WO2000022113A1 - ENZYMATIC SYNTHESIS OF ssDNA - Google Patents

ENZYMATIC SYNTHESIS OF ssDNA Download PDF

Info

Publication number
WO2000022113A1
WO2000022113A1 PCT/US1999/023933 US9923933W WO0022113A1 WO 2000022113 A1 WO2000022113 A1 WO 2000022113A1 US 9923933 W US9923933 W US 9923933W WO 0022113 A1 WO0022113 A1 WO 0022113A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
vector
reverse transcriptase
interest
genetic elements
Prior art date
Application number
PCT/US1999/023933
Other languages
French (fr)
Other versions
WO2000022113A9 (en
Inventor
Michael J. Skillern
Charles A. Conrad
Jonathan F. Elliston
Original Assignee
Ingene, Inc.
Cytogenix, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ingene, Inc., Cytogenix, Inc. filed Critical Ingene, Inc.
Priority to CA002346155A priority Critical patent/CA2346155A1/en
Priority to KR1020017004474A priority patent/KR20010099682A/en
Priority to AU64305/99A priority patent/AU6430599A/en
Priority to BR9914772-6A priority patent/BR9914772A/en
Priority to MXPA01003642A priority patent/MXPA01003642A/en
Priority to EP99951989A priority patent/EP1117776A1/en
Priority to IL14249099A priority patent/IL142490A0/en
Priority to JP2000576004A priority patent/JP2002527061A/en
Publication of WO2000022113A1 publication Critical patent/WO2000022113A1/en
Publication of WO2000022113A9 publication Critical patent/WO2000022113A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/15Nucleic acids forming more than 2 strands, e.g. TFOs
    • 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
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed

