-tion of DNA and RNA by endogenous nucleases within human and other mammalian tissues previously, it has not hithertofore been appreciated that degradation of single-stranded DNA molecules occurs predominantly by a 3'- to 5'- exonucleolytic mechanism. Hence the unique value of the modified derivatives claimed herein could not have been predicted. Put another way, because the pathway of degradation had not been established, no basis existed for the design o~ DNA molecules in which only selected phosphate groups within the chain were modified to effect an increase in stability, such that the resulting oligonucleotide still hybridized to mRNAs to fonn substrates that are reco~nized and cleaved by RNaseH, as is required for use of the oli~onucleotide in therapeutic applications.
As heretofore mentioned, the exact chemical modi-fication at the 3'-phosphodiester linkage is not as important as that the modification be at this precise location. A limited number of other positions on the molecule may also be modified without departing from the scope of this invention. In the case of DNA diagnos-tics, it is necessary to attach a label onto the nucleic acid probe to enable its detection. A radioactive, fluorescent, electrochemical, chemilumenescent or enzyme 133~

marker may serve this purpose. For oligonucleotide probes ranging in length from 15 to about 35 nucleotides in length generally only one label is appended to the molecule. ~nger DNA or RNA molecules use~ as probes may be up to several thousand nucleotides in length. In this case the label may be introduced at a density of up to 1 for every 15 nucleotide units in the chain.
For applications in DNA therapeutics, the oligo-nucleotide may be modified at positions in addition to the 3'-phosphodiester linkage to increase its stability further or to facilitate its transport into cells.
Transport of the oligonucleotide into the target cell may be enhanced by attachment to a carrier molecule normally taken up by the cell such as transferrin the plasma iron binding protein, or epidermal growth factor, or the oligonucleotide may be attached to an antibody against a surface protein of the target cell as in the case of the so-called im~unotoxins. Alternatively, additional phosphate groups of the oligonucleotide may be modified to reduce the ne~ative charge on the molecule and thereby facilitate its transport across the lipid bilayer of the cell membrane. With such modifications, apolar substituents such as hydrocarbon chains or aro-matic groups may also be incorporated into the molecule to further enhance intracellular uptake. Modification of additional phosphate groups within the chain may also increase the stability of the oligonucleotide further by blocking the activity of 5'- to 3'- exonucleases and endonucleases that are present in certain cells (see Example 4). Although degradation by these pathways 133~1335 is much less rapid than degradation from the 3'- end of the chain, the added effect of blocking these activities as well, particularly 5'- to 3'-exonucleolytic cleavage, may be beneficial in many cases. In these various applications, however, it is important that the modifica-tions be restricted to a limited number of the phospho-diester groups. If most or all of the phosphodiester linkages within the chain are modified this will frequently interfere with the hybridization properties of the oligonucleotide, and more importantly, it will preclude the use of such derivatives as therapeutic agents because they will no longer form substrates with targeted mRNAs that are acted upon by RNase~l. It is essential that there be one or more continuous stretches of the oligonucleotide chain in which the phosphodiester linkages are unmodified. The length of this unmodified portion of the chain must be at least 4 nucleotides, and preferably greater than 7 nucleotides in length.
When this is accomplished, the oligonucleotide will be protected from degradation due to the modification of the 3'-terminal phosphodiester linkage, and will also hybridize with mRNAs to form substrates for RNaseH, which will cleave the m~NA across from the unmodified portion of the oligonucleotide, enabling the use of such derivatives as therapeutic agents to block the expression of selected genes. The method of modifying the 3'-phosphodiester linkage will now be described.
The modifying moiety is not critical. It can be selected from any of the following functionalities:

:~339~335 R-O-P=O ~ O-P=O alkyl or aryl O O phosphotriesters R-P=O ~ P=O alkyl or aryl O O phosphonates ~_p=o hydrogen O phosphonates R O
R-N-P=O ~ N-P=O alkyl or aryl Il O 11 I phosphoramidates S-P=O phosphorothioates o Se-P=O phosphoroselenates o ~39935 In these structures, R may vary in length from 1 to about 20 carbon atoms, and may contain halogen substi-tuents (as in Examples 5 and 6) or other heteroatoms, e.g., N, O, or S, attached to the chain. The methods required to prepare these derivatives are simple, straight-forward reactions well known to those skilled in the art. See for example, the following literature refer-ences, each of which discloses modifications of the type expressed herein: Letsinger, et al., Journal of the American Chemical Society, t1982), 104, 6805-6806;
Stec et al., Journal of the American Chemical Society (1984), 106, 6077-6079; Miller et al., ~iochemistry (1986), 25, 5092-5097; and Froehler, et al., Tetrahedron Letters, (1986), 27, 469-472.
The preferred modification is a phosphotriester, for simplicity of synthesis and the diversity of func-tional groups which may be attached to the phosphorus atom. Incorporation of a large l-ydrophobic substituent at this position, for example, may be utilized to faci-litate intracellular uptake. As demonstrated in the present work, phosphotriesters are also resistant to hydrolysis by nucleases, and when incorporated at only a limited number of positions within an oligonucleotide chain, the molecule remains able to hybridize normally with m~NAs to form suhstrates which are cleaved by RNaseH.
In all methods now commonly used, oligonucleotides are synthesized from the 3'- to the 5'- end of the chain. The first residue is coupled to a solid support, such as controlled pore glass, through the 3'-OH group.

1~39935 ~f the synthesis is carried out in solution, rather than on a solid support, the 3'-0~1 of the first residue is blocked by a protecting group which is removed at the culmination of the synthesis. Usually, one nucleo-tide unit is added at a time to the 5'-0~1 group o~ the growing chain. Alternatively, a block of several resi-dues may be added in a single reaction. There are several methods by which the internucleotide link to the 5'-0~ group may be formed. These procedures are reviewed in the ~ollwing monograph, "Oligonucleotide Synthesis: A Practical Approach" (Gait, ~.J., ed) IRL
Press, Oxford (l9~4), The following reaction scheme illustrates the phosphoramidite method, the preferred chemistry at present.

~_O ~ O B2 DMT-O ~ O 2 + ~ ~I
coupling 1~

3 ~ o~ ~ 1 S . S . C=O
~2 oxidation V

~3~5 ~IT-O-~ o ~ 2 o ~-0- P=O
~1~ O ~ 1 ~1 o C=O
~2 DMT is the dimethoxytrityl protecting group; S.S. is the solid support on which the synthesis is carried out. Bl and B2 are nucleic acid bases (either A, G, C or T).

~3~9~35 The immediate product of this reaction is a phospho-triester. At the end of the synthesis, the protecting group R is removed to yield a phosphodiester linkage.
Typically, the ~-cyanoethyl group is used for this purpose. Alternatively, R may be a group which is not removed, in which case the final product is a phos-photriester, the group R remaining permanently attached to the phosphorus atom. It is in this manner that a modified phosphotriester may be incorporated at the first internucleotide linkage from the 3'-end of the chain. By essentially the same chemistry an alkyl or aryl phosphonate may be introduced specifically at this position, in which instance the group R is attached directly to the phosphorous atom rather than through oxygen. Similarly, a hydro~en phosphonate may be intro-duced at this position. Such derivatives may be used directly, or oxidized to give phosphotriesters or phos-phoramidates incorporated at the 3'-terminal internucleotide linkage. The following reference discloses modifications of the hydrogen phosphonate group, Froehler, B.C., Tetrahedron Letters, ~1986), 27, 5575-5578. A phosphoro-thioate or phosphoroselenate group may be incorporated at the 3'-terminal internucleotide link by carrying out the oxidation step in the phosphoramidite method with sulfur, or YSeCN, respectively, Stec. et al., Journal of the American Chemical Society, (1984), 106, 6077-6079. At the completion of the synthesis, following the removal of all protecting groups, the final prol~uct may be used directly or further purified by chromato~raphic methods well known to those skilled in the art, see for 1~3'-3~

example "Oligonucleotide Synthesis: A Practical Approach"
(Gait, M.J., ed.) IRL Press, Oxford (1984).
The procedures just described are generally appli-cable for the synthesis of oli~onucleotides up to approxi-mately 100 residues in length. ~n order to synthesize a longer DNA or RNA molecule in which the 3'-terminal phosphodiester group is modified, as may be required for use as a hybridization probe in diagnostic tests, a combination of both chemical and enzymatic methods is used. To accomplish the synthesis of such a sequence, a short oligonucleotide in which the 3'-terminal phospho-diester linkage was modified, prepared by chemical methods, is enzymatically 1isated onto the 3'-OH group of a longer DNA or RNA chain.
Methods of formulation and administration of the modified oligonucleotides described herein are obYious to those of ordinary skill in the medical arts, see for example, Avis, K. (1985) in Remington's Pharmaceutical Sciences, ~Gennaro, A.R., ed) pp. 151~-1541, Mack Publishing Company, Easton, Pa.
Such method9 of administration may include but are not limited to t~pical, oral, or parenteral routes depending on the disease state.
Appropriate vehicles for parenteral administration include 5~ dextrose, normal saline, Ringer's solution and Ringer's lactate. The material, of course, must be sterile and substantially endotoxin free. It may be possible in certain cases to store the product as a lyophilized powder which would then be reconstituted when needed by addition of an appropriate salt solution.
Ihe following examples are offered to further illus-trate, but not limit, the process, product and medical .
~'t': ~, r ¦l 3 5~ 3 3 ~

techniques of the invention, and serve to point out the unique features of the invention which enable it to over-come limitations of earlier contributions to the field.

EXAMPLE
Demonstration that the Inhibition of Translation of a Targeted mRNA by a Complementary Oli~onucleotide is Dependent on the Cleavage of the mRNA by RNaseH
For the purpose of these experiments two oligonucleo-tides were synthesi~ed: 5'-GAGCACCATGGTTTC, MA-15;
ands 5'-TGTCCAAGTGATTCAGGCCATCGTT, CODMB-25. Both were prepared on a Beckman automated DNA synthesizer using the phophoramidite method. MA-15 is complementary to a se~uence within mouse ~-globin mRNA spanning the initiation codon. CODMB-25 hybridizes to a sequence within the coding region of mouse ~-globin mRNA. Mouse globin mRNA was isolated from reticulocytes obtained from mice rendered anemic by treatment with phenylhydrazine as described by Goosens and Kan (Methods in Enzymology (Antonini, E., Rossi-~ernadi, L., and Chiancone, E., eds.) 76, 805-817, 19~1). In vitro translation reactions were carried out in the rabbit reticulocyte lysate system. The reticylocyte lysates used were purchased from ProMega Biotec, and contain RNase~l. Translation reactions were programmed with 1 u~ of total mouse reticulocyte RNA. The reactions were allowed to proceed for 1 hour at 30~C. Each sample contained 45 uCi of [35S~methionine (Amersham) in a total volume of 25 ul.
Protein synthesis was measured by incorporation of [35S]methionine into material precipitatable in 10%

~39935 trichloracetic acid. In control reactions in the ahsence of oligonucleotide, synthesis of ~ globin accounts for 60~ of the incorporated counts; ~ globin synthesis accounts for 40% of the radioactivity incorporated into protein. Reactions were carried out in both the presence and absence of poly rA/oligo dT t500 ug/ml). Poly rA/oligo dT is a competitive inhibitor of RNaseH, and at the concentration used almost completely blocks the activity of the enzyme. Both MA-15 and CODMB-25 very effectively inhibited translation of the targeted mRNA
(see Table 1). MA-15 is complementary to 12 of 15 residues in the corresponding sequence of ~-globin mRNA, and, therefore, also inhibits ~-globin synthesis to some extent. Poly rA/oligo dT completely blocked the inhibitory effect of the oligonucleotides, indicating that the arrest of translation is mediated entirely by the cleavage of the mRNA by RNaseH.

Table 1. Role of RNaseEI in ~ybrid-Arrested Translation % Inhibition of Globin Synthesis Oligonucleotide - poly rA/oligo dT +poly rA/oligo dT
MA-15 (8 M) 51 3 CODM~-25 (4 uM) 61 5 Oligodeoxynucleotide Methylphosphonates Do Not Form Substrates for RNaseH
Two related substrates for RNaseE~ were compared:
p y rA/oligO dTl2_18 and poly rA/oligo dT18 methyl-phosphonate (MP). Oligo dT12 18 was purchased from ~3~9~3~

Pharmacia PL Biochemicals; the c}-ain length varies from 12 to 18 residues. Oligo dT18MP was synthesized on a Beckman automated DNA synthesizer using the phosphon-amidite method. The 5'-terminal internucleotide link is a normal phosphodiester; the remaining 16 phosphate groups in the chain are methylphosphonates. Poly rA
labeled with tritium was purchased from Amersham.
Each reaction was conducted in a final volume of 100 ul containing 44 picomoles of the substrate, either H-2~1Y rA/oligo dT12 18 or H-poly rA/oligo dT18MP, in 50 mM Tris-HCl buffer pH 8.0, 20 mM KCl, 9 m~ MgC12, 1 mM 2-mercaptoethanol and 25 ug of bovine serum albumin.
The reaction was initiated by the addition of 2.3 units of E.coli RNase~ (Bethesda Research Laboratories) and allowed to proceeed for 30 minutes at 37~C. After the reaction, 0.2 ml of carrier DNA solution (0.5 mg/ml of salmon sperm DNA, 0.1 M sodium pyrophosphate, and 1 mM
ethylenediamine tetraacetic acid) plus 0.3 ml of 10%
trichloracetic acid was added to each sample. The tubes were then incubated on ice for 20 minutes, and afterwards centrifuged at 16,000 X g for 15 minutes.
Cleavage of the poly rA strand by RNase~ releases smaller fragments that are not precipitated by trichloroacetic acid, and hence remain in the supernatant. The super-natant was carefully removed, mixed with liquid scin-tillation cocktail, and counted for radioactivity. The reaction with poly rA/oligo dT proceeded nearly to completion - 85% of the radioactive counts were solu-bilized. In contrast, less than 1~ of poly rA/oligo dT18MP was digested by the enzyme. Thus, when much or all of the phosphate backbone o~ an oligodeoxynucleotide ~39~35 -2~-analogue is modified, it does not form a substrate with the complementary RNA sequence that is recognized and cleaved by RNase~.

Oligonucleotides are Degraded in Llood Exclusively by a 3'- to 5'-Exonucleolytic Activity The purpose of this study was to establish the pathways by which oligonucleotides are degraded in blood. Surprisingly, a single pathway was observed:
sequential degradation from the 3'-to the 5'- end of the DNA molecule.
The oligonucleotide MA-15 described in Example 1 was radioactively labeled with 32p at the 5'- end of the molecule using y32P-adenosine triphosphate (y 32p_ ATP, Amersham) and T4 polynucleotide kinase (Bethesda Research Laboratories):
5-*GAGCACCATGGTTTC
where the asterisk represents the radiolabel. Separ-ately, a radiolabeled derivative was prepared in which 32p was introduced at the 3'most internucleotide link usiny ~3 P-deoxycytidine triphosphate (dCTP) and CNA
polymerase I ~5ethesda Research Laboratories):
5'-GAGCACCATGGTTT*C
The reaction catalyzed by DNA polymerase I is between 5'- GAGCACCATGGTTT and dCTP and requires the comple-mentary DNA strand as a template. A 50 fold excess of unlabeled MA-15 was added after the reaction to insure that the labeled derivative is single stranded. These ~rocedures are standard methods for radiolabeling DNA
fragments, see for example, Maniatis, T., Fritsch, E.F., and Sambrook, J., Molecular Cloning: A Laboratory 1339.~3~

r1anual, Cold Spring Harbor Laboratory, New York, pp.
114-115 and 122-126, lg82~

The radiolabeled oligonucleotide (5 X 10 counts/min) plus 2.5 nanomoles o~ unlabeled MA-15 were added to 48 ul of freshly ~rawn human blood anticoagulated with heparin. Samples were incubated at 37~C, body temperature.
Aliquots of 6 ul were removed at 15 minutes, 30 minutes, 1 hour, and 2 hours. A fraction of each aliquot was loaded onto a polyacrylamide gel and electrophoresed to separate the products on the basis of size. To visual-ize the radiolabeled products, the gel was exposed to x-ray film overnight. A drawing o~ the autoradiogram is shown in Figure 3.
Degradation of the oligonucleotide la~eled at the 5'- end of the molecule (lanes 1-5) gave rise to a ladder of discrete products of decreasing size indicative of sequential exonucleolytic cleavage of the strand from the 3'- toward the 5'- end of the chain. Had cleavage occurred within the middle o~ the strand, products of intermediate length would have appeared throughout the time course. When ~1A-15 was labeled at the 3'-end of the molecule, the resulting autoradiogram, lanes 6-10, showed, over time, disappearance of the full-length 15-mer, and a corresponding increase in the mononucleotide pC. This result likewise demonstratcs a 3'- to 5'- exonuc-leolytic activity. The lack of products o~ intermediate size rules out the presence of a 5'- to 3'- exonucleolytic activity, or an endonucleolytic (internal) cleavage activity.

i339i335 By the same methods as just described, we showed that in blood from other mammalian species, specifically mouse, rabbit, and rat, oligonucleotides are also degraded solely by sequential cleavage of mononucleotides from the 3'-end of the chain.

The Principal Pathway of ~gradation of Oligonucleoti~es in E~uman Cells is by Sequential Exonucleolytic Cleavage of the Strand From the 3'- to the 5'- End of the Chain The purpose of these studies was to determine the pathways by which oligonucleotidec are degraded by nuc-leases present within human cells. Radiolabeled deri-vatives of MA-15 were prepared as in Example 3. Degrada-tion of the oligonucleotide was studied in extracts prepared from the human erythroleukemia cell line K562.
The K562 cell line was first isolated by Lozzio, C.B., and Lozzio, B.B., Blood (1975) 45, 321-334, and has been studied extensively as a mo~el for red cell differ-entiation. The cells were suspended in 10 mM tris buffer p~3 7.4 with 5 mM MgC12, 1 mM CaC12, 100 mM NaCl, and 0.1% triton X-100 and homogenized with a cold motor driven teflon pestle homogenizer for 30 seconds. Cell membranes and organelles were furt3~er disrupted by sonication by five 15 second bursts using a Biosonic sonifier set cn full power. Remaining cell debris was removed from the extracts by cerltrifugation for 5 minutes at 12,000 X g.
~ Trade Mark 133993~

The radiolabeled oligonucleotide was incubated in KS62 cell extracts under the same conditions as decribed in Example 3 and the degradation products analyzed by polyacrylamide gel electrophoresis. Degradation o~ the oligonucleotide was much faster than that in blood.
The primary pathway of degradation was again a 3' to 5' exonucleolytic activity. The half-life for cleavage of the 3'- tenminal ' internucleotide link was 2 minutes.
A slower 5'- to 3'-exonucleolytic activity was also observed. The half-life for cleavage of the S'- terminal nucleotide unit from the chain by this mechanism was 20 minutes.
Using the same methods as above, the pathways of degradation of oligonucleotides in extracts prepared from other mammalian cells have also been studied.
These systems include African green monkey kidney cells tCOS cells) and mouse liver cells. The results suggested a low level of endonucleolytic activity (cleavage in the middle of the strand), as well as a S'- to 3'-exo-nuclease, but again the major pathway of degradation of the oligonucleotide in each of these systems was by a 3'- to 5'-exonuclease.

EXAMPLE S
Modification of Oligonucleotides at the 3'- Terminal ~nternucleotide Link Completely E310cks Their Degradation in Eluman Blood A derivative of the oligonucleotide MA-lS was prepared in which the first internucleotide link from the 3'-end of the molecule was modi~ied as a 2,2,2-trichloro-1,1- dimethylethyl phosphotriester. The phosphotri-ester was introduced into the chain using the corres-13 39~33~

ponding phosphorodichloridite:
CH3 Cl Cl C--C-O-P
CH3 Cl To 20 micromoles o~ 5'-deoxythymidine dissolved in 1 cc of 10~ pyridine in acetonitrile was added 19 micromoles of the phosphorodichlorodite. The reaction was allowed to proceed for 5 minutes at room temperature. The result-ing monochlorodite was added to 1 micromole of deoxycy-tidine attached to controlled pore glass beads (American ~ionetics). The coupling reaction to produce the dinucleo-tide phosphotriester was allowed to proceed for lS
minutes at room temperature. Excess reagents and by-products of the reaction were washed away with anhydrous acetonitrile. The remaining portion of the DNA strand was synthesized by the ~- cyanoethyl phosphoramidite ~ethod on a Beckman automated DNA synthesizer. Only the 3'- terminal internucleotide link is modified. The remaining 13 phosphate groups in the chain are normal phos~hodiesters.
The modified oligonucleotide was labeled with 32p at the 5'-terminal OH group using y P-ATP and T4 polynucleotide kinase as descri~ed in Example 3. The radiolabeled oligonucleotide was incubated in human blood at 37~C. Aliquots were removed at various times and analyzed by polyacrylamide gel electrophoresis. A
drawing of the autoradiogram of the gel is shown in Figure 4. ~o degradation of the oligonucleotide occurred over a period of 18 hours. The gradual decrease in the intensity of the band due to the full length molecule was due to removal of the 32p label from the olisonucleotide 13 3~3~

by a phosphatase present in blood, not to cleavage of the strand.
Similar studies were carried out in which the modi-fied oligonucleotide was incubated in blood from the mouse. Again, no degradation of the oligonucleotide was observed.