Definitions

  • the present invention relates to the production of single stranded DNA (ssDNA) in yeast, prokaryotic, and eukaryotic cells from a set of genetic elements delivered to the cell by a vector system.
  • the ss DNA is produced in the cell with minimal vector sequences which could interfere with the intended function of the ssDNA in the cell. So far as is known, there is no method for producing single-stranded deoxyribonucleic acid (ssDNA) species in eukaryotic cells which do not contain intervening and/or flanking vector sequences.
  • ssDNA single-stranded deoxyribonucleic acid
  • the scientific and patent literature does include the disclosure of cDNA-producing vectors (see A. Ohshima, et al., 89 Proc. Natl. Acad. Sci.
  • a set of genetic elements for delivery into a cell comprising a nucleic acid construct comprising a sequence of interest, and a primer binding site for a reverse transcriptase located in a 3' position with respect to the sequence of interest.
  • the set of genetic elements of the present invention provides an efficient system for directing the synthesis of a stable, single-stranded nucleic acid sequence, both in vivo and in vitro.
  • the single-stranded nucleic acid sequence may be used to provide a desired effect in a cell, tissue or organism. Because production of the single-stranded nucleic acid sequence of interest takes place within the cell, prior art problems arising from delivery of the single-stranded nucleic acid sequence to the cell are overcome, or at least alleviated.
  • nucleic acid construct Because of the arrangement of the nucleic acid construct, with the primer binding site in a position which is 3' to the sequence of interest, there is no limit to the size or type of sequence of interest that may be produced using the nucleic acid construct of the present invention, and the construct may be easily incorporated into a vector for delivery by any desired route to a target cell.
  • Reverse transcription may be carried out by a reverse transcriptase which is endogenous to the cell (e.g. in the case of infection by human immunodeficiency virus or simian immunodeficiency virus) or the set of genetic elements may, preferably, further comprise a reverse transcriptase gene.
  • the set of genetic elements comprises a reverse transcriptase gene
  • the reverse transcriptase gene is, preferably, polycistronically transcribable with the sequence of interest and primer binding site.
  • the reverse transcriptase gene is located on the same nucleic acid construct as the sequence of interest and primer binding site and, more preferably, the reverse transcriptase gene is located in a 5' position with respect to said sequence of interest and 3' primer binding site.
  • the reverse transcriptase gene may encode reverse transcriptase or a reverse transcriptase/RNAse H polyprotein.
  • the gene encoding reverse transcriptase/RNAse H polyprotein may suitably be derived from Moloney murine leukaemia virus, human immunodeficiency virus, or simian immunodeficiency virus.
  • the primer binding site is, preferably, specific for the reverse transcriptase encoded by the reverse transcriptase gene.
  • the primer binding site is, preferably, specific for an endogenous reverse transcriptase.
  • the primer binding site is complementary to a transfer RNA (tRNA).
  • the set of genetic elements of the present invention also comprises a promoter and, optionally, an enhancer for each of said sequence of interest and/or said reverse transcriptase gene.
  • the promoter and/or enhancer is a eukaryotic promoter and/or enhancer.
  • the promoter may be a constitutive, inducible, wide-spectrum or tissue-specific promoter.
  • the set of genetic elements of the present invention further comprises a polyadenylation tail sequence located in a 3' position with respect to the sequence of interest and 3' primer binding site.
  • the polyA tail provides stability of the mRNA transcript.
  • the sequence of interest is an antisense sequence.
  • the present invention thus, has far reaching uses in the field of antisense therapy, particularly in treating pathological conditions by regulating gene function.
  • the sequence of interest may also be an aptamer (i.e. an oligonucleotide that binds to a non-oligonucleotide target e.g. a protein).
  • an aptamer i.e. an oligonucleotide that binds to a non-oligonucleotide target e.g. a protein.
  • the present invention has far reaching therapeutic uses.
  • the nucleic acid construct is DNA.
  • the set of genetic elements according to any one of the preceding claims is incorporated into at least one vector.
  • sequence of interest and 3' primer binding site may be incorporated into a first vector, with the reverse transcriptase gene incorporated into a second vector.
  • the reverse transcriptase gene, sequence of interest and primer binding site may be incorporated into a single vector.
  • the reverse transcriptase gene is, preferably, located in a 5' position with respect to the sequence of interest and 3' primer binding site.
  • nucleic acid constructs of the present invention are such that they may be incorporated into commercially available delivery vectors for mammalian and human therapeutic purposes, and may be administered by any feasible route, depending on the target cell.
  • a vector which comprises: (a) a primer binding site and an insertion site for a sequence of interest, the primer binding site being located in a 3' position with respect to the insertion site; and
  • the reverse transcriptase gene is located in a 5' position with respect to the insertion site and 3' primer binding site.
  • a vector system which comprises a first vector, comprising an insertion site for a sequence of interest and a 3' primer binding site, and a second vector which comprises a reverse transcriptase gene.
  • the vector or vector system of the present invention is a plasmid or modified viral construct.
  • the reverse transcriptase gene is operably linked to an expression control sequence.
  • the vector or vector systems of the present invention may be advantageously employed to deliver antisense, sense, triplex, or any other single-stranded nucleotide sequence of interest into a cell, using known digestion and ligation techniques to splice the sequence of interest into the vector.
  • the vector or vector system described herein provides all the necessary signalling instructions and enzymatic functions to allow a host cell to produce a single-stranded nucleic acid molecule having a desired sequence.
  • the vector or vectors systems of the present invention may also be designed to allow the primer binding site to be removed and exchanged, so that different primer binding sites can be used, depending upon the requirements of the user and the specificity of the reverse transcriptase being used.
  • a host cell stably transformed or transfected with a vector or vector system of the present invention in particular, a eukaryotic cell stably transformed or transfected with a vector or vector system of the present invention.
  • Eukaryotic cells include yeast or plant cells, or mammalian cells.
  • kit for producing a single stranded nucleic acid sequence comprises a vector or vector system according to the present invention, and a restriction endonuclease for the insertion site.
  • kit for producing a single-stranded nucleic acid sequence comprises a vector or vector system according to the present invention, a container for the vector/vector system, and instructions for use of the vector/vector system.
  • an in vivo or in vitro method of producing a single-stranded nucleic acid sequence of interest comprises the steps of introducing a nucleic acid construct into a target cell, the nucleic acid construct comprising a sequence of interest and a primer binding site located in a 3' position with respect to the sequence of interest, transcribing the nucleic acid construct into mRNA and reverse transcribing the mRNA into cDNA.
  • the method further comprises the step of removing the mRNA from an mRNA/cDNA heteroduplex formed by reverse transcription of the mRNA.
  • Reverse transcription may be carried out either by a reverse transcriptase expressed by a reverse transcriptase gene introduced into the target cell, or by a reverse transcriptase which is endogenous to the target cell (e.g. where the target cell has been infected with human immunodeficiency virus or simian immunodeficiency virus).
  • the mRNA transcript may be removed from the mRNA/cDNA heteroduplex by means of RNAse H.
  • the RNAse H is expressed from a gene encoding a reverse transcriptase/RNAse H polyprotein introduced into the target cell.
  • the method may comprise the further step of isolating the mRNA transcript, mRNA/cDNA heteroduplex and/or single stranded cDNA from the target cell.
  • a single-stranded cDNA transcript an inhibitory nucleic acid molecule, (e.g. an antisense sequence or an aptamer), an mRNA transcript and/or a heteroduplex molecule produced by the in vivo or in vitro method of the present invention.
  • An inhibitory nucleic acid molecule may be single-stranded DNA synthesized from the mRNA transcript, or the mRNA transcript itself, which can specifically bind to a complementary nucleic acid sequence. Such inhibitory nucleic acid molecules are particularly useful for regulating gene function.
  • An inhibitory nucleic acid molecule may also be an oligo-nucleotide that specifically binds to an RNA or DNA-binding protein, or an oligo-nucleotide that binds to a biomolecule, e.g. thrombin, bradykinin or PGF2 ⁇ , which does not normally bind to RNA or DNA.
  • a pharmaceutical composition which comprises a set of genetic elements, a vector or vector system, or a host cell according to the present invention, together with a pharmacologically acceptable adjuvant, diluent or carrier.
  • a set of genetic elements, a vector or vector system, or a host cell according to the present invention for use in therapy, especially for use in delivering an inhibitory nucleic acid molecule to a target cell.
  • the set of genetic elements, vector and vector systems, and host cells of the present invention are particularly useful for alleviating pathological conditions by regulating gene expression.
  • a set of genetic elements, vector or vector system, or host cell according to the present invention for the manufacture of a medicament for alleviating a pathological condition by regulating gene expression, especially for alleviating a pathological condition by delivery of an inhibitory nucleic acid molecule to a target cell.
  • Other uses are also disclosed.
  • the sets of genetic elements, vectors, vector systems and host cells of the present invention may be used for the prophylactic or therapeutic treatment of a wide range of conditions or diseases, particularly conditions or diseases which are caused by abnormal or altered gene expression, or conditions or diseases which may be alleviated by regulating gene expression.
  • the sets of genetic elements, vectors, host cells, kits and methods of the present invention may be used to produce single-stranded nucleic acid molecules or virtually any predefined or desired nucleotide base composition in a host cell, and are adaptable and applicable to any in vivo or in vitro system.
  • the nucleic acid construct of the present invention is an artificially synthesised, recombinant, chimeric and/or heterologous product and the sequence of interest may be foreign to the host cell in which it is introduced.
  • Figure 1A referenced in the following description is a schematic view of a plasmid containing genetic elements encoding the sequence of interest and a primer binding site for reverse transcriptase.
  • Figure IB is a schematic view of a plasmid containing a gene for reverse transcriptase.
  • Figure 1C is a schematic view of a plasmid containing genetic elements encoding the sequence of interest, a primer binding site, and a gene for reverse transcriptase.
  • FIG. 2 is a schematic diagram illustrating one embodiment of the method of the present invention.
  • a vector (as used herein, the term “vector” refers to a plasmid or modified viral construct, or any other suitable vehicle, used to deliver and/or manipulate nucleic acid sequences of interest) was designed to produce ssDNA in vivo.
  • the vector contains all necessary signaling instructions and enzymatic functions to allow the host cell to produce the ssDNA encoding a desired sequence (a "sequence of interest”). Described are a set of genetic elements adapted for delivery into a cell by incorporation into the vector for synthesizing ssDNA in vitro or in vivo.
  • the vector also contains appropriate promoter(s)/enhancer(s). Also described herein is a method to construct a vector into which these genetic elements have been incorporated.
  • the reverse transcriptase gene which is the first component of the cassette
  • the reverse transcriptase gene from the Moloney Murine Leukemia Virus (MoMuLV) was used to advantage in the examples described.
  • Many other retroviral reverse transcriptase genes may be used to advantage in the cassette of the present invention, it being preferred that the reverse transcriptase gene is regulated by an appropriate upstream promoter/enhancer such as the Cytomegalovirus (CMV) or Rouse Sarcoma Virus (RSV) promoter for expression in eukaryotic cells.
  • the reverse transcriptase gene also preferably includes a downstream polyadenylation signal sequence so that the mRNA produced from the reverse transcriptase gene includes a 3' poly(A) tail for mRNA stability.
  • the reverse transcriptase produced in the cell synthesizes a complementary DNA (cDNA) from the primary mRNA transcript transcribed from the template encoding the genetic element that includes the sequence of interest as described below.
  • cDNA complementary DNA
  • the second component included in the cassette encodes a nucleic acid sequence that provides the template for synthesis of ssDNA in target cells. It is this element that includes the sequence of interest.
  • this genetic element is preferably regulated by an appropriate wide spectrum or tissue-specific promoter(s)/enhancer(s), such as the SV-40 promoter, or combination of promoter(s)/enhancer(s), located upstream of the genetic element.
  • tissue-specific or wide spectrum promoters/enhancers, or combinations of promoters/enhancers may be used to advantage to regulate the reverse transcriptase gene and sequence of interest.
  • the promoters/enhancers may be constitutive or inducible and may include the CMV or RSV (non-cell type specific) or GFAP (tissue specific) promoters/enhancers listed here and many other viral or mammalian promoters.
  • Representative promoters/enhancers that are appropriate for use in connection with the present invention may include, but are not limited to, HSVtk (McKnight et al., 217 Science 316, 1982), human beta-globulin (Breathnach et al., 50 Ann. Rev. of Biochem.
  • beta-actin Kawamoto et al., 8 Mol. Cell Biol. 267, 1988
  • rat growth hormone Larsen et al, 83 Proc. Natl. Acad. Sci. U.S.A. 8283, 1986
  • MMTV Human et al., 27 Cell 245 1981
  • adenovirus 5 E2 Imperiale, et al, 4 Mol. Cell. Biol. 875, 1984
  • SV40 Angel et al, 49 Cell 729, 1987
  • a-2-macroglobulin Kunz, et al., 17 Nucl. Acids Res.
  • PBS primer-binding site
  • tRNA transfer RNA
  • any PBS that is matched to the reverse transcriptase that comprises the set of genetic elements may be utilized for this purpose.
  • Multiple copies of the sequences of interest, each with its corresponding PBS, can be incorporated into the vector for delivery to a cell in accordance with the method of the present invention if desired, for example, for use in delivering anti-sense sequences to various regions of a gene within the target cell.
  • the mRNA primary transcript transcribed from the genetic element acts as the template used by the reverse transcriptase described above to synthesize and process the sequence of interest, which as noted above, can be any desired ssDNA.
  • the mRNA primary transcript contains a primer binding site (PBS) downstream to the sequence of interest.
  • PBS primer binding site
  • the PBS is exclusively recognized by a "primer tRNA.”
  • tRNAs are endogenous to cells. Each tRNA has the ability to recognize a unique sequence (i.e., codon) on the mRNA transcript coding for an amino acid, and has the ability to covalently link to a specific amino acid (i.e., the tRNA becomes "charged" when bound to a specific amino acid).
  • a "primer tRNA" when bound to the mRNA transcript PBS and not covalently linked (i.e., "uncharged") with an amino acid may be used to initiate ssDNA synthesis by the reverse transcriptase.
  • the MoMuLV reverse transcriptase used in the examples described herein recognizes and uses an uncharged lysine tRNA that in turn recognizes and binds to its unique sequence in the PBS.
  • each PBS incorporated into the vector must contain the unique sequence recognized by the primer tRNA, and the primer tRNA must be one that is recognized by the particular reverse transcriptase utilized.
  • the vector contain other specialized genetic elements to facilitate the identification of cells that carry the set of genetic elements of the present invention and/or to increase the level of expression of the sequence of interest.
  • the specialized genetic elements include selectable marker genes so that the vector can be transformed and amplified in a prokaryotic system.
  • selectable markers are genes that confer to the bacteria (e.g., E. coli) resistance to antibiotics such as ampicillin, chloramphenicol, kanamycin (neomycin), or tetracycline.
  • the vector contain specialized genetic elements for subsequent transfection, identification and expression in a eukaryotic system.
  • multiple selection strategies e.g., Chinese Hamster Ovarian: CHO
  • multiple selection strategies e.g., Chinese Hamster Ovarian: CHO
  • Selectable markers used in eukaryotic systems include, but are not limited to, resistance markers for Zeocin, resistance to G418, resistance to aminoglycoside antibiotics, or phenotypic selection markers such ⁇ -gal or green fluorescence protein.
  • the linear ssDNA can be formed into an intact stem-loop ssDNA structure by the addition of inverted tandem repeats flanking the sequence of interest that form the "stem" portion after duplex formation.
  • the stem-loop structure can function similarly in many applications as the linear ssDNA form.
  • Such a ssDNA structure may be more resistant to intracellular nucleases by retaining the "ends" of a ssDNA in double stranded form.
  • the stem can be designed to contain a predetermined sequence or sequences (i.e., "aptamers") that are recognized and bound by specific DNA-binding proteins.
  • a stem structure is used in the cell as a competitor to titer out a selected protein(s) that regulate specific gene expression.
  • Adenovirus El a modulates gene expression of several adenoviral and cellular genes by affecting the activity of cell-encoded transcription factors resulting in changing normal cells to transformed cells.
  • the duplex structure of the stem thus functions to "bind up" the factor, preventing the protein from binding a promoter and thus inhibiting the expression of a particular deleterious gene.
  • the duplex stem structure may optionally contain multiple binding sites, for example, sites which are recognized by various transcription factors that actively regulate expression of particular gene.
  • adenovirus El a has been found to repress transcription of the collagenase gene via the phorbol ester-responsive element, a promoter element responsible for the induction of transcription by 12-O-tetradecanolyphorbol 13-acetate (TPA), by a number of other mitogens, and by the ras, os, src, and trk oncogenes.
  • TPA 12-O-tetradecanolyphorbol 13-acetate
  • the mechanism involves inhibition of the function of the transcription factor family AP-1.
  • the present invention is used to construct complex secondary ssDNA structures in the loop portion of the DNA transcript produced in accordance with the present invention.
  • Such secondary structure is engineered to serve any of several functions.
  • the sequence of interest optionally includes (but is not limited to) a sequence which is incorporated into the loop portion of the single-stranded cDNA transcript to form so-called "clover leaf or "crucible” like structures such as those found in the long terminal repeats of adenoassociated virus or in retrotransposons. Under correct circumstances, such structure is integrated in site-specific manner into the host genome.
  • a vector incorporating the set of genetic elements of the present invention is adaptable for incorporation into multiple commercially available delivery vectors for mammalian and human therapeutic purposes, multiple delivery routes are feasible depending upon the vector chosen for a particular target cell.
  • viral vectors are presently the most frequently used means for transforming the patient's cells and introducing DNA into the genome.
  • viral vectors, carrying new genetic information are used to infect target cells removed from the body, and these cells are then re-implanted (i.e., ex vivo).
  • Direct in vivo gene transfer into postnatal animals has been reported for formulations of DNA encapsulated in liposomes and DNA entrapped in proteoliposomes containing viral envelope receptor proteins (Nicolau et al, Proc.
  • the vector incorporating the set of genetic elements of the present invention is advantageously employed to deliver antisense, sense, triplex, or any other single-stranded nucleotide sequence of interest, using known digestion and ligation techniques to splice the particular sequence of interest into the vector in the presence or absence of inverted tandem repeats.
  • Those skilled in the art who have the benefit of this disclosure will also recognize that the above-described signals used for expression within eukaryotic cells may be modified in ways known in the art depending upon the particular sequence of interest. The most likely change is to change the promoter so as to confer advantageous expression characteristics on the sequence of interest in the system in which it is desired to express the sequence of interest.
  • the present invention is also utilized to produce inhibitory nucleic acids for use in therapeutics in vivo or in vitro.
  • Inhibitory nucleic acids may be ssDNA synthesized from the mRNA template or the mRNA template itself, which can specifically bind to a complementary nucleic acid sequence. By binding to the appropriate target sequence, an RNA— RNA, a DNA— DNA, or RNA-DNA duplex or triplex is formed. More commonly, these nucleic acids are often termed "antisense” because they are usually complementary to the sense or coding strand of the gene, but the "sense" sequence is also utilized in the cell for therapeutic purposes.
  • oligonucleotides that specifically bind to biomolecules that do not normally bind to RNA or DNA has now been demonstrated for a number of biomolecules that vary widely in size, structure and composition. These molecules include: (1) thrombin, a multifunctional regulatory protein that converts fibrinogen to fibrin in the process of clot formation; (2) bradykinin, a nonapeptide kinin involved in blood pressure regulation and implicated in hypotension; (3) PGF2. alpha., a prostaglandin or fatty acid derivative that exhibits hormonal activity. Additionally, the interaction of oligonucleotides with biomolecules whose natural biological function is primarily extracellular has now been demonstrated. U.S. Pat. No. 5,840,867. The term "inhibitory nucleic acids” as used herein, therefore, refers to both "sense” and "antisense” nucleic acids.
  • an inhibitory nucleic acid By binding to the target nucleic acid, an inhibitory nucleic acid inhibits the function of the target nucleic acid.
  • This inhibitory effect results from, for example, blocking DNA transcription, processing or poly(A) addition to mRNA, DNA replication, translation, or promoting inhibitory mechanisms of the cells, such as promoting RNA degradation.
  • Inhibitory nucleic acid methods therefore encompass a number of different approaches to altering expression of genes.
  • An example of an antiherpes virus inhibitory nucleic acid is ISIS 2922 (ISIS Pharmaceuticals, Carlsbad, CA) which has activity against CMV (see Biotechnology News 14:5).
  • inhibitory nucleic acid therapy approaches can be classified into (1) those that target DNA sequences, (2) those that target RNA sequences (including pre-mRNA and mRNA), (3) those that target proteins (sense strand approaches), and (4) those that cause cleavage or chemical modification of the target nucleic acids.
  • the first approach contemplates several categories. Nucleic acids are designed to bind to the major groove of the duplex DNA to form a triple helical or "triplex" structure. Alternatively, inhibitory nucleic acids are designed to bind to regions of single stranded DNA resulting from the opening of the duplex DNA during replication or transcription. More commonly, inhibitory nucleic acids are designed to bind to mRNA or mRNA precursors.