Modification of Oligonucleoti~es at the 3'-Terminal Internucleotide Link Markedly ~nhihits Their Degradation by Nucleases Present in Human Cells The derivative of MA-15 described in Example 5 in which the 3'-ter~ina~ internucleotide link was modi~ied as a 2,2,2-trichloro-1,1-dimethylethyl phosphotriester was also used in this study. Extracts from K562 cells were prepared as in Example 4. The modified oligonucleotide was incubated in the cell extracts at 37~C. Aliquots were removed at various times, and the products of degradation were analyzed by polyacrylamide gel electrophoresis.
Degradation of this modified oligonucleotide by the rapid 3'- to 5'-exonuclease present in the cell extracts waS completely blocked. This led to a marked increase in stability of the modified derivative compared to the native oligonucleotide. The half-life was prolonged by 10-fold. Degradation occurred by the slower 5'- to 3'-exonucleolytic activity present in the K562 cell extracts (see Example 4). The half-life for cleavage of the 5'-terminal nucleotide from the chain was about 20 minutes.

133g33~

Similar studies were carried out in which the modi-fied oligonucleotide was incubated in extracts prepared from COS cells (African ~reen monkey kidney) and mouse liver cells. In both cases, the half-life of the deri-vative modified at the 3'- terminal internucleotide link was increased 10- to 15-fold compared to the un-modified oligonucleotide.

Oli~onucleotides Modified at the 3'-Terminal Internucleotide Linkage Form Substrates with mRNAs That are Cleaved by RNaseH
Mouse globin mRNA was isolated from mice rendere~
anemic by treatment with phenylhydrazine as described in Example 1. Three oligonucleotides were compared in this study; MA-15, the derivative of MA-15 utilized in Examples 5 and 6 in which the 3'- terminal internucleo-tide link is modified as a trichlorodimethylethyl phos-photriester (MA-15-TCDM), and a derivative further modified at the 5'-terminal 0ll group with naphthyli-socyanate (NPH-MA-15-TCDM). Mouse globin m~NA ~2 micro-grams) was added to l.l nanomoles of the oligonucleotide in a final volume of 22 microliters containing 10 mM
tris-HCl buffer at p~l 7.5, 5 mM MgC12, 2S mM NaCl, and 1 mM dithiothreitol. The reaction~ were initiated by the addition of 2 units of E.coli RNaseH, and were allowed to proceed for 30 minutes at 37~C. The reactions were stopped by extraction of the samples with a 1:1 mixture of phenol and chloroform. The mRNA species remaining after ~,~

$ ~

digestion with RNaseH were isolated by precipitation wi~h ethanol and analyzed on Northern blots.
The RNA samples were reacted with 1 M slyoxal in a ~ volu~e/volume) mixture of 10 m~l sodium phosphate buffer at pll 6.5 and dimethylsulEoxide for 1 hour at 50~C, and then electrophoresed on a 1.5~ agarose gel.
The RNAs were transferred from the gel to a Gene Screen Plus filter ~DuPont-NEN) by capillary blotting accord-ing to the method described by the manufacturer. Pre-hybridization of the filter was done in 1~ sodium do-decylsulfate, 1 M NaCl, 10% dextran sulfate, 50 mM
sodium phosphate pH 6.5 for 2 hours at 35~C. Hybridi-zation was conducted in S ml o~ the prehybridization buffer containing 200 microgram/ml denatured salmon sperm DNA 21us 2 x 106 counts/min of MA-15, radiolabeled with 32p at the 5'-end of the molecule, for 24 hours at 35~C. After the hybridization, the filter was washed in the following order: once in 2 x SSC ~O.lS M NaCl, 0.015 M sodium citrate) for S minutes at room tempera-ture, twice in 2 X SSC plus 1~ sodium dodecylsulfate for 30 minutes at 35~C, and once in 0.1 X SSC for 5 minutes at room temperature. The filter was blotted dry and exposed to t~)~dak XAR-S film for autoradiography.
A drawing of the autoradiogram is shown in Figure 5.
Using radiolabeled MA-15 as a ~robe, a single band due to ~-globin mRNA was detected on the Northern blot. In the absence of oligonucleotide, no cleavage of the mRNA occurred during the ~ncubation with RNase~l (lane 2~. For the reaction in the presence o~ MA-lS
llane 3), a marked decrease in the intensity of the band for ~-globin mRNA is observed due to the cleavage of the mRNA by ~NaseH at the site o~ hybridization (*)Trade Mark '3 3 5 with the probe. A similar decrease in intensity of the ~-globin mRNA band was observed for the reactions with each o~ the modified oligonucleotides (lanes 4 and 5), indicating that they also hybridize with the mRNA to form a substrate that is recognized and cleaved by RNaseH.

Inhibition of Expression of the C-MYC Oncogene in the Murine Plasmacytoma Cell Line MOPC 315 with Complementary Oligonucleotides Modified at the 3'-Terminal Internucleotide Link C-myc is a cellular oncogene aberrantly expressed by a number of human and animal tumor cells both in vivo and in cell culture. In the case of the murine plasmacytoma cell line MOPC 315, the c-myc gene is translocated from its normal position on chromosome 15 to the immunoglobulin locus on chromosome 12 and is expressed at an abnormally high level.
Two oligonucleotide analogs complementary to the sequence of c-myc mRNA coding for amino acid residues 191 to 1~8 were synthesized:
5'-G~TAGGGAAAGACCACTGAGGGT~C
5'-(PN)-GTAGGGAAAGACCACTGAGGGT~C
where (~) represents a trichlorodimethylethyl phosphotriester and (PN) -N~l2 2 2 ~-The TCDM phosphotriester was incorporated into the chain by the procedure described in Example 5. The i339~3~

remaining portion of the sequence containing unmodified phosphodiester groups was synthesized by the phos-phoramidite method. The 5'-terminal group (PN) was introduced using the reagent(*)Aminolink from Applied ~iosystems according to the procedure described by the manufacturer. For comparison, the unmodified oligo-nucleotide having the same base sequence was also prepared, as well as an oligonucleotide complementary to human albumin mRNA:
5'-T~CCCTTCATCCCGAAGTT~C
where (~) again represents the TCDM group.
MOPC 315 cells were maintained in RPMI 1640 culture media containing 10% fetal calf serum at 37~C with 7~
C02. Log ~hase MOPC 315 cells were harvested by centri-fugation (250 X g for 19 minutes at 23~C) and resus-pended at a density of 5 X 106/ml in HB101 serum-free medium (DuPont). One ml aliquots of this cell suspen-sion were incubated in the presence of oligonucleotide for 4 hours at 37~C and 7~ C02. Additional one ml aliquots were incubated concommitantly without oligo-nucleotide. Cell~ from each group were then collected by centrifugation. Total cellular RNA was prepared as described by Chir~win et al., Biochemistry 18, 5294-5299 (197g), and analyzed by Northern blots as described in Example 7. The filters were probed with oligo-nucleotide complementary to c-myc mRNA labeled with 32p using polynucleotide kinase and Y32P-ATP as in previous examples.
Both of the anti-c-myc oligonucleotides modified at the 3'-terminal internucleotide link with the TCDM
group, at a ~o~entration of l micromolar, decrea~ed the level of e-n~c mRNA ~y ~5% when (*)Trade Mark 1~3~3~

compared to the steady-state level of the mRNA in MOPC
315 cells incubated in the absence of oligonucleotide.
The unmodified oligonucleotide against c-myc and the anti-albumin oligonucleotide had no effect on c-myc mRNA levels at a concentration of either 1 or 5 micro-molar. This result is consistent with recent studies of another lymphoid cell line in which no inhibition of expression of the c-myc gene was found at concentrations of an unmodified anti-c-myc oligonucleotide below a concentration of 15 micromolar (Heikkila et al. t1987) ~ature 328, 445-449). The modified derivatives described herein, thus, are between 20- and 50-fold more effective at inhibitins the expression of a targeted gene in intact cells than is the corresponding unmodified oligonucleotide.
In summary, the results presented show that oligo-nucleotides in which the 3'-terminal phosphodiester linkage is modified are resistant to nuclease digestion within cells and body fluids, retain normal hybridization properties with complementary nucleic acid sequences, and when hybridized to specifically targeted mRNA species form substrates which are recognized and cleaved by the enzyme ~NaseH, enabling the oligonucleotide to be used to block the expression of the corresponding gene as is desired in various therapeutic applications.
Other modifications of the oligonucleotide may be made without departing from the scope and .spirit of the invention, the important factor and contribution being the discovery that modification of the 3'-terminal phosphodiester linkage alone is sufficient to markedly X

1~ ~>3~'~35 increase the resistance of the oligonucleotide to degradation, and that such d~rivatives are able to form substrates with RNAs that are recognized and clea~ed by RNase~!.
Additional substituents may be added to further increase the stability o~ the oligonucleotide against degradation, to facilitate ntracellular transport, or to attach labels, e.g., radioactive, fluorescent, or enzy~e markers, for use of the oligonucleotide as a hybridization probe in diagnostic tests. It is lmportant that such further modifications be restricted to a li~ited number of other sites on the molecule. If most or all of the phos~hate grcups within the chain are modified, the hybridization properties of the oligcnucleotide may be adversely affected, and it will not form substrates with mRNAs that are acted upon by RNase~. This latter fact defeats the use of high]y modified oligonucieotide analogs as therapeutic a~ents to block the expresslon of specifically targeted genes.
It therefore can be seen that the invention accom-plishes at least all of the objectives heretofore stated.

Claims (30)