  • Inhibitory nucleic acids are used to prevent maturation of pre-mRNA. Inhibitory nucleic acids may be designed to interfere with RNA processing, splicing or translation. In the second approach, the inhibitory nucleic acids are targeted to mRNA. In this approach, the inhibitory nucleic acids are designed to specifically block translation of the encoded protein. Using this second approach, the inhibitory nucleic acid can be used to selectively suppress certain cellular functions by inhibition of translation of mRNA encoding critical proteins.
  • an inhibitory nucleic acid complementary to regions of c-myc mRNA inhibits c-myc protein expression in a human promyelocytic leukemia cell line, HL60, which overexpresses the c-myc proto-oncogene.
  • HL60 promyelocytic leukemia cell line
  • inhibitory nucleic acids targeting mRNA have been shown to work by several different mechanisms to inhibit translation of the encoded protein(s).
  • the inhibitory nucleic acids introduced into the cell can also utilize the third approach of designing the "sense" strand of the gene or mRNA to trap or compete for the enzymes or binding proteins involved in mRNA translation, as described in Helene and Toulme. Lastly, the inhibitory nucleic acids is used to induce chemical inactivation or cleavage of the target genes or mRNA. Chemical inactivation occurs by the induction of crosslinks between the inhibitory nucleic acid and the target nucleic acid within the cell.
  • the present invention takes the form of a kit comprised of a plasmid having the above-described reverse transcriptase gene cloned therein as well as a multiple cloning site (MCS) into which the user of the kit inserts a particular sequence of interest, which may or may not include the above-described inverted tandem repeats in accordance with the user's intended result.
  • MCS multiple cloning site
  • the MCS is upstream from the genetic element encoding the primer binding site.
  • the resulting plasmid is then purified from the cell culture in which it is maintained, lyophilized or otherwise preserved for packaging and shipping to the user.
  • the kit preferably also includes the restriction endonuclease(s) for the MCS into which the sequence of interest is to be cloned, the ligases and other enzymes for inserting the sequence of interest into the plasmid, and a map of the plasmid, along with suitable reaction buffers.
  • the plasmid pcDNA3.I/Zeo+ was purchased from Invitrogen Corp. (San Diego, CA) and plasmid PBK-RSV from Statagene (La Jolla, CA). Oligodeoxyribonucleotides (ODN) were synthesized by Midland Certified Reagent Co. (Midland, TX). Polymerase chain reactions (PCR) were carried out using Taq DNA polymerase purchased from Boehringer Mannheim Corp. (Indianapolis, IN) in a Robo-gradient thermal cycler (Stratagene (La Jolla, CA). Restriction endonucleases and T4 DNA ligase were obtained from Boehringer Mannheim Corp. (Indianapolis, IN). The ODNs used are listed in the attached Sequence Listing.
  • the following in vivo experiments were designed to determine whether ssDNA could be produced in intact cells.
  • the plasmid utilized included the RSV promoter.
  • Plasmid Constructs The cloning vector pssXB and the plasmids containing the sequences to be expressed as single- stranded DNA were constructed from a common intermediate construct.
  • the host strain for these manipulations was XLl-Blue MRF' (Stratagene, La Jolla, CA).
  • the vector pcDNA3.1Zeo ⁇ (Invitrogen, San Diego, CA) was digested with the restriction enzymes Nhe I and Apa I.
  • This insert contains the Moloney Murine leukemia virus (MoMuLV) reverse transcriptase promoter region.
  • MoMuLV Moloney Murine leukemia virus
  • the plasmid pssDNA-Express-A (pssXA), containing genes for MoMuLV reverse transcriptase, was constructed from the vector pBK-RSV (Stratagene, La Jolla, CA), also using XL-1 Blue MRF' as the host strain.
  • pBK-RSV Stratagene, La Jolla, CA
  • XL-1 Blue MRF' A mouse cell line expressing MoMuLV was obtained from the American Type Culture Collection (ATCC #CRL-1858).
  • Virus RNA was isolated and reverse transcribed from ODN-RT (-) (Table I).
  • the reverse transcript was then PCR amplified according to the manufacturer's intructions using a kit from Promega (Madison, WI), primers ODN-RT (+) and ODN-RT (-), and digested with Sac I and Hind III (sites for these restriction endonucleases are present in the 5' and 3' primers, respectively).
  • the 2.4 kb product obtained includes the sequence of the MoMuLV genome between positions 2546 and 4908.
  • the mature virus reverse transcriptase peptide is encoded by the sequence between positions 2337 and 4349 (Petropoulos, C.J. Retroviral taxonomy, protein structure, sequences and genetic maps. In: Retroviruses,
  • the pBRK-RSV vector was digested with Xba I and Nbe I, which removes the lac promoter region.
  • the Nhe I end was converted to a Sac I end using the linker formed by annealed oligodeoxynucleotides OD ⁇ - ⁇ >S (+) and ODN-N>S (-).
  • the reverse transcriptase amplimers were ligated through the Hind III sites and this construct was subsequently ligated between the Sac I and Xba I sites of pBK-RSV to give pBK-RSV-RT.
  • the set of genetic elements comprising the present invention are also expressed from a single plasmid made by a fusion of, for instance, the pc3.1DNA/Zeo-derived plasmids and the pBK-RSV-derived plasmids such that fused plasmids encode the ss-cDNA-encoding genetic element, the Mo-MuLV-RT gene, and the PBS.
  • pBK-RSV-RT/MboL is digested with Nsil to release a 5.3kb fragment containing the Mo-MuLV-RT gene with an intervening his-pro linker and associated regulatory elements.
  • the 5.3kb DNA fragment is ligated to a linker containing an internal EcoRI site and digested with EcoRI.
  • the pc3.1/Zeo/N-M and the derivative plasmids containing test sequences are digested with Bglll, which recognizes a unique site on pc3.1DNA/Zeo in the cytomegalovirus enhancer/promoter (P CMV).
  • the Bglll ends are ligated to Seq. ID 15 and Seq. ID 16, which contain an internal EcoRI site.
  • the 5.3kb fragment is ligated to pc3.1/Zeo/N-M and derivatives to generate the plasmid.
  • the ss- cDNA is isolated from cells transfected 48-72-hr earlier using triazol reagent (Gibco Life Technologies, Gaithersburg, MD). Assays for specific ss-cDNA species are carried out by both PCR based assays for internal fragment and by denatured single stranded gel electrophoresis with subsequent nylon blotting and probing with an internal biotin-labeled probe.
  • Example 2 Reverse Transcriptase Activity in Transformed Cells To determine the presence of reverse transcriptase activity in extracts of cells containing the pBK-RSV-RT construct, the following assay is used. This assay relies upon reverse transcriptase activity in protein extracts of transformed cells to produce a DNA copy of the Brome Mosaic Virus RNA genome (Silver, et al, 1993). The replication cycle of this virus does not involve a DNA intermediate, eliminating the possibility that an amplification product could be produced without prior reverse transcription.
  • a method and pharmaceutical preparation for diagnosing and treating pathological conditions related to a dopamine receptor abnormality Abnormal activity of the dopaminergic nervous system has been implicated in a number of motor and behavioral disorders including Parkinson's disease, Huntington's disease, tardive dyskinesia, certain forms of schizophrenia and other dystonias and dyskinesias.
  • Dysfunctions of the dopaminergic system may be caused either by a reduced or increased activity of the dopaminergic system or by the inability of the systems to be modulated by a changing external or internal environment.
  • a plasmid is constructed to include a sequence of interest that generates an antisense oligonucleotide capable of binding specifically to an expression-controlling sequence of a nucleic acid encoding the dopamine receptor.
  • the plasmid is administered under conditions whereby the plasmid enters cells expressing the dopamine receptor and generates the inhibitory nucleotide.
  • the inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, thereby selectively controlling expression of one or more dopamine receptor subtypes, and alleviating the pathological conditions related to their expression. Efficacy is tested in accordance with the method described in U.S. Patent No. 5,840,708.
  • KSHV Kaposi's sarcoma-associated herpesvirus
  • Kaposi's sarcoma-associated herpes virus is a new human herpes virus (HHV8) believed to cause Kaposi's sarcoma (KS).
  • Kaposi's sarcoma is the most common neoplasm occurring in persons with acquired immunodeficiency syndrome (AIDS). Approximately 15-20% of AIDS patients develop this neoplasm which rarely occurs in immunocompetent individuals. Epidemiologic evidence suggests that AIDS-associated KS (AIDS-KS) has an infectious etiology.
  • Gay and bisexual AIDS patients are approximately twenty times more likely than hemophiliac AIDS patients to develop KS, and KS may be associated with specific sexual practices among gay men with AIDS.
  • KS is uncommon among adult AIDS patients infected through heterosexual or parenteral HIV transmission, or among pediatric AIDS patients infected through vertical HIV transmission.
  • Agents previously suspected of causing KS include cytomegalovirus, hepatitis B virus, human papillomavirus, Epstein-Barr virus (EBV), human herpesvirus 6, human immunodeficiency virus (HIV), and Mycoplasma penetrans.
  • Non-infectious environmental agents, such as nitrite inhalants also have been proposed to play a role in KS tumorigenesis. Extensive investigations, however, have not demonstrated an etiologic association between any of these agents and AIDS-KS.
  • Virion protein 26 is a component of the nucleocapsid structure in most herpes viruses. This structure serves as a delivery mechanism for the viral genome as it is spread from one infected cell to another. As part of the original infecting virus, it is recognized as a major antigen by the immune system and can therefore be used to screen for antibodies to the herpes virus in patient sera and as a vaccine.
  • a plasmid is constructed using the methods described above to include a sequence of interest.
  • the sequence of interest is an isolated nucleic acid molecule which encodes KSHV virion protein 26 or antisense or triplex oligonucleotide molecule as described in U.S. Patent No. 5,840,708.
  • the plasmid is administered under conditions whereby the plasmid enters infected cells and generates the inhibitory nucleotide.
  • the inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling expression of KSHV virion protein 26, and alleviating the pathological conditions related to expression.
  • Inhibitory nucleotides to modulate the expression of IL-8 and/or IL-8 Receptor to control growth, metastasis and/or angiogenesis in tumors are included in the group consisting of IL-8 and/or IL-8 Receptor and/or angiogenesis in tumors.
  • Interleukin-8 (IL-8, neutrophil activating protein- 1, or NAP-1) is a member of C- X-C chemokine family of related cytokines having broad involvement in inflammatory responses, tissue injury, growth regulation and cellular adhesion. Cerretti, D. P., et al., Molecular Characterization of Receptors for Human Interleukin-8, GRO/Melanoma Growth- Stimulatory Activity and Neutrophil Activating Peptide-2, Molecular Immunology, 30(4), 359-367 (1993); and Koch, A.
  • IL-8 has also been shown to have a potent stimulatory effect on angiogenesis. See, e.g., Koch, A. E., Interleukin-8 as a Macrophage- Derived Mediator of Angiogenesis, Science, 258, 1798-1800 (1992).
  • IL-8 is produced by a variety of normal human somatic cells including monocytes/macrophages, dermal fibroblasts, vascular endothelial cells, keratinocytes, and mesangeal cells. Yasumoto, K., et al., Tumor Necrosis Factor Alpha and Interferon Gamma Synergistically Induce Interleukin 8 Production in a Human Gastric Cancer Cell Line Though Acting Concurrently on AP-1 and NF-kB-like Binding Sites of the Interleukin 8 Gene, J. of Biological Chemistry, 267(31), 22506-11 (1992). Apparently, such cells produce IL-8 only when stressed, and not under conditions of normal growth and homeostasis.
  • IL-8 Factors that induce IL-8 production include inflammation, IL-1, TNF, LPS and thrombin. It is also known that IL-8 is commonly secreted by tumor cells. Because of its effects on growth, it is suspected that IL-8 has a significant role in the metastatic spread of melanoma and other cancers.
  • IL-8 is a ligand for cell-membrane IL-8 Receptor, and it is thought that interaction between IL-8 and IL-8 Receptor is required for IL-8 action.
  • Two IL-8 receptor genes have been identified so far, IL-8 Receptor type A and type B. Both genes belong to the so-called seven transmembrane domain, G protein-coupled receptor family.
  • Receptor A has been shown to be activated by IL-8
  • receptor B has been shown to be activated by IL-8 as well as other cytokines belonging to C-X-C family including Melanoma Growth Stimulatory Activity (MGSA).
  • MGSA Melanoma Growth Stimulatory Activity
  • IL-8 Receptor B present in cancer and other tumor cells is not fully elucidated. There is, however, evidence that activation of IL-8R B (1) is involved in the mechanism of growth regulation of melanoma and tumorigenic fibroblasts; (2) is associated with transformation of lung cells by asbestos, and (3) correlates with metastic potential of melanoma.
  • oligonucleotides which modulate expression of either IL8 or IL-8 Receptor in cancers in vivo. It would be particularly advantageous to provide oligonucleotides which are effective against lung cancer and melanoma because each of these cancers produce their own growth factors.
  • SCLC small cell lung carcinoma
  • NSCLC non-small cell lung carcinoma
  • SCLC comprises approximately one-fourth of the cases, expresses neuroendocrine markers, and generally metastasizes early to lymph nodes, brain, bones, lung and liver.
  • NSCLC comprises the majority of the remaining lung tumor types, and includes adeno-carcinoma, squamous cell carcinoma, and large cell carcinoma.
  • NSCLC is characterized by epithelial-like growth factors and receptors, and is locally invasive.
  • Melanoma cells unlike normal melanocytes, can proliferate in the absence of exogenous growth factors. This independence apparently reflects the production of growth factor and cytokines for autocrine growth stimulation, including TGF-.ANG., TGF-, platelet-derived growth factor A and B chains, basic fibroblast growth factor, IL-8, IL-6, IL-1, granulocyte macrophage colony stimulating factor, and MGSA. Guo Y, et al, Inhibition of Human Melanoma Growth and Metastasis in Vivo by Anti-CD44 Monolclonal Antibody. Cancer Res., 54, 1561-1565 (1994).
  • a plasmid is constructed using the methods described above to include a sequence of interest.
  • the sequence of interest is an isolated nucleic acid molecule as described in U.S. Patent No. 5,849,903.
  • the plasmid is administered (e.g., inhalation or direct injection into solid tumors) under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide.
  • the inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling expression of IL-8 receptors, and alleviating the pathological conditions related to expression.
  • Example 7 Antisense oligonucleotide inhibition of cytomegalovirus infection.
  • Cytomegaloviruses are ubiquitous in nature and are the most common causes of intrauterine infection. Congenital infection is common in newborns of infected mothers. In some populations, as much as 10% of children display perinatal infections. In a small percentage of newborns, the infection is virulent, involving multiple organs.
  • a plasmid is constructed using the methods describe above to include a sequence of interest encoding for an inhibitory nucleotide. Oligonucleotides having a sequence of nucleotide bases specifically hybridizable with a selected sequence of a cytomegalovirus DNA or RNA are described in U.S. Patent No. 5,442,049.
  • the plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide.
  • the inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling replication of CMV, and alleviating the pathological conditions related to CMV infection.
  • This plasmid is used either prophylactically or therapeutically to reduce the severity of disease caused by CMV.
  • Oligonucleotides specifically hybridizable with RNA or DNA deriving from a gene corresponding to one of the open reading frames UL5. UL8. UL9. UL20. UL27. UL29. UL30. UL42. UL52 and IE175 of herpes simplex virus type 1.
  • Oligonucleotides are designed to be specifically hybridizable with DNA or even more preferably, RNA from one of the species herpes simplex virus type 1 (HSV-1), herpes simplex virus type (HSV-2), cytomegalovirus, human herpes virus 6, Epstein Barr virus (EBV) or varicella zoster virus (VZV).
  • HSV-1 herpes simplex virus type 1
  • HSV-2 herpes simplex virus type
  • cytomegalovirus cytomegalovirus
  • EBV Epstein Barr virus
  • VZV varicella zoster virus
  • a plasmid is constructed using the methods described above to include a sequence of interest.
  • the sequence of interest is an isolated nucleic acid molecule as described in U.S. Patent No. 5,514,577.
  • Tthe plasmid is administered (e.g., inhalation or direct injection into solid tumors) under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide.
  • the inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, from one of the species herpes simplex virus type 1 (HSV-1), herpes simplex virus type (HSV-2), cytomegalovirus, human herpes virus 6, Epstein Barr virus (EBV) or varicella zoster virus (VZV) thereby selectively controlling virus infection, and alleviating the pathological conditions related to infection.
  • HSV-1 herpes simplex virus type 1
  • HSV-2 herpes simplex virus type
  • cytomegalovirus cytomegalovirus
  • human herpes virus 6, Epstein Barr virus (EBV) or varicella zoster virus (VZV) varicella zoster virus
  • the proto-oncogene c-myb is the normal cellular homologue of the avian myeloblastosis virus-transforming gene v-myb.
  • the c-myb gene codes for a nuclear protein expressed primarily in hematopoietic cells. It is a proto-oncogene, that is, it codes for a protein which is required for the survival of normal, non-tumor cells. When the gene is altered in the appropriate manner, it has the potential to become an oncogene.
  • Oncogenes are genes whose expression within a cell provides some function in the transformation from normal to tumor cell.
  • An example is the human c-myb gene which has been isolated, cloned, and sequenced. Majello et al, Proc. Natl. Acad. Sci. U.S.A. 83,
  • a plasmid is constructed using the methods describe above to include a sequence of interest encoding for an inhibitory nucleotide. Oligonucleotides having a sequence of nucleotide bases specifically hybridizable with a selected sequence of the DNA or RNA as are described in U.S. Patent No. 5,098,890.
  • the plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide thus acting as an antineoplastic or immunosuppressive agent.
  • inhibitors of ICAM-1, VCAM-1 and ELAM-1 expression would provide a novel therapeutic class of anti-inflammatory agents with activity towards a variety of inflammatory diseases or diseases with an inflammatory component such as asthma, rheumatoid arthritis, allograft rejections, inflammatory bowel disease, various dermatological conditions, and psoriasis.
  • inhibitors of ICAM- 1, VCAM-1, and ELAM-1 may also be effective in the treatment of colds due to rhinovirus infection, AIDS, Kaposi's sarcoma and some cancers and their metastasis.
  • there are no known therapeutic agents which effectively prevent the expression of the cellular adhesion molecules ELAM-1, VCAM-1 and ICAM-1.
  • Monoclonal antibodies may prove to be useful for the treatment of acute inflammatory response due to expression of ICAM-1, VCAM-1 and ELAM-1.
  • the host animal develops antibodies against the monoclonal antibodies thereby limiting their usefulness.
  • monoclonal antibodies are large proteins which may have difficulty in gaining access to the inflammatory site. Soluble forms of the cell adhesion molecules suffer from many of the same limitations as monoclonal antibodies in addition to the expense of their production and their low binding affinity. Thus, there is a long felt need for molecules which effectively inhibit intercellular adhesion molecules.
  • Antisense oligonucleotides avoid many of the pitfalls of current agents used to block the effects of ICAM-1, VCAM-1 and ELAM-1.
  • PCT/US90/02357 discloses DNA sequences encoding Endothelial Adhesion Molecules (ELAMs), including ELAM-1 and VCAM-1 and VCAM- lb.
  • ELAMs Endothelial Adhesion Molecules
  • a number of uses for these DNA sequences are provided, including (1) production of monoclonal antibody preparations that are reactive for these molecules which may be used as therapeutic agents to inhibit leukocyte binding to endothelial cells; (2) production of ELAM peptides to bind to the ELAM ligand on leukocytes which, in turn, may bind to ELAM on endothelial cells, inhibiting leukocyte binding to endothelial cells; (3) use of molecules binding to ELAMS (such as anti-ELAM antibodies, or markers such as the ligand or fragments of it) to detect inflammation; (4) use of ELAM and ELAM ligand DNA sequences to produce nucleic acid molecules that intervene in ELAM or ELAM
  • a plasmid is constructed using the methods describe above to include a sequence of interest encoding for an inhibitory nucleotide for ICAM-1, VCAM-1 or ELAM-1. Oligonucleotides having a sequence of nucleotide bases specifically hybridizable with a selected sequence of ICAM-1, VCAM-1 or ELAM-1 DNA or RNA are described in U.S. Patent No. 5,843,738.
  • the plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide.
  • the inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling the expression of ICAM-1, VCAM- 1 or ELAM-1, and alleviating the pathological conditions related to ICAM-1, VCAM-1 and ELAM-1 expression.
  • This plasmid is used either prophylactically or therapeutically to reduce the severity of inflammation caused by ICAM-1, VCAM-1 and ELAM-1.
  • Protein-Binding Oligonucleotides Specifically Bind Target Molecules
  • Tuerk and Gold describe the use of a procedure termed "systematic evolution of ligands by exponential enrichment.”
  • a pool of RNAs that are completely randomized at specific positions is subjected to selection for binding by a desired nucleic acid-binding protein which has been fixed on a nitrocellulose filter.
  • the bound RNAs then are recovered and amplified as double-stranded DNA that is competent for subsequent in vitro transcription.
  • the newly transcribed RNA then is recycled through this procedure to enrich for oligonucleotides that have consensus sequences for binding by the cognate protein.
  • the oligonucleotides so obtained then may be sequenced for further study. Tuerk and Gold applied this procedure to identify RNA oligonucleotides which are bound by the RNA binding region of T4 DNA polymerase.
  • oligonucleotides that specifically bind to biomolecules that do not normally bind to RNA or DNA has now been demonstrated for a number of biomolecules that vary widely in size, structure and composition. These molecules include: ( 1 ) thrombin, a multifunctional regulatory protein that converts fibrinogen to fibrin in the process of clot formation; (2) bradykinin, a nonapeptide kinin involved in blood pressure regulation and implicated in hypotension; (3) PGF2. alpha., a prostaglandin or fatty acid derivative that exhibits hormonal activity. Additionally, the interaction of oligonucleotides with biomolecules whose natural biological function is primarily extracellular has now been demonstrated.
  • a plasmid is constructed using the methods describe above to include a sequence of interest encoding for an aptamer to thrombin.
  • Aptamers having a sequence of nucleotide bases specifically binding to thrombin are described in U.S. Patent No. 5,840,867.
  • the plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the aptamer.
  • an ex vivo administration is performed where cells are removed from a patient, the plasmid is transfected into the cells, and the cells are then placed back into the patient.
  • the aptamer binds specifically to thrombin, thereby selectively controlling the biological activity of thrombin, and alleviates the pathological conditions related to thrombin' s presence.
  • This plasmid is used either prophylactically or therapeutically.
  • the cassette described herein is described as being made up of three primary components, genetic elements which comprises a sequence of interest and primer binding site, and a reverse transcriptase gene, each of these components being provided with appropriate promoters as described herein.
  • the MoMuLV reverse transcriptase gene described for use as the reverse transcriptase gene of the cassette can be replaced with other reverse transcriptase genes and that promoters other than the CMV promoter may be used to advantage. All such changes and modifications which do not depart from the spirit of the present invention are intended to fall within the scope of the following non-limiting claims.

Abstract

Methods and compositions for producing single-stranded cDNA (ss-cDNA) with a vector-based system in eukaryotic cells. The vector contains all necessary signaling instructions and enzymatic functions to allow the host cell to produce the ssDNA encoding a desired nucleic acid sequence (a 'sequence of interest'). Described are the components included in the vector for synthesizing ssDNA in vivo. They include (1) a reverse transcriptase gene, (2) a genetic element that supplies the template for the desired ssDNA sequence of interest, and (3) a second genetic element located proximal to the genetic element encoding the sequence of interest that supplies the primer site for reverse transcription by the reverse transcriptase molecule. The vector also contains appropriate promoter(s)/enhancer(s). Also described herein is a method to construct a vector including these components.

Description

ENZYMATIC SYNTHESIS OF ssDNA
The present invention relates to the production of single stranded DNA (ssDNA) in yeast, prokaryotic, and eukaryotic cells from a set of genetic elements delivered to the cell by a vector system. The ss DNA is produced in the cell with minimal vector sequences which could interfere with the intended function of the ssDNA in the cell. So far as is known, there is no method for producing single-stranded deoxyribonucleic acid (ssDNA) species in eukaryotic cells which do not contain intervening and/or flanking vector sequences. The scientific and patent literature does include the disclosure of cDNA-producing vectors (see A. Ohshima, et al., 89 Proc. Natl. Acad. Sci. USA 1016-1020 (1992); S. Inouye, et al., 3 Current Opin. Genet. Develop. 713-718 (1993); O. Mirochnitchenko, et al., 269 J. Biol. Chem. 2380-2383 (1994); J.-R. Mao, et al., 270 J. Biol. Chem. 19684-19687 (1995); and U.S. Patent No. 5,436,141), but that system does not appear to have demonstrated the ability to produce ssDNA in eukaryotic cells without intervening vector sequences which can interfere with the intended function of the ssDNA product. It is, therefore, an object of the present invention to provide a DNA construct which directs the synthesis of ssDNA in vitro or in vivo with reduced or eliminated contiguous and/or intervening nucleotide vector sequences.
It is also an object of the present invention to provide a method for producing ssDNA and/or dsDNA in vivo for use as aptamers to which proteins bind for producing a therapeutic effect in a living organism.
It is also an object of the present invention to provide nucleic acid sequences, and a method of introducing such sequences into living cells, for producing a desired effect in a cell, tissue, or organism.
According to the present invention, there is provided a set of genetic elements for delivery into a cell comprising a nucleic acid construct comprising a sequence of interest, and a primer binding site for a reverse transcriptase located in a 3' position with respect to the sequence of interest.
The set of genetic elements of the present invention provides an efficient system for directing the synthesis of a stable, single-stranded nucleic acid sequence, both in vivo and in vitro. The single-stranded nucleic acid sequence may be used to provide a desired effect in a cell, tissue or organism. Because production of the single-stranded nucleic acid sequence of interest takes place within the cell, prior art problems arising from delivery of the single-stranded nucleic acid sequence to the cell are overcome, or at least alleviated. Because of the arrangement of the nucleic acid construct, with the primer binding site in a position which is 3' to the sequence of interest, there is no limit to the size or type of sequence of interest that may be produced using the nucleic acid construct of the present invention, and the construct may be easily incorporated into a vector for delivery by any desired route to a target cell. Reverse transcription may be carried out by a reverse transcriptase which is endogenous to the cell (e.g. in the case of infection by human immunodeficiency virus or simian immunodeficiency virus) or the set of genetic elements may, preferably, further comprise a reverse transcriptase gene.
In the case that the set of genetic elements comprises a reverse transcriptase gene, the reverse transcriptase gene is, preferably, polycistronically transcribable with the sequence of interest and primer binding site.
Preferably, the reverse transcriptase gene is located on the same nucleic acid construct as the sequence of interest and primer binding site and, more preferably, the reverse transcriptase gene is located in a 5' position with respect to said sequence of interest and 3' primer binding site.
The reverse transcriptase gene may encode reverse transcriptase or a reverse transcriptase/RNAse H polyprotein.
The gene encoding reverse transcriptase/RNAse H polyprotein may suitably be derived from Moloney murine leukaemia virus, human immunodeficiency virus, or simian immunodeficiency virus.
Where the set of genetic elements includes a reverse transcriptase gene, the primer binding site is, preferably, specific for the reverse transcriptase encoded by the reverse transcriptase gene. Alternatively, the primer binding site is, preferably, specific for an endogenous reverse transcriptase. Preferably, the primer binding site is complementary to a transfer RNA (tRNA).
Preferably, the set of genetic elements of the present invention also comprises a promoter and, optionally, an enhancer for each of said sequence of interest and/or said reverse transcriptase gene. More preferably, the promoter and/or enhancer is a eukaryotic promoter and/or enhancer.
The promoter may be a constitutive, inducible, wide-spectrum or tissue-specific promoter. Preferably, the set of genetic elements of the present invention further comprises a polyadenylation tail sequence located in a 3' position with respect to the sequence of interest and 3' primer binding site. The polyA tail provides stability of the mRNA transcript.
Preferably, the sequence of interest is an antisense sequence. The present invention, thus, has far reaching uses in the field of antisense therapy, particularly in treating pathological conditions by regulating gene function.
The sequence of interest may also be an aptamer (i.e. an oligonucleotide that binds to a non-oligonucleotide target e.g. a protein). Thus, it can, again, be seen that the present invention, has far reaching therapeutic uses. Preferably, the nucleic acid construct is DNA.
Preferably, the set of genetic elements according to any one of the preceding claims is incorporated into at least one vector.
For example, the sequence of interest and 3' primer binding site may be incorporated into a first vector, with the reverse transcriptase gene incorporated into a second vector.
Alternatively, the reverse transcriptase gene, sequence of interest and primer binding site may be incorporated into a single vector. In this latter case, the reverse transcriptase gene is, preferably, located in a 5' position with respect to the sequence of interest and 3' primer binding site. According to a preferred embodiment of the invention, there is provided a set of genetic elements adapted for delivery into a cell comprising
(a) a sequence of interest and a 3' primer binding site; and
(b) a reverse transcriptase gene, said sequence of interest and 3' primer binding site, and said reverse transcriptase gene being incorporated into at last one vector for delivery into the cell.
The nucleic acid constructs of the present invention are such that they may be incorporated into commercially available delivery vectors for mammalian and human therapeutic purposes, and may be administered by any feasible route, depending on the target cell.
According to the present invention, there is also provided a vector which comprises: (a) a primer binding site and an insertion site for a sequence of interest, the primer binding site being located in a 3' position with respect to the insertion site; and
(b) a reverse transcriptase gene. Preferably, the reverse transcriptase gene is located in a 5' position with respect to the insertion site and 3' primer binding site.
According to another aspect of the present invention, there is provided a vector system which comprises a first vector, comprising an insertion site for a sequence of interest and a 3' primer binding site, and a second vector which comprises a reverse transcriptase gene. Preferably, the vector or vector system of the present invention is a plasmid or modified viral construct.
Preferably, the reverse transcriptase gene is operably linked to an expression control sequence.
The vector or vector systems of the present invention may be advantageously employed to deliver antisense, sense, triplex, or any other single-stranded nucleotide sequence of interest into a cell, using known digestion and ligation techniques to splice the sequence of interest into the vector. The vector or vector system described herein provides all the necessary signalling instructions and enzymatic functions to allow a host cell to produce a single-stranded nucleic acid molecule having a desired sequence. The vector or vectors systems of the present invention may also be designed to allow the primer binding site to be removed and exchanged, so that different primer binding sites can be used, depending upon the requirements of the user and the specificity of the reverse transcriptase being used.
Also provided by the present invention is a host cell stably transformed or transfected with a vector or vector system of the present invention, in particular, a eukaryotic cell stably transformed or transfected with a vector or vector system of the present invention. Eukaryotic cells include yeast or plant cells, or mammalian cells. According to the present invention there is further provided a kit for producing a single stranded nucleic acid sequence, which kit comprises a vector or vector system according to the present invention, and a restriction endonuclease for the insertion site.
According to another aspect of the present invention, there is provided a kit for producing a single-stranded nucleic acid sequence, which kit comprises a vector or vector system according to the present invention, a container for the vector/vector system, and instructions for use of the vector/vector system.
According to the present invention, there is further provided an in vivo or in vitro method of producing a single-stranded nucleic acid sequence of interest, which method comprises the steps of introducing a nucleic acid construct into a target cell, the nucleic acid construct comprising a sequence of interest and a primer binding site located in a 3' position with respect to the sequence of interest, transcribing the nucleic acid construct into mRNA and reverse transcribing the mRNA into cDNA.
Preferably, the method further comprises the step of removing the mRNA from an mRNA/cDNA heteroduplex formed by reverse transcription of the mRNA.
Reverse transcription may be carried out either by a reverse transcriptase expressed by a reverse transcriptase gene introduced into the target cell, or by a reverse transcriptase which is endogenous to the target cell (e.g. where the target cell has been infected with human immunodeficiency virus or simian immunodeficiency virus). The mRNA transcript may be removed from the mRNA/cDNA heteroduplex by means of RNAse H. Preferably, the RNAse H is expressed from a gene encoding a reverse transcriptase/RNAse H polyprotein introduced into the target cell.
Where the single-stranded nucleic acid sequence is prepared by an in vitro method of the present invention, the method may comprise the further step of isolating the mRNA transcript, mRNA/cDNA heteroduplex and/or single stranded cDNA from the target cell.
Also provided by the present invention, are a single-stranded cDNA transcript, an inhibitory nucleic acid molecule, (e.g. an antisense sequence or an aptamer), an mRNA transcript and/or a heteroduplex molecule produced by the in vivo or in vitro method of the present invention. An inhibitory nucleic acid molecule may be single-stranded DNA synthesized from the mRNA transcript, or the mRNA transcript itself, which can specifically bind to a complementary nucleic acid sequence. Such inhibitory nucleic acid molecules are particularly useful for regulating gene function. An inhibitory nucleic acid molecule may also be an oligo-nucleotide that specifically binds to an RNA or DNA-binding protein, or an oligo-nucleotide that binds to a biomolecule, e.g. thrombin, bradykinin or PGF2α, which does not normally bind to RNA or DNA. According to the present invention there is further provided a pharmaceutical composition which comprises a set of genetic elements, a vector or vector system, or a host cell according to the present invention, together with a pharmacologically acceptable adjuvant, diluent or carrier.
According to the present invention there is also provided a set of genetic elements, a vector or vector system, or a host cell according to the present invention for use in therapy, especially for use in delivering an inhibitory nucleic acid molecule to a target cell. The set of genetic elements, vector and vector systems, and host cells of the present invention are particularly useful for alleviating pathological conditions by regulating gene expression. According to a further aspect of the present invention, there is provided the use of a set of genetic elements, vector or vector system, or host cell according to the present invention for the manufacture of a medicament for alleviating a pathological condition by regulating gene expression, especially for alleviating a pathological condition by delivery of an inhibitory nucleic acid molecule to a target cell. Other uses are also disclosed. The sets of genetic elements, vectors, vector systems and host cells of the present invention may be used for the prophylactic or therapeutic treatment of a wide range of conditions or diseases, particularly conditions or diseases which are caused by abnormal or altered gene expression, or conditions or diseases which may be alleviated by regulating gene expression. The sets of genetic elements, vectors, host cells, kits and methods of the present invention may be used to produce single-stranded nucleic acid molecules or virtually any predefined or desired nucleotide base composition in a host cell, and are adaptable and applicable to any in vivo or in vitro system.
According to a preferred embodiment, the nucleic acid construct of the present invention is an artificially synthesised, recombinant, chimeric and/or heterologous product and the sequence of interest may be foreign to the host cell in which it is introduced. Figure 1A referenced in the following description is a schematic view of a plasmid containing genetic elements encoding the sequence of interest and a primer binding site for reverse transcriptase.
Figure IB is a schematic view of a plasmid containing a gene for reverse transcriptase.
Figure 1C is a schematic view of a plasmid containing genetic elements encoding the sequence of interest, a primer binding site, and a gene for reverse transcriptase.
Figure 2 is a schematic diagram illustrating one embodiment of the method of the present invention. A vector (as used herein, the term "vector" refers to a plasmid or modified viral construct, or any other suitable vehicle, used to deliver and/or manipulate nucleic acid sequences of interest) was designed to produce ssDNA in vivo. The vector contains all necessary signaling instructions and enzymatic functions to allow the host cell to produce the ssDNA encoding a desired sequence (a "sequence of interest"). Described are a set of genetic elements adapted for delivery into a cell by incorporation into the vector for synthesizing ssDNA in vitro or in vivo. They include (1) a reverse transcriptase gene, (2) a genetic element that supplies the template for the desired ssDNA sequence of interest, and (3) a second genetic element located proximal to the genetic element encoding the sequence of interest that supplies the primer site for reverse transcription by the reverse transcriptase molecule. The vector also contains appropriate promoter(s)/enhancer(s). Also described herein is a method to construct a vector into which these genetic elements have been incorporated.
Regarding the reverse transcriptase gene which is the first component of the cassette, the reverse transcriptase gene from the Moloney Murine Leukemia Virus (MoMuLV) was used to advantage in the examples described. Many other retroviral reverse transcriptase genes may be used to advantage in the cassette of the present invention, it being preferred that the reverse transcriptase gene is regulated by an appropriate upstream promoter/enhancer such as the Cytomegalovirus (CMV) or Rouse Sarcoma Virus (RSV) promoter for expression in eukaryotic cells. The reverse transcriptase gene also preferably includes a downstream polyadenylation signal sequence so that the mRNA produced from the reverse transcriptase gene includes a 3' poly(A) tail for mRNA stability. As known to those skilled in the art, multiple poly(A) tails are available and are routinely used for production of expressed eukaryotic genes. The reverse transcriptase produced in the cell synthesizes a complementary DNA (cDNA) from the primary mRNA transcript transcribed from the template encoding the genetic element that includes the sequence of interest as described below. The RNase H activity of the reverse transcriptase, along with endogenous RNase H activity within the cell, degrades the mRNA component of the heteroduplex RNA/cDNA hybrid to produce ssDNA in vivo.
The second component included in the cassette encodes a nucleic acid sequence that provides the template for synthesis of ssDNA in target cells. It is this element that includes the sequence of interest. As is the case for the above reverse transcriptase gene, this genetic element is preferably regulated by an appropriate wide spectrum or tissue- specific promoter(s)/enhancer(s), such as the SV-40 promoter, or combination of promoter(s)/enhancer(s), located upstream of the genetic element. Those skilled in the art who have the benefit of this disclosure will also recognize that a number of tissue- specific or wide spectrum promoters/enhancers, or combinations of promoters/enhancers may be used to advantage to regulate the reverse transcriptase gene and sequence of interest. Although a list of all available promoters/enhancers is not needed to exemplify the invention, the promoters/enhancers may be constitutive or inducible and may include the CMV or RSV (non-cell type specific) or GFAP (tissue specific) promoters/enhancers listed here and many other viral or mammalian promoters. Representative promoters/enhancers that are appropriate for use in connection with the present invention may include, but are not limited to, HSVtk (McKnight et al., 217 Science 316, 1982), human beta-globulin (Breathnach et al., 50 Ann. Rev. of Biochem. 349, 1981), beta-actin (Kawamoto et al., 8 Mol. Cell Biol. 267, 1988), rat growth hormone (Larsen et al, 83 Proc. Natl. Acad. Sci. U.S.A. 8283, 1986), MMTV (Huang et al., 27 Cell 245 1981), adenovirus 5 E2 (Imperiale, et al, 4 Mol. Cell. Biol. 875, 1984), SV40 (Angel et al, 49 Cell 729, 1987), a-2-macroglobulin (Kunz, et al., 17 Nucl. Acids Res. 1121, 1989), MHC class I gene H-2kb (Blanar et al., 8 EMBO J. 1139, 1989), and thyroid stimulating hormone (Chatterjee et α/., 86 Proc. Natl. Acad. Sci. U.S.A. 9114, 1989). For expression in eukaryotic cells, the sequence of interest is followed downstream by a genetic element encoding for a primer-binding site (PBS) for initiation of cDNA synthesis by reverse transcription. The PBS is a sequence that is complementary to a transfer RNA (tRNA) which resides within the eukaryotic target cell. The PBS included in the presently preferred set of genetic elements described herein was taken from the actual
18 nucleotide sequence region of MoMuLV. However, any PBS that is matched to the reverse transcriptase that comprises the set of genetic elements may be utilized for this purpose. Multiple copies of the sequences of interest, each with its corresponding PBS, can be incorporated into the vector for delivery to a cell in accordance with the method of the present invention if desired, for example, for use in delivering anti-sense sequences to various regions of a gene within the target cell.