  1. CLAIMS:

    An in vitro method of blocking expression of a targeted gene in a eukaryotic cell which expresses the targeted gene comprising:
    contacting the cell with an oligodeoxynucleotide, which oligodeoxynucleotide has (a) complementarity for and is hybridizable to an mRNA
    region within mRNA transcribed from the targeted gene, such complementarity being sufficient to ensure specificity of hybridization of the oligodeoxynucleotide to the region within the mRNA transcribed from the targeted gene;
    (b) a modified 3'-terminal internucleotide phosphodiester linkage, which linkage is resistant to 3' to 5' exonuclease degradation;
    (c) one or more additional modified internucleotide phosphodiester linkages, which additional modified internucleotide phosphodiester linkage(s) is/are resistant to nuclease degradation; and (d) a continuous stretch of at least four internucleotide phosphodiester linkages which are unmodified;
    under conditions such that said oligodeoxynucleotide enters the cell in sufficient quantity to block the expression of the targeted gene.
  2. 2. The method of Claim 1 wherein the eukaryotic cell is in cell culture or tissue culture.
  3. 3. The method of Claim 1 wherein the modified 3'-terminal internucleotide phosphodiester linkage of the oligodeoxynucleotide is a phosphotriester, phosphonate, phosphoramidate, phosphorothioate, or phosphoroselenate linkage.
  4. 4. The method of Claim 1 wherein the additional modified internucleotide phosphodiester linkage of the oligodeoxynucleotide is at the 5'-terminal internucleotide phosphodiester linkage.
  5. 5. The method of Claim 1 wherein the additional modified internucleotide phosphodiester linkage of the oligodeoxynucleotide is a phosphotriester, phosphonate, phosphoramidate, phosphorothioate, or phosphoroselenate linkage.
  6. 6. The method of Claim 1 wherein the oligodeoxynucleotide further contains one or more additional modified internucleotide phosphodiester linkages, which additional modified internucleotide phosphodiester linkage(s) enhance(s) membrane crossing ability of said oligonucleotide, thus facilitating transport of said oligodeoxynucleotide into the cell.
  7. 7. The method of Claim 6 wherein the additional modified internucleotide phosphodiester linkage of the oligodeoxynucleotide is at the 5'-terminal internucleotide phosphodiester linkage.
  8. 8. The method of Claim 1 wherein the oligodeoxynucleotide is from about 10 to about 75 nucleotides in length.
  9. 9. The method of Claim 1 wherein the oligodeoxynucleotide is attached to a carrier molecule, which carrier molecule is capable of being taken up by a cell.
  10. 10. The method of Claim 1 wherein the oligodeoxynucleotide continuous stretch is greater than seven internucleotide phosphodiester linkages.
  11. 11. The method of Claim 1 wherein all oligodeoxynucleotide phosphodiester linkages outside the continuous stretch are modified.
  12. 12. An oligodeoxynucleotide having: (a) a modified 3'-terminal internucleotide phosphodiester linkage, which modified 3'-terminal internucleotide phosphodiester linkage is resistant to 3' to 5' exonuclease degradation; (b) one or more additional modified internucleotide phosphodiester linkages, which additional modified phosphodiester linkage(s) is/are resistant to nuclease degradation; and (c) a continuous stretch of at least four internucleotide phosphodiester linkages that are unmodified.
  13. 13. The oligonucleotide of Claim 12 further having one or more additional modified internucleotide phosphodiester linkages, which additional modified internucleotide phosphodiester linkage (5) facilitate(s) intracellular transport of said oligodeoxynucleotide.
  14. 14. The oligodeoxynucleotide of Claim 12 wherein said oligodeoxynucleotide is from about 10 to about 75 nucleotides in length.
  15. 15. The oligodexoynucleotide of Claim 12 wherein said oligodeoxynucleotide is attached to a carrier molecule, which carrier molecule is capable of being taken up by a cell.
  16. 16. The oligodeoxynucleotide of Claim 12 wherein the additional modified internucleotide phosphodiester linkage is at the 5'-terminal internucleotide phosphodiester linkage.
  17. 17. The oligodeoxynucleotide of Claim 12 wherein the continuous stretch of internucleotide phosphodiester linkages which are unmodified is greater than seven internucleotide phosphodiester linkages.
  18. 18. The oligodeoxynucleotide of Claim 12 wherein all phosphodiester linkages outside the continuous stretch are modified.
  19. 19. The oligodeoxynucleotide of Claim 12 wherein the modified 3'-terminal internucleotide phosphodiester linkage is a phosphotriester linkage, a phosphonate linkage, a phosphoramidate linkage, a phosphorothioate linkage, or a phosphoroselenate linkage.
  20. 20. The oligodeoxynucleotide of Claim 12 wherein the additional modified internucleotide phosphodiester linkage(s) is/are a phosphotriester linkage(s), phosphonate linkage(s), phosphoramidate linkage(s), phosphorothioate linkage(s), or a phosphoroselenate linkage(s).
  21. 21. The oligodeoxynucleotide of Claim 12 having: (a) a phosphotriester linkage at the 3'-terminal internucleotide position; (b) one or more additional phosphotriester linkage(s) at the 5' end of the oligodeoxynucleotide; and (c) a continuous stretch of at least four internucleotide phosphodiester linkages which are unmodified.
  22. 22. An oligodeoxynucleotide of Claim 12 having: (a) a phosphoramidate linkage at the 3'-terminal internucleotide position; (b) one or more phosphoramidate linkage(s) at the S' end of the oligodeoxynucleotide; and (c) a continuous stretch of at least four internucleotide phosphodiester linkages which are unmodified.
  23. 23. The oligodeoxynucleotide of Claims 12-22, the nucleotide sequence of which has complementarity for and is hybridizable to a nucleotide sequence within the mRNA
    transcribed from a targeted gene.
  24. 24. The oligodeoxynucleotide of Claims 12-23 for use in blocking expression of a targeted gene.
  25. 25. The oligodeoxynucleotide of Claims 12-23 for therapeutic use.
  26. 26. The oligodeoxynucleotide of Claim 24 wherein the RNA-DNA duplex is a substrate for RNaseH and the mRNA within the mRNA-DNA duplex is cleaved by RNaseH to block expression of the targeted gene.
  27. 27. An oligodeoxynucleotide of seven or greater internucleotide linkages having: (a) complementarity for and being hybridizable to a nucleotide sequence within the mRNA
    sequence of a targeted gene; (b) a modified 3'-terminal internucleotide phosphodiester linkage, which modified 3'-terminal internucleotide phosphodiester linkage is resistant to 3' to 5' exonuclease degradation; (c) one or more additional modified internucleotide phosphodiester linkages, which additional modified phosphodiester linkage(s) is/are resistant to nuclease degradation; and (d) a continuous stretch of at least four internucleotide phosphodiester linkages that are unmodified.
  28. 28. The oligodeoxynucleotide of Claim 16 for use in blocking expression of a targeted gene.
  29. 29. The oligodeoxynucleotide of Claim 27 for therapeutic use.
  30. 30. The oligodeoxynucleotide of Claim 28 wherein the RNA-DNA duplex is a substrate for RNaseH and the mRNA within the mRNA-DNA duplex is cleaved by RNaseH to block expression of the targeted gene.
CA000582338A 1987-11-30 1988-11-04 Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes Expired - Lifetime CA1339935C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12656487A 1987-11-30 1987-11-30
US126,564 1987-11-30

Publications (1)

Publication Number Publication Date
CA1339935C true CA1339935C (en) 1998-06-30

Family

ID=22425523

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000582338A Expired - Lifetime CA1339935C (en) 1987-11-30 1988-11-04 Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes

Country Status (7)

Country Link
US (3) US5491133A (en)
EP (1) EP0348458B1 (en)
JP (1) JP3019994B2 (en)
AT (1) ATE151467T1 (en)
CA (1) CA1339935C (en)
DE (1) DE3855864T2 (en)
WO (1) WO1989005358A1 (en)