The mRNA primary transcript transcribed from the genetic element acts as the template used by the reverse transcriptase described above to synthesize and process the sequence of interest, which as noted above, can be any desired ssDNA. The mRNA primary transcript contains a primer binding site (PBS) downstream to the sequence of interest. The PBS is exclusively recognized by a "primer tRNA." To those skilled in the art, tRNAs are endogenous to cells. Each tRNA has the ability to recognize a unique sequence (i.e., codon) on the mRNA transcript coding for an amino acid, and has the ability to covalently link to a specific amino acid (i.e., the tRNA becomes "charged" when bound to a specific amino acid). However, a "primer tRNA" when bound to the mRNA transcript PBS and not covalently linked (i.e., "uncharged") with an amino acid, may be used to initiate ssDNA synthesis by the reverse transcriptase. For example, the MoMuLV reverse transcriptase used in the examples described herein, recognizes and uses an uncharged lysine tRNA that in turn recognizes and binds to its unique sequence in the PBS. Thus, each PBS incorporated into the vector must contain the unique sequence recognized by the primer tRNA, and the primer tRNA must be one that is recognized by the particular reverse transcriptase utilized. It is preferred that the vector contain other specialized genetic elements to facilitate the identification of cells that carry the set of genetic elements of the present invention and/or to increase the level of expression of the sequence of interest. The specialized genetic elements include selectable marker genes so that the vector can be transformed and amplified in a prokaryotic system. For example, the most commonly used selectable markers are genes that confer to the bacteria (e.g., E. coli) resistance to antibiotics such as ampicillin, chloramphenicol, kanamycin (neomycin), or tetracycline. It is also preferred that the vector contain specialized genetic elements for subsequent transfection, identification and expression in a eukaryotic system. For expression in eukaryotic cells, multiple selection strategies (e.g., Chinese Hamster Ovarian: CHO) may be used that confer to the cell resistance to an antibiotic or other drug or alter the phenotype of the cell such as morphological changes, loss of contact inhibition, or increased growth rate. Selectable markers used in eukaryotic systems include, but are not limited to, resistance markers for Zeocin, resistance to G418, resistance to aminoglycoside antibiotics, or phenotypic selection markers such β-gal or green fluorescence protein.
It will also be evident to those skilled in the art from this description that the linear ssDNA can be formed into an intact stem-loop ssDNA structure by the addition of inverted tandem repeats flanking the sequence of interest that form the "stem" portion after duplex formation. The stem-loop structure can function similarly in many applications as the linear ssDNA form. Such a ssDNA structure may be more resistant to intracellular nucleases by retaining the "ends" of a ssDNA in double stranded form.
It will also be evident to those skilled in the art that the stem (duplex DNA) can be designed to contain a predetermined sequence or sequences (i.e., "aptamers") that are recognized and bound by specific DNA-binding proteins. Among other uses, such a stem structure is used in the cell as a competitor to titer out a selected protein(s) that regulate specific gene expression. For example, a ssDNA stem-loop of the present invention in a cell such that the "stem" contains a binding site for a selected transcription factor such as adenovirus El a. Adenovirus El a, like other oncogenes, modulates gene expression of several adenoviral and cellular genes by affecting the activity of cell-encoded transcription factors resulting in changing normal cells to transformed cells. (Jones et al., Genes Dev. 2, 267-281 (1988)). The duplex structure of the stem thus functions to "bind up" the factor, preventing the protein from binding a promoter and thus inhibiting the expression of a particular deleterious gene. To those skilled in the art, it will be clear that the duplex stem structure may optionally contain multiple binding sites, for example, sites which are recognized by various transcription factors that actively regulate expression of particular gene. For example, adenovirus El a has been found to repress transcription of the collagenase gene via the phorbol ester-responsive element, a promoter element responsible for the induction of transcription by 12-O-tetradecanolyphorbol 13-acetate (TPA), by a number of other mitogens, and by the ras, os, src, and trk oncogenes. The mechanism involves inhibition of the function of the transcription factor family AP-1. Offringa et al., 62 Cell 527-538 (1990).
In another aspect which will be recognized by those skilled in the art, the present invention is used to construct complex secondary ssDNA structures in the loop portion of the DNA transcript produced in accordance with the present invention. Such secondary structure is engineered to serve any of several functions. For instance, the sequence of interest optionally includes (but is not limited to) a sequence which is incorporated into the loop portion of the single-stranded cDNA transcript to form so-called "clover leaf or "crucible" like structures such as those found in the long terminal repeats of adenoassociated virus or in retrotransposons. Under correct circumstances, such structure is integrated in site-specific manner into the host genome.
Because a vector incorporating the set of genetic elements of the present invention is adaptable for incorporation into multiple commercially available delivery vectors for mammalian and human therapeutic purposes, multiple delivery routes are feasible depending upon the vector chosen for a particular target cell. For example, viral vectors are presently the most frequently used means for transforming the patient's cells and introducing DNA into the genome. In an indirect method, viral vectors, carrying new genetic information, are used to infect target cells removed from the body, and these cells are then re-implanted (i.e., ex vivo). Direct in vivo gene transfer into postnatal animals has been reported for formulations of DNA encapsulated in liposomes and DNA entrapped in proteoliposomes containing viral envelope receptor proteins (Nicolau et al, Proc. Natl. Acad Sci USA 80: 1068-1072 (1983); Kaneda et al, Science 243:375-378 (1989); Mannino et al, Biotechniques 6:682-690 (1988). Positive results have also been described with calcium phosphate co-precipitated DNA (Benvenisty and Reshef, Proc. Natl. Acad Sci USA 83:9551-9555 (1986)). Such systems include intravenous, intramuscular, and subcutaneous injection, as well as direct intra-tumoral and intra-cavitary injections. The set of genetic elements, when incorporated into the vector of choice, can also be administered through transmucosal, rectal, oral, or inhalation-type methods of delivery.
The vector incorporating the set of genetic elements of the present invention is advantageously employed to deliver antisense, sense, triplex, or any other single-stranded nucleotide sequence of interest, using known digestion and ligation techniques to splice the particular sequence of interest into the vector in the presence or absence of inverted tandem repeats. Those skilled in the art who have the benefit of this disclosure will also recognize that the above-described signals used for expression within eukaryotic cells may be modified in ways known in the art depending upon the particular sequence of interest. The most likely change is to change the promoter so as to confer advantageous expression characteristics on the sequence of interest in the system in which it is desired to express the sequence of interest. There are so many possible promoters and other signals, and they are so dependent on the particular target cell for which the sequence of interest has been selected, that it is impossible to list all che potential enhancers, inducible and constitutive promoter systems, and/or poly(A) tailing systems which may be preferred for a particular target cell and sequence of interest.
The present invention is also utilized to produce inhibitory nucleic acids for use in therapeutics in vivo or in vitro. Inhibitory nucleic acids may be ssDNA synthesized from the mRNA template or the mRNA template itself, which can specifically bind to a complementary nucleic acid sequence. By binding to the appropriate target sequence, an RNA— RNA, a DNA— DNA, or RNA-DNA duplex or triplex is formed. More commonly, these nucleic acids are often termed "antisense" because they are usually complementary to the sense or coding strand of the gene, but the "sense" sequence is also utilized in the cell for therapeutic purposes. For example, the identification of oligonucleotides that specifically bind to biomolecules that do not normally bind to RNA or DNA has now been demonstrated for a number of biomolecules that vary widely in size, structure and composition. These molecules include: (1) thrombin, a multifunctional regulatory protein that converts fibrinogen to fibrin in the process of clot formation; (2) bradykinin, a nonapeptide kinin involved in blood pressure regulation and implicated in hypotension; (3) PGF2. alpha., a prostaglandin or fatty acid derivative that exhibits hormonal activity. Additionally, the interaction of oligonucleotides with biomolecules whose natural biological function is primarily extracellular has now been demonstrated. U.S. Pat. No. 5,840,867. The term "inhibitory nucleic acids" as used herein, therefore, refers to both "sense" and "antisense" nucleic acids.
By binding to the target nucleic acid, an inhibitory nucleic acid inhibits the function of the target nucleic acid. This inhibitory effect results from, for example, blocking DNA transcription, processing or poly(A) addition to mRNA, DNA replication, translation, or promoting inhibitory mechanisms of the cells, such as promoting RNA degradation. Inhibitory nucleic acid methods therefore encompass a number of different approaches to altering expression of genes. An example of an antiherpes virus inhibitory nucleic acid is ISIS 2922 (ISIS Pharmaceuticals, Carlsbad, CA) which has activity against CMV (see Biotechnology News 14:5). These different types of inhibitory nucleic acid technologies are described in Helene, C. and Toulme, J. (1990) Biochim. Biophys. Acta. 1049:99-125, which is referred to hereinafter as "Helene and Toulme."
In brief, inhibitory nucleic acid therapy approaches can be classified into (1) those that target DNA sequences, (2) those that target RNA sequences (including pre-mRNA and mRNA), (3) those that target proteins (sense strand approaches), and (4) those that cause cleavage or chemical modification of the target nucleic acids. The first approach contemplates several categories. Nucleic acids are designed to bind to the major groove of the duplex DNA to form a triple helical or "triplex" structure. Alternatively, inhibitory nucleic acids are designed to bind to regions of single stranded DNA resulting from the opening of the duplex DNA during replication or transcription. More commonly, inhibitory nucleic acids are designed to bind to mRNA or mRNA precursors. Inhibitory nucleic acids are used to prevent maturation of pre-mRNA. Inhibitory nucleic acids may be designed to interfere with RNA processing, splicing or translation. In the second approach, the inhibitory nucleic acids are targeted to mRNA. In this approach, the inhibitory nucleic acids are designed to specifically block translation of the encoded protein. Using this second approach, the inhibitory nucleic acid can be used to selectively suppress certain cellular functions by inhibition of translation of mRNA encoding critical proteins. For example, an inhibitory nucleic acid complementary to regions of c-myc mRNA inhibits c-myc protein expression in a human promyelocytic leukemia cell line, HL60, which overexpresses the c-myc proto-oncogene. See Wickstrom E. L., et al. (1988) PNAS 85: 1028-1032 and Harel-Bellan, A., et al. (1988) Exp. Med. 168:2309- 2318. As described in Helene and Toulme, inhibitory nucleic acids targeting mRNA have been shown to work by several different mechanisms to inhibit translation of the encoded protein(s).
The inhibitory nucleic acids introduced into the cell can also utilize the third approach of designing the "sense" strand of the gene or mRNA to trap or compete for the enzymes or binding proteins involved in mRNA translation, as described in Helene and Toulme. Lastly, the inhibitory nucleic acids is used to induce chemical inactivation or cleavage of the target genes or mRNA. Chemical inactivation occurs by the induction of crosslinks between the inhibitory nucleic acid and the target nucleic acid within the cell.
In another embodiment, the present invention takes the form of a kit comprised of a plasmid having the above-described reverse transcriptase gene cloned therein as well as a multiple cloning site (MCS) into which the user of the kit inserts a particular sequence of interest, which may or may not include the above-described inverted tandem repeats in accordance with the user's intended result. The MCS is upstream from the genetic element encoding the primer binding site. The resulting plasmid is then purified from the cell culture in which it is maintained, lyophilized or otherwise preserved for packaging and shipping to the user. The kit preferably also includes the restriction endonuclease(s) for the MCS into which the sequence of interest is to be cloned, the ligases and other enzymes for inserting the sequence of interest into the plasmid, and a map of the plasmid, along with suitable reaction buffers.
Except where otherwise indicated, standard techniques are described by Seabrook, et al. (1989) (J. Seabrook, et al, Molecular Cloning: A Laboratory Manual (2nd Ed.), Cold Spring Harbor Press (1989), hereinafter referred to as "Maniatis, et al (1989)") were utilized in the examples set out below. Several experimental designs are presented to illustrate the method of producing ssDNA in vivo.
EXAMPLES The following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention. Materials
The plasmid pcDNA3.I/Zeo+ was purchased from Invitrogen Corp. (San Diego, CA) and plasmid PBK-RSV from Statagene (La Jolla, CA). Oligodeoxyribonucleotides (ODN) were synthesized by Midland Certified Reagent Co. (Midland, TX). Polymerase chain reactions (PCR) were carried out using Taq DNA polymerase purchased from Boehringer Mannheim Corp. (Indianapolis, IN) in a Robo-gradient thermal cycler (Stratagene (La Jolla, CA). Restriction endonucleases and T4 DNA ligase were obtained from Boehringer Mannheim Corp. (Indianapolis, IN). The ODNs used are listed in the attached Sequence Listing. Example 1 In vivo Synthesis of ssDNA in Eukaryotic Cells
The following in vivo experiments were designed to determine whether ssDNA could be produced in intact cells. To control expression of the genetic elements cloned into the plasmid in these host cells, the plasmid utilized included the RSV promoter.
However, those skilled in the art who have the benefit of this disclosure will recoginze that any of the eukaryotic promoters listed above can be used for this purpose.
Plasmid Constructs. The cloning vector pssXB and the plasmids containing the sequences to be expressed as single- stranded DNA were constructed from a common intermediate construct. The host strain for these manipulations was XLl-Blue MRF' (Stratagene, La Jolla, CA).
In the first cloning stage, to obtain the common intermediate, the vector pcDNA3.1Zeo~ (Invitrogen, San Diego, CA) was digested with the restriction enzymes Nhe I and Apa I. The double-stranded oligodeoxyribonucleotide having compatible Nhe I and Apa I ends, which is formed by annealing the synthetic, single-stranded oligodeoxynucleotides ODN-PMMV(+) and ODN-PMMV(-) (see Table I ), was ligated into the digested pcDNA3.1Zeo+ to give pcPMMV. This insert contains the Moloney Murine leukemia virus (MoMuLV) reverse transcriptase promoter region. It also contains two Not I sites, unique in pcPMMV. In this construct and in the plasmids deriving from this construct, the strands designated (+) are positioned to be transcribed into RNA from the cytomegalovirus (CMV) promoter of pcDNA3.1/Zeo(+).
The plasmid pssDNA-Express-A (pssXA), containing genes for MoMuLV reverse transcriptase, was constructed from the vector pBK-RSV (Stratagene, La Jolla, CA), also using XL-1 Blue MRF' as the host strain. A mouse cell line expressing MoMuLV was obtained from the American Type Culture Collection (ATCC #CRL-1858). Virus RNA was isolated and reverse transcribed from ODN-RT (-) (Table I). The reverse transcript was then PCR amplified according to the manufacturer's intructions using a kit from Promega (Madison, WI), primers ODN-RT (+) and ODN-RT (-), and digested with Sac I and Hind III (sites for these restriction endonucleases are present in the 5' and 3' primers, respectively). The 2.4 kb product obtained includes the sequence of the MoMuLV genome between positions 2546 and 4908. The mature virus reverse transcriptase peptide is encoded by the sequence between positions 2337 and 4349 (Petropoulos, C.J. Retroviral taxonomy, protein structure, sequences and genetic maps. In: Retroviruses,
757, Appendix 2, Coffin, J.M. (Ed.). Cold Spring Harbor Laboratory Press, Cold Spring
Harbor, New York, USA 1997), but peptides truncated at the amino terminus retain full activity (Sun, et al (1998)). The pBRK-RSV vector was digested with Xba I and Nbe I, which removes the lac promoter region. The Nhe I end was converted to a Sac I end using the linker formed by annealed oligodeoxynucleotides ODΝ-Ν>S (+) and ODN-N>S (-). The reverse transcriptase amplimers were ligated through the Hind III sites and this construct was subsequently ligated between the Sac I and Xba I sites of pBK-RSV to give pBK-RSV-RT.
Those skilled in the art will recognize that the set of genetic elements comprising the present invention are also expressed from a single plasmid made by a fusion of, for instance, the pc3.1DNA/Zeo-derived plasmids and the pBK-RSV-derived plasmids such that fused plasmids encode the ss-cDNA-encoding genetic element, the Mo-MuLV-RT gene, and the PBS. pBK-RSV-RT/MboL is digested with Nsil to release a 5.3kb fragment containing the Mo-MuLV-RT gene with an intervening his-pro linker and associated regulatory elements. The 5.3kb DNA fragment is ligated to a linker containing an internal EcoRI site and digested with EcoRI. The pc3.1/Zeo/N-M and the derivative plasmids containing test sequences are digested with Bglll, which recognizes a unique site on pc3.1DNA/Zeo in the cytomegalovirus enhancer/promoter (P CMV). The Bglll ends are ligated to Seq. ID 15 and Seq. ID 16, which contain an internal EcoRI site. After digestion with EcoRI, the 5.3kb fragment is ligated to pc3.1/Zeo/N-M and derivatives to generate the plasmid.
Tissue culture studies. Stable and transient transfections are carried out by using lipofectant (Boehringer Mannhiem Corp.) using the manufacturer's accompanying instructions. All plasmid constructs were transfected into Cos-7, U251 and HeLa cell lines Assays for ssDNA were performed by PCR and by dot-blot analyses 24-48 hours after transfection. Reverse transcriptase activity was assayed using the RT-PCR assay developed by Silver, et al. (Silver, J., et al 21 Nucleic Acids Res. 3593-4 (1993)). The ss- cDNA is isolated from cells transfected 48-72-hr earlier using triazol reagent (Gibco Life Technologies, Gaithersburg, MD). Assays for specific ss-cDNA species are carried out by both PCR based assays for internal fragment and by denatured single stranded gel electrophoresis with subsequent nylon blotting and probing with an internal biotin-labeled probe.
The experiments described above demonstrate a method of production of ssDNA in vivo by multiple stepwise reactions using eukaryotic reverse transcriptase reactions and various cDNA priming reactions. Any nucleotide sequence of interest is produced by this method in a prokaryotic or eukaryotic cell. The cells were actually co-transfected with two plasmids, one plasmid carrying the genetic elements encoding the sequence of interest and primer binding site for reverse transcriptase, shown in Fig. 1A and the other carrying the gene for reverse transcriptase shown in Fig. IB. Those skilled in the art, however, will recognize that a single plasmid including the genetic elements encoding the sequence of interest and PBS for reverse transcriptase, and the gene for reverse transcriptase also can be used for this purpose (Fig. 1C).
Example 2 Reverse Transcriptase Activity in Transformed Cells To determine the presence of reverse transcriptase activity in extracts of cells containing the pBK-RSV-RT construct, the following assay is used. This assay relies upon reverse transcriptase activity in protein extracts of transformed cells to produce a DNA copy of the Brome Mosaic Virus RNA genome (Silver, et al, 1993). The replication cycle of this virus does not involve a DNA intermediate, eliminating the possibility that an amplification product could be produced without prior reverse transcription.
Example 3
Demonstration of the presence of single-stranded DNA in Transformed Mammalian Cells. A PCR strategy is used to detect single-stranded DNA in transformed cells. The product obtained from RNA extraction procedures, which presumably includes the single- stranded DNA is used as a template in PCR amplifications using primers specific for the expected single-stranded DNA molecule, which is not otherwise present in the cells. A band of the expected size is produced from untreated RNA ssDNA preparations and from such preparations that were treated with RNAase A. Use of preparations treated with SI nuclease, a highly specific, single-stranded DNA endonuclease, does not result in an amplified product. Example 4
A method and pharmaceutical preparation for diagnosing and treating pathological conditions related to a dopamine receptor abnormality. Abnormal activity of the dopaminergic nervous system has been implicated in a number of motor and behavioral disorders including Parkinson's disease, Huntington's disease, tardive dyskinesia, certain forms of schizophrenia and other dystonias and dyskinesias. Dysfunctions of the dopaminergic system may be caused either by a reduced or increased activity of the dopaminergic system or by the inability of the systems to be modulated by a changing external or internal environment.
For a patient suffering from one of the above mentioned disorders, a plasmid is constructed to include a sequence of interest that generates an antisense oligonucleotide capable of binding specifically to an expression-controlling sequence of a nucleic acid encoding the dopamine receptor. The plasmid is administered under conditions whereby the plasmid enters cells expressing the dopamine receptor and generates the inhibitory nucleotide. The inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, thereby selectively controlling expression of one or more dopamine receptor subtypes, and alleviating the pathological conditions related to their expression. Efficacy is tested in accordance with the method described in U.S. Patent No. 5,840,708.
Example 5
Inhibitory nucleotides to Kaposi's sarcoma-associated herpesvirus (KSHV) virion protein 26 (VP26)
Kaposi's sarcoma-associated herpes virus (KSHV) is a new human herpes virus (HHV8) believed to cause Kaposi's sarcoma (KS). Kaposi's sarcoma is the most common neoplasm occurring in persons with acquired immunodeficiency syndrome (AIDS). Approximately 15-20% of AIDS patients develop this neoplasm which rarely occurs in immunocompetent individuals. Epidemiologic evidence suggests that AIDS-associated KS (AIDS-KS) has an infectious etiology. Gay and bisexual AIDS patients are approximately twenty times more likely than hemophiliac AIDS patients to develop KS, and KS may be associated with specific sexual practices among gay men with AIDS. KS is uncommon among adult AIDS patients infected through heterosexual or parenteral HIV transmission, or among pediatric AIDS patients infected through vertical HIV transmission. Agents previously suspected of causing KS include cytomegalovirus, hepatitis B virus, human papillomavirus, Epstein-Barr virus (EBV), human herpesvirus 6, human immunodeficiency virus (HIV), and Mycoplasma penetrans. Non-infectious environmental agents, such as nitrite inhalants, also have been proposed to play a role in KS tumorigenesis. Extensive investigations, however, have not demonstrated an etiologic association between any of these agents and AIDS-KS.
Virion protein 26 (VP26) is a component of the nucleocapsid structure in most herpes viruses. This structure serves as a delivery mechanism for the viral genome as it is spread from one infected cell to another. As part of the original infecting virus, it is recognized as a major antigen by the immune system and can therefore be used to screen for antibodies to the herpes virus in patient sera and as a vaccine.
For an infected patient, a plasmid is constructed using the methods described above to include a sequence of interest. The sequence of interest is an isolated nucleic acid molecule which encodes KSHV virion protein 26 or antisense or triplex oligonucleotide molecule as described in U.S. Patent No. 5,840,708. The plasmid is administered under conditions whereby the plasmid enters infected cells and generates the inhibitory nucleotide. The inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling expression of KSHV virion protein 26, and alleviating the pathological conditions related to expression.
Example 6
Inhibitory nucleotides to modulate the expression of IL-8 and/or IL-8 Receptor to control growth, metastasis and/or angiogenesis in tumors.
Interleukin-8 (IL-8, neutrophil activating protein- 1, or NAP-1) is a member of C- X-C chemokine family of related cytokines having broad involvement in inflammatory responses, tissue injury, growth regulation and cellular adhesion. Cerretti, D. P., et al., Molecular Characterization of Receptors for Human Interleukin-8, GRO/Melanoma Growth- Stimulatory Activity and Neutrophil Activating Peptide-2, Molecular Immunology, 30(4), 359-367 (1993); and Koch, A. E., et al., In situ expression of cytokines and cellular adhesion molecules in the skin of patients with systemic sclerosis, Pathobiology, 61(5-6), 239-46 (1993). IL-8 has also been shown to have a potent stimulatory effect on angiogenesis. See, e.g., Koch, A. E., Interleukin-8 as a Macrophage- Derived Mediator of Angiogenesis, Science, 258, 1798-1800 (1992).
It is known that IL-8 is produced by a variety of normal human somatic cells including monocytes/macrophages, dermal fibroblasts, vascular endothelial cells, keratinocytes, and mesangeal cells. Yasumoto, K., et al., Tumor Necrosis Factor Alpha and Interferon Gamma Synergistically Induce Interleukin 8 Production in a Human Gastric Cancer Cell Line Though Acting Concurrently on AP-1 and NF-kB-like Binding Sites of the Interleukin 8 Gene, J. of Biological Chemistry, 267(31), 22506-11 (1992). Apparently, such cells produce IL-8 only when stressed, and not under conditions of normal growth and homeostasis. Factors that induce IL-8 production include inflammation, IL-1, TNF, LPS and thrombin. It is also known that IL-8 is commonly secreted by tumor cells. Because of its effects on growth, it is suspected that IL-8 has a significant role in the metastatic spread of melanoma and other cancers.
IL-8 is a ligand for cell-membrane IL-8 Receptor, and it is thought that interaction between IL-8 and IL-8 Receptor is required for IL-8 action. Two IL-8 receptor genes have been identified so far, IL-8 Receptor type A and type B. Both genes belong to the so-called seven transmembrane domain, G protein-coupled receptor family. Receptor A has been shown to be activated by IL-8, and receptor B has been shown to be activated by IL-8 as well as other cytokines belonging to C-X-C family including Melanoma Growth Stimulatory Activity (MGSA).
The role and function of IL-8 Receptor B present in cancer and other tumor cells is not fully elucidated. There is, however, evidence that activation of IL-8R B (1) is involved in the mechanism of growth regulation of melanoma and tumorigenic fibroblasts; (2) is associated with transformation of lung cells by asbestos, and (3) correlates with metastic potential of melanoma.
Given the growth stimulatory effect of IL-8 on cells responsive to various tumor growth factors, it would be advantageous to provide antisense oligonucleotides which modulate expression of either IL8 or IL-8 Receptor in cancers in vivo. It would be particularly advantageous to provide oligonucleotides which are effective against lung cancer and melanoma because each of these cancers produce their own growth factors.
There are at least two major types of lung cancer, small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC). SCLC comprises approximately one-fourth of the cases, expresses neuroendocrine markers, and generally metastasizes early to lymph nodes, brain, bones, lung and liver. NSCLC comprises the majority of the remaining lung tumor types, and includes adeno-carcinoma, squamous cell carcinoma, and large cell carcinoma. NSCLC is characterized by epithelial-like growth factors and receptors, and is locally invasive.
Melanoma cells, unlike normal melanocytes, can proliferate in the absence of exogenous growth factors. This independence apparently reflects the production of growth factor and cytokines for autocrine growth stimulation, including TGF-.ANG., TGF-, platelet-derived growth factor A and B chains, basic fibroblast growth factor, IL-8, IL-6, IL-1, granulocyte macrophage colony stimulating factor, and MGSA. Guo Y, et al, Inhibition of Human Melanoma Growth and Metastasis in Vivo by Anti-CD44 Monolclonal Antibody. Cancer Res., 54, 1561-1565 (1994).
For a patient suffering from any of the above diseases, a plasmid is constructed using the methods described above to include a sequence of interest. The sequence of interest is an isolated nucleic acid molecule as described in U.S. Patent No. 5,849,903. To control growth, metastasis and/or angiogenesis, the plasmid is administered (e.g., inhalation or direct injection into solid tumors) under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide. The inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling expression of IL-8 receptors, and alleviating the pathological conditions related to expression.
Example 7 Antisense oligonucleotide inhibition of cytomegalovirus infection.
Cytomegaloviruses (CMVs) are ubiquitous in nature and are the most common causes of intrauterine infection. Congenital infection is common in newborns of infected mothers. In some populations, as much as 10% of children display perinatal infections. In a small percentage of newborns, the infection is virulent, involving multiple organs.
Pronounced involvement of the reticuloendothelial and central nervous system is typical; and the infection is a major cause of mental retardation. Careful testing demonstrates that as many as 50% of severely, prenatally infected adults may display neuropsychiatric disease or deafness. Although extraneural organs are usually spared chronic morbidity, the virus can be detected in the kidney for years. A plasmid is constructed using the methods describe above to include a sequence of interest encoding for an inhibitory nucleotide. Oligonucleotides having a sequence of nucleotide bases specifically hybridizable with a selected sequence of a cytomegalovirus DNA or RNA are described in U.S. Patent No. 5,442,049. The plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide. The inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling replication of CMV, and alleviating the pathological conditions related to CMV infection. This plasmid is used either prophylactically or therapeutically to reduce the severity of disease caused by CMV.
Example 8
Oligonucleotides specifically hybridizable with RNA or DNA deriving from a gene corresponding to one of the open reading frames UL5. UL8. UL9. UL20. UL27. UL29. UL30. UL42. UL52 and IE175 of herpes simplex virus type 1.
Oligonucleotides are designed to be specifically hybridizable with DNA or even more preferably, RNA from one of the species herpes simplex virus type 1 (HSV-1), herpes simplex virus type (HSV-2), cytomegalovirus, human herpes virus 6, Epstein Barr virus (EBV) or varicella zoster virus (VZV). Such oligonucleotides are conveniently and desirably presented as a pharmaceutical composition in a pharmaceutically acceptable carrier as described in U.S. Patent No. 5,514,577.
For a patient suffering from any of the above infections, a plasmid is constructed using the methods described above to include a sequence of interest. The sequence of interest is an isolated nucleic acid molecule as described in U.S. Patent No. 5,514,577. Tthe plasmid is administered (e.g., inhalation or direct injection into solid tumors) under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide. The inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, from one of the species herpes simplex virus type 1 (HSV-1), herpes simplex virus type (HSV-2), cytomegalovirus, human herpes virus 6, Epstein Barr virus (EBV) or varicella zoster virus (VZV) thereby selectively controlling virus infection, and alleviating the pathological conditions related to infection. Example 9
Antisense oligonucleotides to proto-oncogenes. and in particular to the c-myb gene, and the use of such oligonucleotides as antineoplastic and immunosuppressive agents.
The proto-oncogene c-myb is the normal cellular homologue of the avian myeloblastosis virus-transforming gene v-myb. The c-myb gene codes for a nuclear protein expressed primarily in hematopoietic cells. It is a proto-oncogene, that is, it codes for a protein which is required for the survival of normal, non-tumor cells. When the gene is altered in the appropriate manner, it has the potential to become an oncogene.
Oncogenes are genes whose expression within a cell provides some function in the transformation from normal to tumor cell. An example is the human c-myb gene which has been isolated, cloned, and sequenced. Majello et al, Proc. Natl. Acad. Sci. U.S.A. 83,
9636-9640 (1986).
A plasmid is constructed using the methods describe above to include a sequence of interest encoding for an inhibitory nucleotide. Oligonucleotides having a sequence of nucleotide bases specifically hybridizable with a selected sequence of the DNA or RNA as are described in U.S. Patent No. 5,098,890. The plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide thus acting as an antineoplastic or immunosuppressive agent.
Example 10 Antisense oligonucleotides Against ICAM-1 Gene Expression in Interleukin- 1 beta-Stimulated Cells.
It is has been hoped that inhibitors of ICAM-1, VCAM-1 and ELAM-1 expression would provide a novel therapeutic class of anti-inflammatory agents with activity towards a variety of inflammatory diseases or diseases with an inflammatory component such as asthma, rheumatoid arthritis, allograft rejections, inflammatory bowel disease, various dermatological conditions, and psoriasis. In addition, inhibitors of ICAM- 1, VCAM-1, and ELAM-1 may also be effective in the treatment of colds due to rhinovirus infection, AIDS, Kaposi's sarcoma and some cancers and their metastasis. To date, there are no known therapeutic agents which effectively prevent the expression of the cellular adhesion molecules ELAM-1, VCAM-1 and ICAM-1. The use of neutralizing monoclonal antibodies against ICAM-1 in animal models provide evidence that such inhibitors if identified would have therapeutic benefit for asthma; Wegner et al, Science 1990, 247, 456-459, renal allografts; Cosimi et al., J. Immunol. 1990, 144, 4604-4612, and cardiac allografts; Isobe et al., Science 1992, 255, 1125-1127. The use of a soluble form of ICAM- 1 molecule was also effective in preventing rhinovirus infection of cells in culture. Marlin et al, 344 Nature 70-72 (1990). Current agents which affect intercellular adhesion molecules include synthetic peptides, monoclonal antibodies, and soluble forms of the adhesion molecules. To date, synthetic peptides which block the interactions with VCAM-1 or ELAM-1 have not been identified. Monoclonal antibodies may prove to be useful for the treatment of acute inflammatory response due to expression of ICAM-1, VCAM-1 and ELAM-1. However, with chronic treatment, the host animal develops antibodies against the monoclonal antibodies thereby limiting their usefulness. In addition, monoclonal antibodies are large proteins which may have difficulty in gaining access to the inflammatory site. Soluble forms of the cell adhesion molecules suffer from many of the same limitations as monoclonal antibodies in addition to the expense of their production and their low binding affinity. Thus, there is a long felt need for molecules which effectively inhibit intercellular adhesion molecules. Antisense oligonucleotides avoid many of the pitfalls of current agents used to block the effects of ICAM-1, VCAM-1 and ELAM-1.
PCT/US90/02357 (Hession, et al.) discloses DNA sequences encoding Endothelial Adhesion Molecules (ELAMs), including ELAM-1 and VCAM-1 and VCAM- lb. A number of uses for these DNA sequences are provided, including (1) production of monoclonal antibody preparations that are reactive for these molecules which may be used as therapeutic agents to inhibit leukocyte binding to endothelial cells; (2) production of ELAM peptides to bind to the ELAM ligand on leukocytes which, in turn, may bind to ELAM on endothelial cells, inhibiting leukocyte binding to endothelial cells; (3) use of molecules binding to ELAMS (such as anti-ELAM antibodies, or markers such as the ligand or fragments of it) to detect inflammation; (4) use of ELAM and ELAM ligand DNA sequences to produce nucleic acid molecules that intervene in ELAM or ELAM ligand expression at the translational level using antisense nucleic acid and ribozymes to block translation of a specific mRNA either by masking mRNA with antisense nucleic acid or cleaving it with a ribozyme.
A plasmid is constructed using the methods describe above to include a sequence of interest encoding for an inhibitory nucleotide for ICAM-1, VCAM-1 or ELAM-1. Oligonucleotides having a sequence of nucleotide bases specifically hybridizable with a selected sequence of ICAM-1, VCAM-1 or ELAM-1 DNA or RNA are described in U.S. Patent No. 5,843,738. The plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the inhibitory nucleotide. The inhibitory nucleotide binds specifically to expression-controlling sequences of such RNA molecules, or encoding sequences, thereby selectively controlling the expression of ICAM-1, VCAM- 1 or ELAM-1, and alleviating the pathological conditions related to ICAM-1, VCAM-1 and ELAM-1 expression. This plasmid is used either prophylactically or therapeutically to reduce the severity of inflammation caused by ICAM-1, VCAM-1 and ELAM-1. Example 11
Protein-Binding Oligonucleotides (Aptamers) Specifically Bind Target Molecules
The field of rational drug design using biomolecule targeting and aptamer development utilizes oligonucleotides to bind to specific proteins and thus interfere with their function. Described in U.S. Pat. No. 5,840,867, are aptamers to biomolecular targets such as proteins in general, and thrombin in particular. The novel compounds and methods disclosed may be applied broadly to biotechnology diagnostics and therapeutics.
Conventional methods of detection and isolation of proteins and other molecules have employed antibodies and the like which specifically bind such substances. Recently, however, the de novo design of specifically binding oligonucleotides for non- oligonucleotide targets that generally bind nucleic acids has been described. See, e.g., Blackwell, T. K., et al., Science (1990) 250: 1 104-1 110; Blackwell, T. K., et al., Science (1990) 250: 1 149-1 152; Tuerk, C, and Gold, L„ Science (1990) 249:505-510; Joyce, G. F., Gene (1989) 82:83-87. Such oligonucleotides have been termed "aptamers" herein. Tuerk and Gold describe the use of a procedure termed "systematic evolution of ligands by exponential enrichment." In this method, a pool of RNAs that are completely randomized at specific positions is subjected to selection for binding by a desired nucleic acid-binding protein which has been fixed on a nitrocellulose filter. The bound RNAs then are recovered and amplified as double-stranded DNA that is competent for subsequent in vitro transcription. The newly transcribed RNA then is recycled through this procedure to enrich for oligonucleotides that have consensus sequences for binding by the cognate protein. The oligonucleotides so obtained then may be sequenced for further study. Tuerk and Gold applied this procedure to identify RNA oligonucleotides which are bound by the RNA binding region of T4 DNA polymerase.
The identification of oligonucleotides that specifically bind to biomolecules that do not normally bind to RNA or DNA has now been demonstrated for a number of biomolecules that vary widely in size, structure and composition. These molecules include: ( 1 ) thrombin, a multifunctional regulatory protein that converts fibrinogen to fibrin in the process of clot formation; (2) bradykinin, a nonapeptide kinin involved in blood pressure regulation and implicated in hypotension; (3) PGF2. alpha., a prostaglandin or fatty acid derivative that exhibits hormonal activity. Additionally, the interaction of oligonucleotides with biomolecules whose natural biological function is primarily extracellular has now been demonstrated.
A plasmid is constructed using the methods describe above to include a sequence of interest encoding for an aptamer to thrombin. Aptamers having a sequence of nucleotide bases specifically binding to thrombin are described in U.S. Patent No. 5,840,867. The plasmid is administered to the patient under conditions whereby the plasmid enters cells and generates the aptamer. Alterntively, an ex vivo administration is performed where cells are removed from a patient, the plasmid is transfected into the cells, and the cells are then placed back into the patient. The aptamer binds specifically to thrombin, thereby selectively controlling the biological activity of thrombin, and alleviates the pathological conditions related to thrombin' s presence. This plasmid is used either prophylactically or therapeutically.
Although described with reference to the figures and specific examples set out herein, those skilled in the art will recognize that certain changes can be made to the specific elements set out herein without changing the manner in which those elements function to achieve their intended respective results. For instance, the cassette described herein is described as being made up of three primary components, genetic elements which comprises a sequence of interest and primer binding site, and a reverse transcriptase gene, each of these components being provided with appropriate promoters as described herein. Those skilled in the art will recognize that, for instance, the MoMuLV reverse transcriptase gene described for use as the reverse transcriptase gene of the cassette can be replaced with other reverse transcriptase genes and that promoters other than the CMV promoter may be used to advantage. All such changes and modifications which do not depart from the spirit of the present invention are intended to fall within the scope of the following non-limiting claims.
Figure imgf000029_0001