Families Citing this family (427)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5256775A (en) * 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
CA2058632C (en) * 1989-06-05 2004-08-24 Brian C. Froehler Exonuclease-resistant oligonucleotides and methods for preparing the same
CA2029273A1 (en) * 1989-12-04 1991-06-05 Christine L. Brakel Modified nucleotide compounds
US5872232A (en) * 1990-01-11 1999-02-16 Isis Pharmaceuticals Inc. 2'-O-modified oligonucleotides
US5623065A (en) * 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US6399754B1 (en) * 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
US5149797A (en) * 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
AU7579991A (en) * 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
EP0541722B1 (en) * 1990-08-03 1995-12-20 Sterling Winthrop Inc. Compounds and methods for inhibiting gene expression
CA2102804A1 (en) * 1991-05-10 1992-11-11 Sudhir Agrawal 3'-end blocked oligonucleotides
US7015315B1 (en) * 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
US5965722A (en) * 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
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
US5792608A (en) * 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US20060270624A1 (en) * 1991-12-24 2006-11-30 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
JPH06511387A (en) 1991-12-24 1994-12-22 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for β-amyloid modulation
DK0618925T4 (en) 1991-12-24 2012-07-09 Isis Pharmaceuticals Inc Antisense oligonucleotides
US5856455A (en) * 1991-12-24 1999-01-05 Isis Pharmaceuticals, Inc. Gapped 2'-modified oligonucleotides
US5434257A (en) * 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
US6346614B1 (en) 1992-07-23 2002-02-12 Hybridon, Inc. Hybrid oligonucleotide phosphorothioates
TW244371B (en) * 1992-07-23 1995-04-01 Tri Clover Inc
US5523389A (en) * 1992-09-29 1996-06-04 Isis Pharmaceuticals, Inc. Inhibitors of human immunodeficiency virus
IL105244A (en) * 1993-03-31 2003-07-31 Dpharm Ltd Prodrugs with enhanced penetration into cells
US6413949B1 (en) 1995-06-07 2002-07-02 D-Pharm, Ltd. Prodrugs with enhanced penetration into cells
WO1995003427A2 (en) * 1993-07-19 1995-02-02 Gen-Probe Incorporated Oligonucleotide screening assay
DE69433036T2 (en) 1993-09-03 2004-05-27 Isis Pharmaceuticals, Inc., Carlsbad AMINODERIVATIZED NUCLEOSIDES AND OLIGONUCLEOSIDES
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US6313106B1 (en) 1995-06-07 2001-11-06 D-Pharm Ltd. Phospholipid derivatives of valproic acid and mixtures thereof
GB9518864D0 (en) 1995-09-14 1995-11-15 Cambridge Res & Innovation Oligonucleotides and their uses
US5998203A (en) * 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
US7070962B1 (en) 1996-02-01 2006-07-04 Aventis Pharmaceuticals Inc. Positive controls in polynucleotide amplification
AU3230797A (en) 1996-06-05 1998-01-05 Chiron Corporation Dna encoding dp. 75 and a process for its use
US20050119470A1 (en) * 1996-06-06 2005-06-02 Muthiah Manoharan Conjugated oligomeric compounds and their use in gene modulation
US20040161777A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Modified oligonucleotides for use in RNA interference
US20040171031A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Sugar 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
US9096636B2 (en) * 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US20040147022A1 (en) * 1996-06-06 2004-07-29 Baker Brenda F. 2'-methoxy substituted oligomeric compounds and compositions for use in gene modulations
US20040203024A1 (en) * 1996-06-06 2004-10-14 Baker Brenda F. Modified oligonucleotides for use in RNA interference
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US7812149B2 (en) * 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US20050053976A1 (en) * 1996-06-06 2005-03-10 Baker Brenda F. Chimeric oligomeric compounds and their use in gene modulation
US5849902A (en) * 1996-09-26 1998-12-15 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US5989912A (en) 1996-11-21 1999-11-23 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US5885834A (en) * 1996-09-30 1999-03-23 Epstein; Paul M. Antisense oligodeoxynucleotide against phosphodiesterase
AU731909B2 (en) 1997-07-01 2001-04-05 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
AU8512598A (en) * 1997-07-25 1999-02-16 Hybridon, Inc. Oligonuclotides having 3' terminal stereospecific phosphorothioates
US7090975B2 (en) 1998-03-13 2006-08-15 Promega Corporation Pyrophosphorolysis and incorporation of nucleotide method for nucleic acid detection
US7321828B2 (en) 1998-04-13 2008-01-22 Isis Pharmaceuticals, Inc. System of components for preparing oligonucleotides
US20040186071A1 (en) * 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
EP1071753A2 (en) 1998-04-20 2001-01-31 Ribozyme Pharmaceuticals, Inc. Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
AU745880B2 (en) * 1998-05-21 2002-04-11 Isis Pharmaceuticals, Inc. Compositions and methods for non-parenteral delivery of oligonucleotides
JP2002515514A (en) * 1998-05-21 2002-05-28 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for local delivery of oligonucleotides
CA2344653A1 (en) * 1998-09-29 2000-04-06 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
AU775997B2 (en) * 1998-12-02 2004-08-19 Bristol-Myers Squibb Company DNA-protein fusions and uses thereof
US6358712B1 (en) 1999-01-05 2002-03-19 Trustee Of Boston University Ordered gene assembly
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
EP1169480A4 (en) * 1999-04-14 2005-02-02 Musc Found For Res Dev Tissue-specific and pathogen-specific toxic agents and ribozymes
JP3513068B2 (en) * 1999-05-12 2004-03-31 浜松ホトニクス株式会社 Method for detecting target nucleic acid in cytoplasm of living cell
AU4739600A (en) * 1999-05-17 2000-12-05 Mcgill University Method for subtracting cdnas by suppressing the synthesis of specifically targeted mrnas
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US7668658B2 (en) * 1999-10-13 2010-02-23 Sequenom, Inc. 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
US20030176385A1 (en) * 2000-02-15 2003-09-18 Jingfang Ju Antisense modulation of protein expression
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
GB0021303D0 (en) * 2000-08-30 2000-10-18 Medinnova Sf Use
WO2002059337A1 (en) * 2001-01-26 2002-08-01 Georgetown University School Of Medicine Anti-apoptopic gene scc-s2 and diagnostic and therapeutic uses thereof
AU2002303262A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene shinc-1 and diagnostic and therapeutic uses thereof
WO2002081639A2 (en) * 2001-04-06 2002-10-17 Georgetown University Gene brcc2 and diagnostic and therapeutic uses thereof
WO2002081642A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc-3 and diagnostic and therapeutic uses thereof
AU2002305151A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene scc-112 and diagnostic and therapeutic uses thereof
US7803915B2 (en) * 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
DK2000545T3 (en) 2001-06-20 2011-11-28 Genentech Inc Compositions and methods for diagnosis and treatment of lung tumor
US20050107595A1 (en) * 2001-06-20 2005-05-19 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2451643C (en) 2001-06-21 2012-11-13 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
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
EP2174953A1 (en) 2001-09-18 2010-04-14 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
NZ566396A (en) 2001-10-09 2009-07-31 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expressions
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
US20030224380A1 (en) * 2001-10-25 2003-12-04 The General Hospital Corporation Genes and polymorphisms on chromosome 10 associated with Alzheimer's disease and other neurodegenerative diseases
US20030170678A1 (en) * 2001-10-25 2003-09-11 Neurogenetics, Inc. Genetic markers for Alzheimer's disease and methods using the same
WO2003054143A2 (en) * 2001-10-25 2003-07-03 Neurogenetics, Inc. Genes and polymorphisms on chromosome 10 associated with alzheimer's disease and other neurodegenerative diseases
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
AU2002367318B2 (en) 2002-01-02 2007-07-12 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
US7244565B2 (en) * 2002-04-10 2007-07-17 Georgetown University Gene shinc-3 and diagnostic and therapeutic uses thereof
US7138512B2 (en) * 2002-04-10 2006-11-21 Georgetown University Gene SHINC-2 and diagnostic and therapeutic uses thereof
JP2005536190A (en) 2002-04-16 2005-12-02 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
AU2003228809A1 (en) * 2002-05-03 2003-11-17 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof, and related methods
US20030228317A1 (en) * 2002-05-22 2003-12-11 Prafulla Gokhale Gene BRCC-1 and diagnostic and therapeutic uses thereof
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
US20050221326A1 (en) * 2002-06-12 2005-10-06 Avi Orr-Urtreger Oligonucleotides antibodies and kits including same for treating prostate cancer and determining predisposition thereto
AU2003256325A1 (en) * 2002-06-26 2004-01-19 The Penn State Research Foundation Methods and materials for treating human papillomavirus infections
US6889143B2 (en) * 2002-09-12 2005-05-03 Intergrated Dna Technologies, Inc. Methods and systems for estimating the melting temperature (TM) for polynucleotide molecules
US7229976B2 (en) 2002-09-26 2007-06-12 Isis Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
EP1560597A4 (en) * 2002-10-29 2007-06-27 Pharmacia Corp Differentially expressed genes involved in cancer, the polypeptides encoded thereby, and methods of using the same
AU2003287505A1 (en) 2002-11-05 2004-06-03 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
AU2003290598A1 (en) * 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
ES2420914T3 (en) 2002-11-13 2013-08-27 Genzyme Corporation Antisense modulation of apolipoprotein B expression
CA2505801A1 (en) 2002-11-13 2004-05-27 Rosanne Crooke Antisense modulation of apolipoprotein b expression
CA2506127C (en) 2002-11-15 2013-07-09 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US7468356B2 (en) 2003-02-11 2008-12-23 Antisense Therapeutics Ltd. Modulation of insulin like growth factor I receptor expression
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20070141570A1 (en) * 2003-03-07 2007-06-21 Sequenom, Inc. Association of polymorphic kinase anchor proteins with cardiac phenotypes and related methods
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
US20040198640A1 (en) * 2003-04-02 2004-10-07 Dharmacon, Inc. Stabilized polynucleotides for use in RNA interference
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
WO2005002507A2 (en) 2003-06-03 2005-01-13 Isis Pharmaceuticals, Inc. Modulation of survivin expression
JP2007502868A (en) 2003-06-13 2007-02-15 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Vaccines, immunotherapeutic agents and methods of use
US7790691B2 (en) * 2003-06-20 2010-09-07 Isis Pharmaceuticals, Inc. Double stranded compositions comprising a 3′-endo modified strand for use in gene modulation
EP1636342A4 (en) * 2003-06-20 2008-10-08 Isis Pharmaceuticals Inc Oligomeric compounds for use in gene modulation
US7173015B2 (en) 2003-07-03 2007-02-06 The Trustees Of The University Of Pennsylvania Inhibition of Syk kinase expression
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, 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
NZ545134A (en) 2003-09-18 2009-06-26 Lilly Co Eli Modulation of eIF4E expression
AU2004274021B2 (en) * 2003-09-18 2009-08-13 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
EP1678194B1 (en) 2003-10-10 2013-06-26 Alchemia Oncology Pty Limited The modulation of hyaluronan synthesis and degradation in the treatment of disease
CN103495153A (en) 2003-10-23 2014-01-08 奇尼塔二有限责任公司 Method for detecting mutations in a gene associated with resistance to viral infection, OAS1
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
ES2493016T3 (en) 2003-11-17 2014-09-11 Genentech, Inc. Compositions comprising antibodies against CD79b conjugated to a growth inhibitory agent or a cytotoxic agent and methods for the treatment of tumors of hematopoietic origin
US20050120397A1 (en) * 2003-11-24 2005-06-02 Hermann Steller Compounds and methods for regulation of spermatid differentiation
US7695932B2 (en) * 2003-12-24 2010-04-13 Massachusetts Institute Of Technology Gene targets for enhanced carotenoid production
US7741070B2 (en) * 2003-12-24 2010-06-22 Massachusetts Institute Of Technology Gene targets for enhanced carotenoid production
US7608699B2 (en) * 2003-12-29 2009-10-27 Rockefeller University Synthetic nuclear localization signal derived from lentiviral integrase and methods of use thereof
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
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US20090280567A1 (en) * 2004-02-06 2009-11-12 Dharmacon, Inc. Stabilized sirnas as transfection controls and silencing reagents
ATE447024T1 (en) * 2004-02-06 2009-11-15 Dharmacon Inc STABILIZED RNAS AS TRANSFECTION CONTROLS AND SILENCING REAGENTS
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
US8790919B2 (en) * 2004-03-15 2014-07-29 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNase H
KR101147147B1 (en) * 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Modified polynucleotides for reducing off-target effects in rna interference
US20050244869A1 (en) * 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
EP2540734B1 (en) 2004-04-05 2016-03-30 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
AU2004320622B2 (en) * 2004-06-03 2012-06-14 Isis Pharmaceuticals, Inc. Chimeric gapped oligomeric compositions
US8394947B2 (en) * 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US8680062B2 (en) 2004-07-06 2014-03-25 Deliversir Ltd. System for delivering therapeutic agents into living cells and cells nuclei
PL2409713T3 (en) 2004-08-10 2016-02-29 Kastle Therapeutics Llc Oligonucleotides for use in modulating lipoprotein and cholesterol levels in humans
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
EP1799859B1 (en) 2004-09-17 2014-07-02 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
NZ554277A (en) 2004-10-20 2010-09-30 Antisense Therapeutics Ltd Antisense modulation of integrin alpha 4 expression
US20060166234A1 (en) * 2004-11-22 2006-07-27 Barbara Robertson Apparatus and system having dry control gene silencing compositions
US7923207B2 (en) 2004-11-22 2011-04-12 Dharmacon, Inc. Apparatus and system having dry gene silencing pools
US7935811B2 (en) * 2004-11-22 2011-05-03 Dharmacon, Inc. Apparatus and system having dry gene silencing compositions
AU2006223498A1 (en) 2005-03-10 2006-09-21 Genentech, Inc. Methods and compositions for modulating vascular integrity
US20060223777A1 (en) * 2005-03-29 2006-10-05 Dharmacon, Inc. Highly functional short hairpin RNA
EP1879626A4 (en) * 2005-04-19 2011-03-23 Massachusetts Inst Technology Amphiphilic polymers and methods of use thereof
US8097597B2 (en) 2005-05-05 2012-01-17 Antisense Pharma Gmbh Use of low doses of oligonucleotides antisense to TGF-β genes in the treatment of brain tumors
JP5329949B2 (en) 2005-05-31 2013-10-30 エコーレ ポリテクニーク フェデラーレ デ ローザンヌ Triblock copolymers for cytoplasmic delivery of gene-based drugs
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
EP1966377A2 (en) 2005-11-21 2008-09-10 Isis Pharmaceuticals, Inc. Modulation of eif4e-bp2 expression
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
US8288354B2 (en) 2005-12-28 2012-10-16 The Scripps Research Institute Natural antisense and non-coding RNA transcripts as drug targets
US7951934B2 (en) 2006-01-26 2011-05-31 Isis Pharmaceuticals, Inc. Compositions and their uses directed to huntingtin
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
JP5342881B2 (en) 2006-01-27 2013-11-13 アイシス ファーマシューティカルズ, インコーポレーテッド 6-modified bicyclic nucleic acid analogues
JP5213723B2 (en) 2006-01-27 2013-06-19 アイシス ファーマシューティカルズ, インコーポレーテッド Oligomer compounds and compositions for use in modulating microRNA
WO2007134014A2 (en) * 2006-05-05 2007-11-22 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of gcgr
ES2389737T3 (en) * 2006-05-11 2012-10-31 Isis Pharmaceuticals, Inc. 5 'modified bicyclic nucleic acid analogs
US7666854B2 (en) * 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
WO2007137301A2 (en) * 2006-05-23 2007-11-29 Isis Pharmaceuticals, Inc. Modulation of chrebp expression
US8198253B2 (en) 2006-07-19 2012-06-12 Isis Pharmaceuticals, Inc. Compositions and their uses directed to HBXIP
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
US9155749B2 (en) 2006-09-14 2015-10-13 Medgenics Medical Israel Ltd. Long lasting drug formulations
WO2008036825A2 (en) * 2006-09-22 2008-03-27 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by rna interference
EP1935428A1 (en) * 2006-12-22 2008-06-25 Antisense Pharma GmbH Oligonucleotide-polymer conjugates
CA2691066C (en) 2007-02-09 2018-07-31 Northwestern University Particles for detecting intracellular targets
EP2144633B1 (en) * 2007-04-23 2014-07-16 Deliversir Ltd A system for delivering therapeutic agents into living cells and cells nuclei
US9156865B2 (en) 2007-04-23 2015-10-13 Deliversir Ltd System for delivering therapeutic agents into living cells and cells nuclei
US9556210B2 (en) 2007-04-23 2017-01-31 Sabag-Rfa Ltd. System for delivering therapeutic agents into living cells and cells nuclei
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
EP2826863B1 (en) 2007-05-30 2017-08-23 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
WO2008154401A2 (en) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
US8278283B2 (en) * 2007-07-05 2012-10-02 Isis Pharmaceuticals, Inc. 6-disubstituted or unsaturated bicyclic nucleic acid analogs
AU2008286771B2 (en) 2007-08-15 2013-08-15 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
WO2009039442A1 (en) * 2007-09-21 2009-03-26 California Institute Of Technology Nfia in glial fate determination, glioma therapy and astrocytoma treatment
WO2009045536A2 (en) * 2007-10-05 2009-04-09 The University Of North Carolina At Chapel Hill Receptor targeted oligonucleotides
US8637478B2 (en) * 2007-11-13 2014-01-28 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
USRE47320E1 (en) 2007-11-20 2019-03-26 Ionis Pharmaceuticals, Inc. Modulation of CD40 expression
WO2009067647A1 (en) * 2007-11-21 2009-05-28 Isis Pharmaceuticals, Inc. Carbocyclic alpha-l-bicyclic nucleic acid analogs
US7845686B2 (en) * 2007-12-17 2010-12-07 S & B Technical Products, Inc. Restrained pipe joining system for plastic pipe
EP2265627A2 (en) * 2008-02-07 2010-12-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
US8188060B2 (en) 2008-02-11 2012-05-29 Dharmacon, Inc. Duplex oligonucleotides with enhanced functionality in gene regulation
WO2009117589A1 (en) * 2008-03-21 2009-09-24 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucleosides and methods for their use
EP2285819B1 (en) * 2008-04-04 2013-10-16 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
US8082730B2 (en) * 2008-05-20 2011-12-27 Caterpillar Inc. Engine system having particulate reduction device and method
WO2010017248A2 (en) 2008-08-04 2010-02-11 University Of Miami Sting (stimulator of interferon genes), a regulator of innate immune responses
US20110237646A1 (en) * 2008-08-07 2011-09-29 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression for the treatment of cns related disorders
EP3081648A1 (en) 2008-08-25 2016-10-19 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
US10138485B2 (en) 2008-09-22 2018-11-27 Rxi Pharmaceuticals Corporation Neutral nanotransporters
US8604192B2 (en) * 2008-09-24 2013-12-10 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acids analogs
US8501805B2 (en) * 2008-09-24 2013-08-06 Isis Pharmaceuticals, Inc. Substituted alpha-L-bicyclic nucleosides
AU2009305636A1 (en) 2008-10-15 2010-04-22 Ionis Pharmaceuticals, Inc. Modulation of Factor 11 expression
AU2009308217B2 (en) 2008-10-24 2016-01-21 Ionis Pharmaceuticals, Inc. 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
WO2010048585A2 (en) 2008-10-24 2010-04-29 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
CA2744207C (en) 2008-11-24 2019-05-28 Northwestern University Polyvalent rna-nanoparticle compositions
CN102317458B (en) 2008-12-04 2018-01-02 库尔纳公司 Pass through treatment of the suppression of erythropoietin(EPO) (EPO) natural antisense transcript to EPO relevant diseases
CA2745811C (en) 2008-12-04 2021-07-13 Joseph Collard Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
KR101749352B1 (en) 2008-12-04 2017-06-20 큐알엔에이, 인크. Treatment of sirtuin 1(sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
US8592386B2 (en) 2008-12-17 2013-11-26 Sarepta Therapeutics, Inc. Antisense compositions and methods for modulating contact hypersensitivity or contact dermatitis
US20100233270A1 (en) * 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
US20100184844A1 (en) * 2009-01-08 2010-07-22 Northwestern University Inhibition of Bacterial Protein Production by Polyvalent Oligonucleotide Modified Nanoparticle Conjugates
KR101546673B1 (en) * 2009-01-15 2015-08-25 삼성전자주식회사 Toner for electrophotographic and process for preparing the same
WO2010091308A2 (en) 2009-02-06 2010-08-12 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
US8536320B2 (en) 2009-02-06 2013-09-17 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
KR101805199B1 (en) 2009-02-12 2017-12-05 큐알엔에이, 인크. Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
EP3009150B1 (en) 2009-02-12 2019-11-13 CuRNA, Inc. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US20110319317A1 (en) 2009-03-04 2011-12-29 Opko Curna, Llc Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirt1
WO2010107733A2 (en) 2009-03-16 2010-09-23 Curna, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases 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
EP3248618A1 (en) 2009-04-22 2017-11-29 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
ES2661787T3 (en) 2009-05-01 2018-04-04 Curna, Inc. Treatment of hemoglobin-related diseases (hbf / hbg) by inhibition of natural antisense transcript for hbf / hbg
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
CN102575251B (en) 2009-05-18 2018-12-04 库尔纳公司 The relevant disease of the reprogramming factor is treated by inhibiting the natural antisense transcript for the reprogramming factor
KR101703695B1 (en) 2009-05-22 2017-02-08 큐알엔에이, 인크. Treatment of transcription factor e3 (tfe3) and insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to tfe3
EP2435571B1 (en) 2009-05-28 2016-12-14 CuRNA, Inc. Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
JP5944311B2 (en) 2009-06-16 2016-07-05 クルナ・インコーポレーテッド Treatment of collagen gene-related diseases by suppression of natural antisense transcripts against collagen genes
EP2443238B1 (en) 2009-06-16 2017-03-22 CuRNA, Inc. Treatment of paraoxonase 1 (pon1) related diseases by inhibition of natural antisense transcript to 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)
WO2010151674A2 (en) 2009-06-26 2010-12-29 Curna, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
JP6128848B2 (en) 2009-08-05 2017-05-17 クルナ・インコーポレーテッド Treatment of insulin gene (INS) -related diseases by suppression of natural antisense transcripts against the insulin gene (INS)
WO2011017521A2 (en) 2009-08-06 2011-02-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
WO2011031482A2 (en) 2009-08-25 2011-03-17 Curna, Inc. Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
CA2772715C (en) 2009-09-02 2019-03-26 Genentech, Inc. Mutant smoothened and methods of using the same
US8664377B2 (en) * 2009-09-30 2014-03-04 Samsung Electronics Co., Ltd. Method and apparatus for isolating nucleic acids
CN102711826B (en) 2009-10-22 2017-03-29 霍夫曼-拉罗奇有限公司 For the method and composition that the HEPSIN for regulating and controlling macrophage-stimulating albumen is activated
CA2779099C (en) 2009-10-30 2021-08-10 Northwestern University Templated nanoconjugates
BR112012011381B8 (en) 2009-11-13 2021-05-25 Avi Biopharma Inc Isolated antiviral antisense oligonucleotide and pharmaceutical composition comprising the same
CA2781887C (en) 2009-11-30 2018-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2011084455A2 (en) 2009-12-16 2011-07-14 Opko Curna, Llc. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
CN102869776B (en) 2009-12-23 2017-06-23 库尔纳公司 HGF relevant diseases are treated by suppressing the natural antisense transcript of HGF (HGF)
KR101793753B1 (en) 2009-12-23 2017-11-03 큐알엔에이, 인크. Treatment of uncoupling protein 2 (ucp2) related diseases by inhibition of natural antisense transcript to ucp2
WO2011090741A2 (en) 2009-12-29 2011-07-28 Opko Curna, Llc TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
NO2521784T3 (en) 2010-01-04 2018-05-05
US8912157B2 (en) 2010-01-06 2014-12-16 Curna, Inc. Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
DK2524039T3 (en) 2010-01-11 2018-03-12 Curna Inc TREATMENT OF GENDER HORMON-BINDING GLOBULIN (SHBG) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTS TO SHBG
US8779118B2 (en) 2010-01-11 2014-07-15 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2011091390A2 (en) 2010-01-25 2011-07-28 Opko Curna, Llc Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
WO2011103528A2 (en) 2010-02-22 2011-08-25 Opko Curna Llc Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
EP2538981B1 (en) 2010-02-23 2017-12-20 F. Hoffmann-La Roche AG Compositions and methods for the diagnosis and treatment of tumor
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
WO2011105900A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-alpha (c8-alpha) and uses thereof
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
US9068185B2 (en) 2010-03-12 2015-06-30 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
WO2011112732A2 (en) 2010-03-12 2011-09-15 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
US9506057B2 (en) 2010-03-26 2016-11-29 Integrated Dna Technologies, Inc. Modifications for antisense compounds
US9044494B2 (en) 2010-04-09 2015-06-02 Curna, Inc. Treatment of fibroblast growth factor 21 (FGF21) related diseases by inhibition of natural antisense transcript to FGF21
EP2563380B1 (en) 2010-04-26 2018-05-30 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-trna synthetase
WO2011139799A2 (en) 2010-04-27 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl trna synthetases
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
US8993723B2 (en) 2010-04-28 2015-03-31 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of alanyl-tRNA synthetases
KR101869570B1 (en) 2010-04-28 2018-06-20 아이오니스 파마수티컬즈, 인코포레이티드 Modified nucleosides and oligomeric compounds prepared therefrom
AU2011248457B2 (en) 2010-04-29 2017-02-16 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl tRNA synthetases
US8986680B2 (en) 2010-04-29 2015-03-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Asparaginyl tRNA synthetases
US9068177B2 (en) 2010-04-29 2015-06-30 Atyr Pharma, Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
WO2011139917A1 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression
EP2566515B1 (en) 2010-05-03 2017-08-02 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases
JP6008840B2 (en) 2010-05-03 2016-10-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of phenylalanyl αtRNA synthetase
US8981045B2 (en) 2010-05-03 2015-03-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-tRNA synthetases
MA34291B1 (en) 2010-05-03 2013-06-01 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING A TUMOR
EP2566966A4 (en) 2010-05-03 2013-12-11 Curna Inc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CA2798139C (en) 2010-05-04 2019-09-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-trna synthetase complex
CN103429739B (en) 2010-05-12 2018-11-13 哥伦比亚大学纽约管理委员会 Prepare the method generated with the enteroendocrine cell of excreting insulin
CA3090304A1 (en) 2010-05-13 2011-11-17 Sarepta Therapeutics, Inc. Antisense modulation of interleukins 17 and 23 signaling
EP2568996B1 (en) 2010-05-14 2017-10-04 aTyr Pharma, Inc. Therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-trna synthetases
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
JP6027965B2 (en) 2010-05-17 2016-11-16 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of leucyl-tRNA synthetase
CA2799207C (en) 2010-05-26 2019-03-26 Curna, Inc. Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
WO2011156278A1 (en) 2010-06-07 2011-12-15 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
EP2580228B1 (en) 2010-06-08 2016-03-23 Ionis Pharmaceuticals, Inc. Substituted 2'-amino and 2'-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2011159758A2 (en) 2010-06-15 2011-12-22 Medgenics Medical Israel Ltd. Long lasting drug formulations
JP6116479B2 (en) 2010-07-12 2017-04-19 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of glycyl-tRNA synthetase
US8980860B2 (en) 2010-07-14 2015-03-17 Curna, Inc. Treatment of discs large homolog (DLG) related diseases by inhibition of natural antisense transcript to DLG
EP2608801B1 (en) 2010-08-25 2019-08-21 aTyr Pharma, Inc. INNOVATIVE DISCOVERY OF THERAPEUTIC, DIAGNOSTIC, AND ANTIBODY COMPOSITIONS RELATED TO PROTEIN FRAGMENTS OF TYROSYL-tRNA SYNTHETASES
WO2012033848A1 (en) 2010-09-07 2012-03-15 Integrated Dna Technologies, Inc. Modifications for antisense compounds
US8481680B2 (en) 2010-10-05 2013-07-09 Genentech, Inc. Mutant smoothened and methods of using the same
US8993533B2 (en) 2010-10-06 2015-03-31 Curna, Inc. Treatment of sialidase 4 (NEU4) related diseases by inhibition of natural antisense transcript to NEU4
US20140031250A1 (en) 2010-10-07 2014-01-30 David Tsai Ting Biomarkers of Cancer
CA2815212A1 (en) 2010-10-22 2012-04-26 Curna, Inc. Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
CN103201387B (en) 2010-10-27 2018-02-02 库尔纳公司 IFRD1 relevant diseases are treated by suppressing the natural antisense transcript of interferon correlative development regulatory factor 1 (IFRD1)
EP3260540A1 (en) 2010-11-12 2017-12-27 The General Hospital Corporation Polycomb-associated non-coding rnas
EP2453017A1 (en) 2010-11-12 2012-05-16 Antisense Pharma GmbH Oligonucleotides for use in prophylaxis and/or treatment of TGF-beta1 and TGF-beta2, TGF-beta2 and TGF-beta3, TGF-beta1 and TGF-beta3, or TGF-beta1, TGF-beta2, and TGF-beta3 mRNA overexpressing diseases
US9920317B2 (en) 2010-11-12 2018-03-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
EP2640853B1 (en) 2010-11-17 2018-12-26 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
WO2012071238A2 (en) 2010-11-23 2012-05-31 Opko Curna Llc Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
WO2012097261A2 (en) 2011-01-14 2012-07-19 The General Hospital Corporation Methods targeting mir-128 for regulating cholesterol/lipid metabolism
EP2668199B1 (en) 2011-01-27 2017-09-06 Ramot at Tel Aviv University, Ltd. Glycogen synthase kinase-3 inhibitors
US9688719B2 (en) 2011-01-27 2017-06-27 Ramot At Tel-Aviv University Ltd. Glycogen synthase kinase-3 inhibitors
EP2670404B1 (en) 2011-02-02 2018-08-29 The Trustees of Princeton University Sirtuin modulators as virus production modulators
SI2670411T1 (en) 2011-02-02 2019-06-28 Excaliard Pharmaceuticals, Inc. Antisense compounds targeting connective tissue growth factor (ctgf) for use in a method of treating keloids or hypertrophic scars
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
RU2620980C2 (en) 2011-06-09 2017-05-30 Курна, Инк. Treatment of diseases associated with frataxin (fxn), by inhibiting natural antisense fxn transcript
EP2756080B1 (en) 2011-09-14 2019-02-20 Translate Bio MA, Inc. Multimeric oligonucleotide compounds
EP2755692B1 (en) 2011-09-14 2020-11-25 Northwestern University Nanoconjugates able to cross the blood-brain barrier
EP2766482B1 (en) 2011-10-11 2016-12-07 The Brigham and Women's Hospital, Inc. Micrornas in neurodegenerative disorders
EP2790736B1 (en) 2011-12-12 2018-01-31 Oncoimmunin, Inc. In vivo delivery of oligonucleotides
JP6348848B2 (en) 2012-02-13 2018-06-27 ガミダ セル リミテッド Proliferation of mesenchymal stem cells
JP2015511494A (en) 2012-03-15 2015-04-20 キュアナ,インク. Treatment of BDNF-related diseases by inhibition of natural antisense transcripts against brain-derived neurotrophic factor (BDNF)
WO2013138662A1 (en) 2012-03-16 2013-09-19 4S3 Bioscience, Inc. Antisense conjugates for decreasing expression of dmpk
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
EP2850092B1 (en) 2012-04-09 2017-03-01 Ionis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
JP2015518485A (en) 2012-04-20 2015-07-02 アプタミアール セラピューティクス インコーポレイテッド Thermogenic miRNA regulator
EP2841572B1 (en) 2012-04-27 2019-06-19 Duke University Genetic correction of mutated genes
WO2013173599A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating hemoglobin gene family expression
AU2013262663A1 (en) 2012-05-16 2015-01-22 The General Hospital Corporation D/B/A Massachusetts General Hospital Compositions and methods for modulating gene expression
KR20160074368A (en) 2012-05-16 2016-06-28 라나 테라퓨틱스, 인크. Compositions and methods for modulating utrn expression
CA2873766A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics Inc. Compositions and methods for modulating atp2a2 expression
WO2013173608A1 (en) 2012-05-16 2013-11-21 Rana Therapeutics, Inc. Compositions and methods for modulating mecp2 expression
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
EA201492123A1 (en) 2012-05-16 2015-10-30 Рана Терапьютикс, Инк. COMPOSITIONS AND METHODS FOR MODULATING THE EXPRESSION OF THE SMN GENES FAMILY
WO2013184209A1 (en) 2012-06-04 2013-12-12 Ludwig Institute For Cancer Research Ltd. Mif for use in methods of treating subjects with a neurodegenerative disorder
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
WO2014022852A1 (en) 2012-08-03 2014-02-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
CN104736551B (en) 2012-08-15 2017-07-28 Ionis制药公司 The method for preparing oligomeric compounds using improved end-blocking scheme
AU2013315225B2 (en) 2012-09-14 2018-11-08 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
AU2013336237A1 (en) 2012-10-22 2015-06-11 Sabag-Rfa Ltd A system for delivering therapeutic agents into living cells and cells nuclei
WO2014130945A1 (en) 2013-02-25 2014-08-28 Integrated Dna Technologies, Inc. Modifications for antisense compounds
SG10201906382QA (en) 2013-05-01 2019-08-27 Ionis Pharmaceuticals Inc Compositions and methods for modulating hbv and ttr expression
AU2014280847B2 (en) 2013-06-13 2019-07-04 Antisense Therapeutics Ltd Combination therapy
WO2014207743A2 (en) 2013-06-24 2014-12-31 Ramot At Tel-Aviv University Ltd. Glycogen synthase kinase-3 inhibitors
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
CA3228845A1 (en) 2014-01-31 2015-08-06 Temple University Of The Commonwealth System Of Higher Education Bag3 and uses thereof in the treatment of heart failure with reduced ejection fraction
EP3102197B1 (en) 2014-02-04 2018-08-29 Genentech, Inc. Mutant smoothened and methods of using the same
EP3119789B1 (en) 2014-03-17 2020-04-22 Ionis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
US10006027B2 (en) 2014-03-19 2018-06-26 Ionis Pharmaceuticals, Inc. Methods for modulating Ataxin 2 expression
RU2019130898A (en) 2014-03-19 2019-11-11 Ионис Фармасьютикалз, Инк. COMPOSITIONS FOR MODULATION OF ATAXIN 2 EXPRESSION
RU2704619C2 (en) 2014-04-01 2019-10-30 Биоген Ма Инк. Compositions for modulating expression of sod-1
US10221416B2 (en) 2014-04-24 2019-03-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
SG11201608502TA (en) 2014-05-01 2016-11-29 Ionis Pharmaceuticals Inc Compositions and methods for modulating complement factor b expression
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
EP3161159B1 (en) 2014-06-25 2020-08-05 The General Hospital Corporation Targeting human satellite ii (hsatii)
EP3160503B1 (en) 2014-06-26 2021-02-17 The Trustees of Columbia University in the City of New York Inhibition of serotonin expression in gut enteroendocrine cells results in conversion to insulin-positive cells
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
EP3198012B1 (en) 2014-09-26 2019-09-04 University of Massachusetts Rna-modulating agents
WO2016070060A1 (en) 2014-10-30 2016-05-06 The General Hospital Corporation Methods for modulating atrx-dependent gene repression
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
WO2016115490A1 (en) 2015-01-16 2016-07-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of dux4
EP3256487A4 (en) 2015-02-09 2018-07-18 Duke University Compositions and methods for epigenome editing
EP3256591A4 (en) 2015-02-13 2018-08-08 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
US10900036B2 (en) 2015-03-17 2021-01-26 The General Hospital Corporation RNA interactome of polycomb repressive complex 1 (PRC1)
WO2016164463A1 (en) 2015-04-07 2016-10-13 The General Hospital Corporation Methods for reactivating genes on the inactive x chromosome
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
CN108139375A (en) 2015-06-26 2018-06-08 贝斯以色列女执事医疗中心股份有限公司 Target the cancer therapy for inhibiting the four transmembrane proteins 33 (TSPAN33) in cell derived from marrow sample
EP3314027A4 (en) 2015-06-29 2019-07-03 Caris Science, Inc. Therapeutic oligonucleotides
CA2993652A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
WO2017053781A1 (en) 2015-09-25 2017-03-30 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
EP4089175A1 (en) 2015-10-13 2022-11-16 Duke University Genome engineering with type i crispr systems in eukaryotic cells
WO2017075670A1 (en) 2015-11-05 2017-05-11 Children's Hospital Los Angeles "mobilizing leukemia cells"
AU2016355178B9 (en) 2015-11-19 2019-05-30 Massachusetts Institute Of Technology Lymphocyte antigen CD5-like (CD5L)-interleukin 12B (p40) heterodimers in immunity
WO2017096395A1 (en) 2015-12-04 2017-06-08 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
AU2015416656B2 (en) 2015-12-07 2023-02-23 Erasmus University Medical Center Rotterdam Enzymatic replacement therapy and antisense therapy for Pompe disease
AU2017205462A1 (en) 2016-01-05 2018-06-07 Ionis Pharmaceuticals, Inc. Methods for reducing LRRK2 expression
AU2017213826A1 (en) 2016-02-04 2018-08-23 Curis, Inc. Mutant smoothened and methods of using the same
JP7033072B2 (en) 2016-02-25 2022-03-09 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド Treatment for fibrosis targeting SMOC2
AU2017234678A1 (en) 2016-03-16 2018-08-16 Ionis Pharmaceuticals, Inc. Methods of modulating KEAP1
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
JP2019516393A (en) 2016-03-18 2019-06-20 カリス サイエンス インコーポレイテッド Oligonucleotide probes and uses thereof
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
EP3471781A4 (en) 2016-06-17 2020-05-06 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
NL2017294B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Natural cryptic exon removal by pairs of antisense oligonucleotides.
NL2017295B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Antisense oligomeric compound for Pompe disease
WO2018067900A1 (en) 2016-10-06 2018-04-12 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
US11033570B2 (en) 2016-12-02 2021-06-15 Cold Spring Harbor Laboratory Modulation of Lnc05 expression
US11180756B2 (en) 2017-03-09 2021-11-23 Ionis Pharmaceuticals Morpholino modified oligomeric compounds
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
EP3612232A1 (en) 2017-04-21 2020-02-26 The Broad Institute, Inc. Targeted delivery to beta cells
WO2018209270A1 (en) 2017-05-11 2018-11-15 Northwestern University Adoptive cell therapy using spherical nucleic acids (snas)
WO2019036613A1 (en) 2017-08-18 2019-02-21 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
US10517889B2 (en) 2017-09-08 2019-12-31 Ionis Pharmaceuticals, Inc. Modulators of SMAD7 expression
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
MX2020007369A (en) 2018-01-15 2020-10-28 Ionis Pharmaceuticals Inc Modulators of dnm2 expression.
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
US11732260B2 (en) 2018-03-02 2023-08-22 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-β precursor protein
US11661601B2 (en) 2018-03-22 2023-05-30 Ionis Pharmaceuticals, Inc. Methods for modulating FMR1 expression
EP4051799A2 (en) 2018-03-30 2022-09-07 Rheinische Friedrich-Wilhelms-Universität Bonn Aptamers for targeted activaton of t cell-mediated immunity
CN116536272A (en) 2018-04-06 2023-08-04 儿童医疗中心有限公司 Compositions and methods for somatic reprogramming and imprinting
SG11202008660TA (en) 2018-04-11 2020-10-29 Ionis Pharmaceuticals Inc Modulators of ezh2 expression
CR20200604A (en) 2018-05-09 2021-02-09 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
BR112020020957B1 (en) 2018-05-09 2022-05-10 Ionis Pharmaceuticals, Inc Oligomeric compounds, population and pharmaceutical composition thereof and their uses
US11833168B2 (en) 2018-06-14 2023-12-05 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing STMN2 expression
US11332746B1 (en) 2018-06-27 2022-05-17 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing LRRK2 expression
KR20210044771A (en) 2018-07-02 2021-04-23 압타미르 테라퓨틱스, 인코포레이티드 Targeted delivery of therapeutic agents to human adipocytes
MX2021000922A (en) 2018-07-25 2021-03-31 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn2 expression.
US20210292766A1 (en) 2018-08-29 2021-09-23 University Of Massachusetts Inhibition of Protein Kinases to Treat Friedreich Ataxia
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
BR112021008967A2 (en) 2018-11-15 2021-08-17 Ionis Pharmaceuticals, Inc. irf5 expression modulators
JP2022519532A (en) 2019-01-31 2022-03-24 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 Modulator of YAP1 expression
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
AU2020227824A1 (en) 2019-02-27 2021-08-26 Ionis Pharmaceuticals, Inc. Modulators of MALAT1 expression
WO2020205463A1 (en) 2019-03-29 2020-10-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating ube3a-ats
EP3956450A4 (en) 2019-07-26 2022-11-16 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
EP4013767A4 (en) 2019-08-15 2023-10-25 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
TW202140787A (en) 2020-02-28 2021-11-01 美商Ionis製藥公司 Compounds and methods for modulating smn2
EP4143321A2 (en) 2020-05-01 2023-03-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating atxn1
JP2023532518A (en) 2020-06-29 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド Compounds and methods for modulating PLP1
US11447521B2 (en) 2020-11-18 2022-09-20 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
CN117500815A (en) 2021-06-18 2024-02-02 Ionis制药公司 Compounds and methods for reducing IFNAR1 expression
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024044352A1 (en) 2022-08-26 2024-02-29 The General Hospital Corporation Methods and compositions for prognosis and treatment of dilated cardiomyopathy and heart failure