Claims

WHAT IS CLAIMED IS:
1. A set of genetic elements for delivery into a cell comprising: a nucleic acid construct comprising a sequence of interest; and a primer binding site for a reverse transcriptase located in a 3' position with respect to the sequence of interest.
2. A set of genetic elements according to Claim 1, further comprising a reverse transcriptase gene.
3. A set of genetic elements according to Claim 2, wherein the reverse transcriptase gene is polycistronically transcribable with the sequence of interest and primer binding site.
4. A set of genetic elements according to Claim 2 or 3, wherein the reverse transcriptase gene is located on the same nucleic acid construct as the sequence of interest and primer binding site.
5 . A set of genetic elements according to any one of Claims 2 to 4, wherein the reverse transcriptase gene is located in a 5' position with respect to said sequence of interest and 3' primer binding site.
6. A set of genetic elements according to any one of Claims 2 to 4, wherein the reverse transcriptase gene encodes a reverse transcriptase/RNAse H polyprotein.
7. A set of genetic elements according to Claim 6, wherein the gene encoding reverse transcriptase/RNAse H polyprotein is from Moloney murine leukaemia virus, human immunodeficiency virus, or simian immunodeficiency virus.
8. A set of genetic elements according to any one of Claims 2 to 7, wherein the primer binding site is specific for a reverse transcriptase encoded by the reverse transcriptase gene.
9. A set of genetic elements according to Claim 1, wherein the primer binding site is specific for an endogenous reverse transcriptase.
10. A set of genetic elements according to any one of the preceding claims, wherein the primer binding site is complementary to a transfer RNA (tRNA).
1 1. A set of genetic elements according to any one of the preceding claims, further comprising a promoter and, optionally, an enhancer for each of said sequence of interest and/or said reverse transcriptase gene.
12. A set of genetic elements according to Claim 11, wherein the promoter and/or enhancer is a eukaryotic promoter and/or enhancer.
13. A set of genetic elements according to Claim 11 or 12, wherein the promoter is a constitutive, inducible, wide-spectrum or tissue specific promoter.
14. A set of genetic elements according to any one of the preceding claims, further comprising a polyadenylation tail sequence located in a 3' position with respect to the sequence of interest and 3' primer binding site.
15. A set of genetic elements according to any one of the preceding claims, wherein the sequence of interest is an antisense sequence.
16. A set of genetic elements according to any one of Claims 1 to 14, wherein the sequence of interest is an aptamer.
17. A set of genetic elements according to any one of the preceding claims, wherein the nucleic acid construct is DNA.
18. A set of genetic elements according to any one of the preceding claims incorporated into at least one vector.
19. A set of genetic elements according to any one of Claims 2-17, wherein the sequence of interest and 3' primer binding site are incorporated into a first vector, and wherein the reverse transcriptase gene is incorporated into a second vector.
20. A set of genetic elements according to any one of Claims 2-17, wherein the reverse transcriptase gene, sequence of interest and primer binding site are incorporated into a single vector.
21. A set of genetic elements according to Claim 20, wherein the reverse transcriptase gene is located in a 5' position with respect to the sequence of interest and 3' primer binding site.
22. A set of genetic elements adapted for delivery into a cell comprising
(a) a sequence of interest and a 3' primer binding site; and
(b) a reverse transcriptase gene, said sequence of interest and 3' primer binding site, and said reverse transcriptase gene being incorporated into at last one vector for delivery into the cell.
23. A vector which comprises: (a) a primer binding site and an insertion site for a sequence of interest, the primer binding site being located in a 3' position with respect to the insertion site; and
(b) a reverse transcriptase gene.
24. A vector according to Claim 23, wherein the reverse transcriptase gene is located in a 5' position with respect to the insertion site and 3' primer binding site
25. A vector system which comprises a first vector, comprising an insertion site for a sequence of interest and a 3' primer binding site and a second vector which comprises a reverse transcriptase gene.
26. A vector or vector system according to any one of Claims 23 to 25, wherein the vector is a plasmid or modified viral construct.
27. A vector or vector system according to any one of Claims 23 to 26, wherein the reverse transcriptase gene is operably linked to an expression control sequence.
28. A host cell stably transformed or transfected with a vector or vector system according to any of Claims 23 to 27.
29. A host cell according to Claim 28, which is a eukaryotic cell.
30. A kit for producing a single-stranded nucleic acid sequence, which kit comprises a vector or vector system according to any one of Claims 23 to 27, and a restriction endonuclease for the insertion site.
31. A kit for producing a single-stranded nucleic acid sequence, which kit comprises a vector or vector system according to any one of Claims 23 to 27, a container for the vector/vector system and instructions for use of the vector/vector system.
32. An in vivo or in vitro method of producing a single-stranded nucleic acid sequence of interest, which method comprises the steps of introducing a nucleic acid construct into a target cell, the nucleic acid construct comprising a sequence of interest and a primer binding site located in a 3' position with respect to the sequence of interest, transcribing the nucleic acid construct into mRNA and reverse transcribing the mRNA into cDNA.
33. A method according to Claim 32, further comprising the step of removing the mRNA from an mRNA/cDNA heteroduplex formed by reverse transcription of the mRNA.
34. A method according to Claim 32 or 33, wherein reverse transcription is carried out by a reverse transcriptase expressed by a reverse transcriptase gene introduced into the target cell.
35. A method according to Claim 32 or 33, wherein reverse transcription is carried out by a reverse transcriptase which is endogenous to the target cell.
36. A method according to any one of Claims 33 to 35, wherein the mRNA transcript is removed from the mRNA/cDNA heteroduplex by means of RNAse H.
37. A method according to Claim 36, wherein the RNAse H is expressed from a gene encoding a reverse transcriptase/RNAse H polyprotein introduced into the target cell.
38. A method according to any one of Claims 32 to 37, further comprising the step of isolating the mRNA transcript, mRNA/cDNA heteroduplex and/or single-stranded cDNA from the target cell.
39. A single-stranded cDNA transcript produced by the method of any one of Claims 32 to 38.
40. An inhibitory nucleic acid molecule produced by the method of any one of Claims 32 to 38.
41. An inhibitory nucleic acid molecule according to Claim 40, which is an antisense sequence or an aptamer.
42. An mRNA transcript produced by the method of any one of Claims 32 to 38.
43. A heteroduplex molecule produced by the method of any one of Claims 32 to 38.
44. A pharmaceutical composition which comprises a set of genetic elements according to any one of Claims 1 to 22, together with a pharmacologically acceptable adjuvant, diluent or carrier.
45. A pharmaceutical composition which comprises a vector or vector system according to any one of Claims 23 to 27, together with a pharmacologically acceptable adjuvant, diluent or carrier.
46. A pharmaceutical composition which comprises a host cell according to Claim 28 or 29, together with a pharmacologically acceptable adjuvant, diluent or carrier. 51
47. A set of genetic elements according to any one of Claims 1 to 22 for use in therapy, especially for use in delivering an inhibitory nucleic acid molecule to a target cell.
48. A vector or vector system according to any one of Claims 23 to 27 for use in therapy, especially for use in delivering an inhibitory nucleic acid molecule to a target cell.
49. A host cell according to Claim 28 or 29 for use in therapy, especially for use in delivering an inhibitory nucleic acid molecule to a target cell.
50. Use of a set of genetic elements according to any one of Claims 1 to 22, for the manufacture of a medicament for alleviating a pathological condition by regulating gene expression, especially for alleviating a pathological condition by delivery of an inhibitory nucleic acid molecule to a target cell.
51. Use of a vector or vector system according to any one of Claims 23 to 27, for the manufacture of a medicament for alleviating a pathological condition by regulating gene expression, especially for alleviating a pathological condition by delivery of an inhibitory nucleic acid molecule to a target cell.
52. Use of a host cell according to Claim 28 or 29 for the manufacture of a medicament for alleviating a pathological condition by regulating gene expression, especially for alleviating a pathological condition by delivery of an inhibitory nucleic acid molecule to a target cell.
PCT/US1999/023933 1998-10-09 1999-10-12 ENZYMATIC SYNTHESIS OF ssDNA WO2000022113A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA002346155A CA2346155A1 (en) 1998-10-09 1999-10-12 Enzymatic synthesis of ssdna
KR1020017004474A KR20010099682A (en) 1998-10-09 1999-10-12 Enzymatic Synthesis of ssDNA
AU64305/99A AU6430599A (en) 1998-10-09 1999-10-12 Enzymatic synthesis of ssdna
BR9914772-6A BR9914772A (en) 1998-10-09 1999-10-12 Set of genetic elements, vector, host cell, set for the production of a nucleic acid sequence, method for in vivo or in vitro production of a nucleic acid sequence, cdna transcription, inhibitor nucleic acid molecule, mrna transcription, heteroduplex molecule and pharmaceutical composition
MXPA01003642A MXPA01003642A (en) 1998-10-09 1999-10-12 ENZYMATIC SYNTHESIS OF ssDNA.
EP99951989A EP1117776A1 (en) 1998-10-09 1999-10-12 ENZYMATIC SYNTHESIS OF ssDNA
IL14249099A IL142490A0 (en) 1998-10-09 1999-10-12 ENZYMATIC SYNTHESIS OF ssDNA
JP2000576004A JP2002527061A (en) 1998-10-09 1999-10-12 Enzymatic synthesis of ssDNA

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US16979398A 1998-10-09 1998-10-09
US39778399A 1999-09-16 1999-09-16
US09/169,793 1999-09-16
US09/397,783 1999-09-16

Publications (2)

Publication Number Publication Date
WO2000022113A1 true WO2000022113A1 (en) 2000-04-20
WO2000022113A9 WO2000022113A9 (en) 2000-08-24

Family

ID=26865381

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/023933 WO2000022113A1 (en) 1998-10-09 1999-10-12 ENZYMATIC SYNTHESIS OF ssDNA

Country Status (9)

Country Link
EP (1) EP1117776A1 (en)
JP (1) JP2002527061A (en)
KR (1) KR20010099682A (en)
AU (1) AU6430599A (en)
BR (1) BR9914772A (en)
CA (1) CA2346155A1 (en)
IL (1) IL142490A0 (en)
MX (1) MXPA01003642A (en)
WO (1) WO2000022113A1 (en)