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3818109A (en) * 1971-03-19 1974-06-18 Univ Kansas State Conversion of whey solids to an edible yeast cell mass
US3968257A (en) * 1972-09-06 1976-07-06 Mueller Hans Processes for the production of protein-containing nutriments and fodders
US4310662A (en) 1979-12-26 1982-01-12 Genentech, Inc. Nucleosidic phosphorylating agent and methods
US4307189A (en) 1980-03-21 1981-12-22 Malon Kit Method for the quantitative determination of terminal deoxynucleotidyl transferase in biological samples
US4511713A (en) * 1980-11-12 1985-04-16 The Johns Hopkins University Process for selectively controlling unwanted expression or function of foreign nucleic acids in animal or mammalian cells
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) * 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
JP2547714B2 (en) * 1981-10-23 1996-10-23 モルキユラ− バイオシステムズ インコ−ポレテツド Oligonucleotide therapeutic agent and method for producing the same
US4591564A (en) 1982-11-10 1986-05-27 The University Of Montana Transferase enzymes which modify the 3'-termini of ribonucleic acid and methods
US4521509A (en) * 1982-11-24 1985-06-04 Research Corporation Method for degrading DNA
WO1984003285A1 (en) 1983-02-22 1984-08-30 Molecular Biosystems Inc Defined sequence single strand oligonucleotides incorporating reporter groups, process for the chemical synthesis thereof, and nucleosides useful in such synthesis
US4507433A (en) 1983-10-07 1985-03-26 The Johns Hopkins University Preparation of oligodeoxyribonucleoside alkyl or arylphosphonates
US4672110A (en) 1983-11-29 1987-06-09 Northwestern University Method of deprotecting polynucleotide trimethylethyl phosphotriesters
US4676987A (en) * 1984-04-24 1987-06-30 Stauffer Chemical Co. Production of fermented whey containing calcium propionate
US4600531A (en) 1984-06-27 1986-07-15 University Of Iowa Research Foundation Production of alpha-alpha cross-linked hemoglobins in high yield
US4598064A (en) 1984-06-27 1986-07-01 University Of Iowa Research Foundation Alpha-alpha cross-linked hemoglobins
US4698303A (en) * 1985-02-15 1987-10-06 Engenics, Inc. Production of lactic acid by continuous fermentation using an inexpensive raw material and a simplified method of lactic acid purification
FR2582653B1 (en) * 1985-05-30 1987-08-14 Centre Nat Rech Scient NEW COUPLING COMPOUNDS, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN THE DEVELOPMENT OF INTERFERON EFFECTS
GB8515276D0 (en) 1985-06-17 1985-07-17 Amersham Int Plc Nucleic acid sequencing
JPS62151155A (en) * 1985-09-17 1987-07-06 Ajinomoto Co Inc Production of seasoning using cheese whey as raw material
US4800159A (en) * 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US4806463A (en) 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
DE3788914T2 (en) 1986-09-08 1994-08-25 Ajinomoto Kk Compounds for cleaving RNA at a specific position, oligomers used in the preparation of these compounds and starting materials for the synthesis of these oligomers.
US5276019A (en) * 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5004810A (en) * 1988-09-30 1991-04-02 Schering Corporation Antiviral oligomers
IT1247995B (en) * 1991-06-06 1995-01-05 Himont Inc PROCESS FOR THE PRODUCTION OF AQUATIC LACTIC ACID SOLUTIONS PURIFIED FROM FERMENTATION BRODS.
US5585479A (en) * 1992-07-24 1996-12-17 The United States Of America As Represented By The Secretary Of The Navy Antisense oligonucleotides directed against human ELAM-I RNA

Also Published As

Publication number Publication date
EP0348458B1 (en) 1997-04-09
JPH02502516A (en) 1990-08-16
DE3855864T2 (en) 1997-09-25
US6197944B1 (en) 2001-03-06
US5491133A (en) 1996-02-13
WO1989005358A1 (en) 1989-06-15
EP0348458A1 (en) 1990-01-03
JP3019994B2 (en) 2000-03-15
US5962425A (en) 1999-10-05
ATE151467T1 (en) 1997-04-15
EP0348458A4 (en) 1992-12-09
DE3855864D1 (en) 1997-05-15

Similar Documents

Publication Publication Date Title
CA1339935C (en) Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
US5532130A (en) Methods and compositions for sequence-specific hybridization of RNA by 2'-5' oligonucleotides
US5885834A (en) Antisense oligodeoxynucleotide against phosphodiesterase
AU712680B2 (en) Three component chimeric antisense oligonucleotides
US6015886A (en) Oligonucleotide phosphate esters
JP3529135B2 (en) How to cut a specific RNA strand
JP2003012688A (en) Oligonucleotide n3'->p5' phosphoramidate: synthesis and compound: hybridization and nuclease resistance characteristic
EP0736093B1 (en) ANTISENSE NUCLEIC ACIDS FOR THE PREVENTION AND TREATMENT OF DISORDERS IN WHICH EXPRESSION OF c-erbB-2 PLAYS A ROLL
Damha et al. Antisense L/D-oligodeoxynucleotide chimeras: nuclease stability, base-pairing properties, and activity at directing ribonuclease H
JPH03190895A (en) Nucleotide modification compound
KR20010102992A (en) Non-invasive method for detecting target rna
CA2154578A1 (en) Oligonucleotide alkylphosphonates and alkylphosphonothioates
IE920981A1 (en) Single-stranded circular oligonucleotides
Miller Development of antisense and antigene oligonucleotide analogs
IL94752A (en) Formation of triple helix complexes of double stranded DNA using nucleoside oligomers method and kit for detecting double stranded DNA sequences
EP0650526A1 (en) Formation of triple helix complexes of single stranded nucleic acids using nucleoside oligomers
AU620364B2 (en) Dna and rna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
NZ244125A (en) Oligomer containing 8-modified purine base and method using these to detect specific segments of double stranded nucleic acid by forming a triple helix and method for preventing expression of the segment
Thomas et al. Capping of bcr-abl antisense oligonucleotides enhances antiproliferative activity against chronic myeloid leukemia cell lines
Tsukahara et al. Nonenzymatic sequence-specific cleavage of duplex DNA via triple-helix formation by homopyrimidine phosphorothioate oligonucleotides
Noe et al. Chemistry of antisense oligonucleotides
KR100257972B1 (en) Oligonucleotides having phosphorothioate linkages of high chiral purity
CA2331333C (en) Antisense oligonucleotide constructs based on .beta.-arabinofuranose and its analogues
JP2004187609A (en) Method for determining optimal antisense sequence
JPH09224673A (en) Pair of miniribozyme, miniribozyme-dimer, cutting-deactivation of target rna by them and medicine

Legal Events

Date Code Title Description
MKEX Expiry

Effective date: 20150630