Cited By (175)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001025419A1 (en) * 1999-10-04 2001-04-12 Cytogenix, Inc. ALTERING GENE EXPRESSION WITH ssDNA PRODUCED IN VIVO
WO2003052071A2 (en) * 2001-12-14 2003-06-26 Yale University Intracellular generation of single-stranded dna
EP1581054A2 (en) * 2002-12-06 2005-10-05 Cytogenix, Inc. Treatment of hsv-related patholgies using ssdna
WO2007115168A2 (en) 2006-03-31 2007-10-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 gene
JP2008514204A (en) * 2004-09-28 2008-05-08 サイトジェニックス, インコーポレイテッド Single-stranded antibacterial oligonucleotide and use thereof
US7423142B2 (en) 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7473525B2 (en) 2001-01-09 2009-01-06 Alnylam Europe Ag Compositions and methods for inhibiting expression of anti-apoptotic genes
US7605251B2 (en) 2006-05-11 2009-10-20 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US7691824B2 (en) 2006-04-28 2010-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the JC virus
US7737266B2 (en) 2006-09-18 2010-06-15 Board Of Regents, The University Of Texas System RNAi modulation of SCAP and therapeutics uses thereof
WO2010068816A1 (en) 2008-12-10 2010-06-17 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
WO2010089221A1 (en) 2009-02-03 2010-08-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of ptp1b genes
WO2010099341A1 (en) 2009-02-26 2010-09-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mig-12 gene
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
US7812150B2 (en) 2006-05-19 2010-10-12 Alnylam Pharmaceuticals, Inc. RNAi modulation of Aha and therapeutic uses thereof
US7888498B2 (en) 2006-05-22 2011-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
US7919473B2 (en) 2004-03-12 2011-04-05 Alnylam Pharmaceuticals, Inc. IRNA agents targeting VEGF
EP2316943A1 (en) 2007-07-05 2011-05-04 Novartis AG DSRNA for treating viral infection
WO2011054939A2 (en) 2009-11-09 2011-05-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of kif10 genes
WO2011073218A1 (en) 2009-12-18 2011-06-23 F. Hoffmann-La Roche Ag Compositons and methods for inhibiting expression of il-18 genes
WO2011095495A1 (en) 2010-02-05 2011-08-11 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of ikk2 genes
EP2363134A1 (en) 2005-11-09 2011-09-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor V Leiden mutant gene
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
WO2011153323A2 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
EP2441837A1 (en) 2007-03-26 2012-04-18 Alnylam Pharmaceuticals Inc. dsRNA compositions and methods for treating HPV infections
WO2012052258A1 (en) 2010-10-18 2012-04-26 Arrowhead Research Corporation Compositions and methods for inhibiting expression of rrm2 genes
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
US8273869B2 (en) 2009-06-15 2012-09-25 Alnylam Pharmaceuticals, Inc. Lipid formulated dsRNA targeting the PCSK9 gene
US8273868B2 (en) 2001-10-12 2012-09-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting viral replication
WO2012177947A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
WO2012178033A2 (en) 2011-06-23 2012-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
WO2012177784A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Angiopoietin-like 3 (angptl3) irna compostions and methods of use thereof
WO2013003520A1 (en) 2011-06-30 2013-01-03 Arrowhead Research Corporation Compositions and methods for inhibiting gene expression of hepatitis b virus
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
US8470799B2 (en) 2006-09-21 2013-06-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the HAMP gene
EP2617828A1 (en) 2007-12-10 2013-07-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
WO2013155204A2 (en) 2012-04-10 2013-10-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2013163430A2 (en) 2012-04-26 2013-10-31 Alnylam Pharmaceuticals Serpinc1 irna compositions and methods of use thereof
EP2690175A2 (en) 2008-09-02 2014-01-29 Alnylam Pharmaceuticals Compositions and methods for combined inhibition of mutant EGFR gene and IL-6 expression
US8658608B2 (en) 2005-11-23 2014-02-25 Yale University Modified triple-helix forming oligonucleotides for targeted mutagenesis
EP2712926A2 (en) 2008-03-05 2014-04-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 and VEGF genes
WO2014089313A1 (en) 2012-12-05 2014-06-12 Alnylam Pharmaceuticals PCSK9 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2014130922A1 (en) 2013-02-25 2014-08-28 Trustees Of Boston University Compositions and methods for treating fungal infections
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
WO2014190137A1 (en) 2013-05-22 2014-11-27 Alnylam Pharmaceuticals, Inc. SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2014190157A1 (en) 2013-05-22 2014-11-27 Alnylam Pharmaceuticals, Inc. Tmprss6 compositions and methods of use thereof
WO2015050990A1 (en) 2013-10-02 2015-04-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US9051567B2 (en) 2009-06-15 2015-06-09 Tekmira Pharmaceuticals Corporation Methods for increasing efficacy of lipid formulated siRNA
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
EP2937418A1 (en) 2008-10-20 2015-10-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
WO2015175510A1 (en) 2014-05-12 2015-11-19 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
WO2015179724A1 (en) 2014-05-22 2015-11-26 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
US9228188B2 (en) 2011-06-21 2016-01-05 Alnylam Pharmaceuticals, Inc. Compositions and method for inhibiting hepcidin antimicrobial peptide (HAMP) or HAMP-related gene expression
WO2016057893A1 (en) 2014-10-10 2016-04-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2016077321A1 (en) 2014-11-10 2016-05-19 Alnylam Pharmaceuticals, Inc. Hepatitis b virus (hbv) irna compositions and methods of use thereof
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2016130806A2 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2016168286A1 (en) 2015-04-13 2016-10-20 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2016179342A2 (en) 2015-05-06 2016-11-10 Alnylam Pharmaceuticals, Inc. Factor xii (hageman factor) (f12), kallikrein b, plasma (fletcher factor) 1 (klkb1), and kininogen 1 (kng1) irna compositions and methods of use thereof
WO2016201301A1 (en) 2015-06-12 2016-12-15 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
EP3109321A1 (en) 2008-09-25 2016-12-28 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
WO2017040078A1 (en) 2015-09-02 2017-03-09 Alnylam Pharmaceuticals, Inc. PROGRAMMED CELL DEATH 1 LIGAND 1 (PD-L1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2017048843A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2017100542A1 (en) 2015-12-10 2017-06-15 Alnylam Pharmaceuticals, Inc. Sterol regulatory element binding protein (srebp) chaperone (scap) irna compositions and methods of use thereof
WO2017100236A1 (en) 2015-12-07 2017-06-15 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
WO2017184689A1 (en) 2016-04-19 2017-10-26 Alnylam Pharmaceuticals, Inc. High density lipoprotein binding protein (hdlbp/vigilin) irna compositions and methods of use thereof
WO2017214518A1 (en) 2016-06-10 2017-12-14 Alnylam Pharmaceuticals, Inc. COMPLETMENT COMPONENT C5 iRNA COMPOSTIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
US9902955B2 (en) 1999-01-30 2018-02-27 Alnylam Pharmaceuticals, Inc. Method and medicament for inhibiting the expression of a given gene
EP3312281A2 (en) 2013-03-14 2018-04-25 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2018098117A1 (en) 2016-11-23 2018-05-31 Alnylam Pharmaceuticals, Inc. SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US9988627B2 (en) 2013-10-04 2018-06-05 Novartis Ag Formats for organic compounds for use in RNA interference
WO2018112320A1 (en) 2016-12-16 2018-06-21 Alnylam Pharmaceuticals, Inc. Methods for treating or preventing ttr-associated diseases using transthyretin (ttr) irna compositions
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
WO2018195165A1 (en) 2017-04-18 2018-10-25 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
US10130651B2 (en) 2015-08-07 2018-11-20 Arrowhead Pharmaceuticals, Inc. RNAi Therapy for Hepatitis B Virus Infection
WO2019014530A1 (en) 2017-07-13 2019-01-17 Alnylam Pharmaceuticals Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
US10227588B2 (en) 2013-10-04 2019-03-12 Novartis Ag 3′end caps for RNAi agents for use in RNA interference
WO2019089922A1 (en) 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
WO2019099610A1 (en) 2017-11-16 2019-05-23 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
WO2019126097A1 (en) 2017-12-18 2019-06-27 Alnylam Pharmaceuticals, Inc. High mobility group box-1 (hmgb1) irna compositions and methods of use thereof
WO2019122182A1 (en) 2017-12-20 2019-06-27 F. Hoffmann-La Roche Ag Non-replicative transduction particles and transduction particle-based reporter systems
EP3564393A1 (en) 2011-06-21 2019-11-06 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
WO2019222166A1 (en) 2018-05-14 2019-11-21 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
US10519446B2 (en) 2013-10-04 2019-12-31 Novartis Ag Organic compounds to treat hepatitis B virus
WO2020037125A1 (en) 2018-08-16 2020-02-20 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2020060986A1 (en) 2018-09-18 2020-03-26 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2020104649A2 (en) 2018-11-23 2020-05-28 Sanofi Novel rna compositions and methods for inhibiting angptl8
EP3674409A1 (en) 2011-03-29 2020-07-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of tmprss6 gene
WO2020136154A1 (en) 2018-12-27 2020-07-02 F. Hoffmann-La Roche Ag Non-replicative transduction particles and transduction particle-based reporter systems for detection of acinetobacter baumannii
WO2020141124A2 (en) 2018-12-31 2020-07-09 F. Hoffmann-La Roche Ag Non-replicative transduction particles with one or more non-native tail fibers and transduction particle-based reporter systems
EP3693463A1 (en) 2013-10-04 2020-08-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
EP3699592A1 (en) 2013-03-13 2020-08-26 Geneweave Biosciences Inc. Non-replicative transduction particles and transduction particle-based reporter systems
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021022109A1 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. SERPIN FAMILY F MEMBER 2 (SERPINF2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2021037972A1 (en) 2019-08-27 2021-03-04 Sanofi Compositions and methods for inhibiting pcsk9
WO2021046122A1 (en) 2019-09-03 2021-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
EP3798306A1 (en) 2013-12-12 2021-03-31 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021071830A1 (en) 2019-10-07 2021-04-15 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
WO2021076828A1 (en) 2019-10-18 2021-04-22 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
WO2021081026A1 (en) 2019-10-22 2021-04-29 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
WO2021087325A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
WO2021087036A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2021096763A1 (en) 2019-11-13 2021-05-20 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2021102373A1 (en) 2019-11-22 2021-05-27 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
WO2021119226A1 (en) 2019-12-13 2021-06-17 Alnylam Pharmaceuticals, Inc. Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2021136752A1 (en) 2019-12-31 2021-07-08 F. Hoffmann-La Roche Ag Quantitative pcr screening of inducible prophage from bacterial isolates
WO2021151079A1 (en) 2020-01-24 2021-07-29 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
WO2021167841A1 (en) 2020-02-18 2021-08-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2021178736A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021188611A1 (en) 2020-03-18 2021-09-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
WO2021195307A1 (en) 2020-03-26 2021-09-30 Alnylam Pharmaceuticals, Inc. Coronavirus irna compositions and methods of use thereof
US11136597B2 (en) 2016-02-16 2021-10-05 Yale University Compositions for enhancing targeted gene editing and methods of use thereof
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021207167A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021207189A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
WO2021222549A1 (en) 2020-04-30 2021-11-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2021222065A1 (en) 2020-04-27 2021-11-04 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
WO2021257782A1 (en) 2020-06-18 2021-12-23 Alnylam Pharmaceuticals, Inc. XANTHINE DEHYDROGENASE (XDH) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022066847A1 (en) 2020-09-24 2022-03-31 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
WO2022076291A1 (en) 2020-10-05 2022-04-14 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
WO2022079221A1 (en) 2020-10-16 2022-04-21 Sanofi Rna compositions and methods for inhibiting lipoprotein(a)
WO2022079222A1 (en) 2020-10-16 2022-04-21 Sanofi Novel rna compositions and methods for inhibiting angptl3
WO2022087041A1 (en) 2020-10-21 2022-04-28 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
WO2022087329A1 (en) 2020-10-23 2022-04-28 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
WO2022103999A1 (en) 2020-11-13 2022-05-19 Alnylam Pharmaceuticals, Inc. COAGULATION FACTOR V (F5) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022119873A1 (en) 2020-12-01 2022-06-09 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
WO2022150260A1 (en) 2021-01-05 2022-07-14 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT 9 (C9) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022174000A2 (en) 2021-02-12 2022-08-18 Alnylam Pharmaceuticals, Inc. Superoxide dismutase 1 (sod1) irna compositions and methods of use thereof for treating or preventing superoxide dismutase 1- (sod1-) associated neurodegenerative diseases
WO2022182864A1 (en) 2021-02-25 2022-09-01 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods and methods of use thereof
WO2022182574A1 (en) 2021-02-26 2022-09-01 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022187435A1 (en) 2021-03-04 2022-09-09 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
WO2022212153A1 (en) 2021-04-01 2022-10-06 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
WO2022212231A2 (en) 2021-03-29 2022-10-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
WO2022231999A1 (en) 2021-04-26 2022-11-03 Alnylam Pharmaceuticals, Inc. Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
US11517584B2 (en) 2016-08-04 2022-12-06 Arrowhead Pharmaceuticals, Inc. RNAi agents for Hepatitis B virus infection
WO2022256395A1 (en) 2021-06-02 2022-12-08 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2022256290A2 (en) 2021-06-04 2022-12-08 Alnylam Pharmaceuticals, Inc. HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2022260939A2 (en) 2021-06-08 2022-12-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders
WO2023278576A1 (en) 2021-06-30 2023-01-05 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023003995A1 (en) 2021-07-23 2023-01-26 Alnylam Pharmaceuticals, Inc. Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023003922A1 (en) 2021-07-21 2023-01-26 Alnylam Pharmaceuticals, Inc. Metabolic disorder-associated target gene irna compositions and methods of use thereof
WO2023003805A1 (en) 2021-07-19 2023-01-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
WO2023014765A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. iRNA COMPOSITIONS AND METHODS FOR SILENCING ANGIOTENSINOGEN (AGT)
WO2023014677A1 (en) 2021-08-03 2023-02-09 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
WO2023019246A1 (en) 2021-08-13 2023-02-16 Alnylam Pharmaceuticals, Inc. Factor xii (f12) irna compositions and methods of use thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
WO2023044094A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
WO2023064530A1 (en) 2021-10-15 2023-04-20 Alnylam Pharmaceuticals, Inc. Extra-hepatic delivery irna compositions and methods of use thereof
WO2023076451A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2023076450A2 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0530112A2 (en) * 1991-08-30 1993-03-03 University Of Medicine And Dentistry Of New Jersey Method for synthesizing singlestranded stem-loop DNAS, the products and uses therefor
EP0532380A2 (en) * 1991-08-30 1993-03-17 University Of Medicine And Dentistry Of New Jersey Method for synthesizing stable single-stranded cDNA in eukaryotes by means of bacterial retron, products and uses therefor
EP0562206A2 (en) * 1992-01-06 1993-09-29 University Of Medicine & Dentistry Of New Jersey Single-stranded DNA-RNA hybrid molecules and reverse transcriptases
WO1994001550A1 (en) * 1992-07-02 1994-01-20 Hybridon, Inc. Self-stabilized oligonucleotides as therapeutic agents
WO1994013689A1 (en) * 1992-12-09 1994-06-23 Miller Jeffrey E METHODS AND COMPOSITIONS FOR cDNA SYNTHESIS
WO1994020639A1 (en) * 1991-08-30 1994-09-15 The University Of Medicine And Dentistry Of New Jersey Over expession of single-stranded molecules
WO1994023026A1 (en) * 1993-03-26 1994-10-13 Genset Staple and semi-staple oligonucleotides, method of preparation and applications
WO1995035369A1 (en) * 1994-06-22 1995-12-28 Miller Jeffrey E METHODS AND COMPOSITIONS FOR cDNA SYNTHESIS

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0530112A2 (en) * 1991-08-30 1993-03-03 University Of Medicine And Dentistry Of New Jersey Method for synthesizing singlestranded stem-loop DNAS, the products and uses therefor
EP0532380A2 (en) * 1991-08-30 1993-03-17 University Of Medicine And Dentistry Of New Jersey Method for synthesizing stable single-stranded cDNA in eukaryotes by means of bacterial retron, products and uses therefor
WO1994020639A1 (en) * 1991-08-30 1994-09-15 The University Of Medicine And Dentistry Of New Jersey Over expession of single-stranded molecules
EP0562206A2 (en) * 1992-01-06 1993-09-29 University Of Medicine & Dentistry Of New Jersey Single-stranded DNA-RNA hybrid molecules and reverse transcriptases
WO1994001550A1 (en) * 1992-07-02 1994-01-20 Hybridon, Inc. Self-stabilized oligonucleotides as therapeutic agents
WO1994013689A1 (en) * 1992-12-09 1994-06-23 Miller Jeffrey E METHODS AND COMPOSITIONS FOR cDNA SYNTHESIS
WO1994023026A1 (en) * 1993-03-26 1994-10-13 Genset Staple and semi-staple oligonucleotides, method of preparation and applications
WO1995035369A1 (en) * 1994-06-22 1995-12-28 Miller Jeffrey E METHODS AND COMPOSITIONS FOR cDNA SYNTHESIS

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
J.-R. MAO ET AL.: "Gene regulation by antisense DNA produced in vivo", J. BIOL. CHEM., vol. 270, no. 34, 25 August 1995 (1995-08-25), AM. SOC. BIOCHEM. MOL.BIOL.,INC.,BALTIMORE,US, pages 19684 - 19687, XP002132578 *
O. MIROCHNITCHENKO ET AL.: "Production of single-stranded DNA in mammalian cells by means of a bacterial retron", J. BIOL. CHEM., vol. 269, no. 4, 28 January 1994 (1994-01-28), AM. SOC. BIOCHEM. MOL.BIOL.,INC.,BALTIMORE,US, pages 2380 - 2383, XP002132577 *
OHSHIMA A ET AL: "IN-VIVO DUPLICATION OF GENETIC ELEMENTS BY THE FORMATION OF STEM-LOOP DNA WITHOUT AN RNA INTERMEDIATE", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 1992, vol. 89, no. 3, 1992, pages 1016 - 1020, XP002132579, ISSN: 0027-8424 *

Cited By (246)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9902955B2 (en) 1999-01-30 2018-02-27 Alnylam Pharmaceuticals, Inc. Method and medicament for inhibiting the expression of a given gene
US7419964B2 (en) 1999-09-16 2008-09-02 Cytogenix, Inc. Treatment of HSV-related pathologies using ssDNA
WO2001025419A1 (en) * 1999-10-04 2001-04-12 Cytogenix, Inc. ALTERING GENE EXPRESSION WITH ssDNA PRODUCED IN VIVO
US7473525B2 (en) 2001-01-09 2009-01-06 Alnylam Europe Ag Compositions and methods for inhibiting expression of anti-apoptotic genes
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7868160B2 (en) 2001-01-09 2011-01-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7423142B2 (en) 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US8273868B2 (en) 2001-10-12 2012-09-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting viral replication
WO2003052071A2 (en) * 2001-12-14 2003-06-26 Yale University Intracellular generation of single-stranded dna
WO2003052071A3 (en) * 2001-12-14 2003-11-20 Univ Yale Intracellular generation of single-stranded dna
EP1581054A2 (en) * 2002-12-06 2005-10-05 Cytogenix, Inc. Treatment of hsv-related patholgies using ssdna
EP1581054A4 (en) * 2002-12-06 2006-07-26 Cytogenix Inc Treatment of hsv-related patholgies using ssdna
US7947659B2 (en) 2004-03-12 2011-05-24 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
US7919473B2 (en) 2004-03-12 2011-04-05 Alnylam Pharmaceuticals, Inc. IRNA agents targeting VEGF
EP2382997A1 (en) 2004-09-15 2011-11-02 Alnylam Pharmaceuticals Compositions and methods for inhibiting expression of anti-apoptotic genes
JP2008514204A (en) * 2004-09-28 2008-05-08 サイトジェニックス, インコーポレイテッド Single-stranded antibacterial oligonucleotide and use thereof
EP2363134A1 (en) 2005-11-09 2011-09-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor V Leiden mutant gene
US8658608B2 (en) 2005-11-23 2014-02-25 Yale University Modified triple-helix forming oligonucleotides for targeted mutagenesis
US7718629B2 (en) 2006-03-31 2010-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 gene
WO2007115168A2 (en) 2006-03-31 2007-10-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 gene
US7691824B2 (en) 2006-04-28 2010-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the JC virus
US7605251B2 (en) 2006-05-11 2009-10-20 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
EP3872179A1 (en) 2006-05-11 2021-09-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the pcsk9 gene
EP3578656A1 (en) 2006-05-11 2019-12-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the pcsk9 gene
EP3249052A1 (en) 2006-05-11 2017-11-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the pcsk9 gene
EP2835429A1 (en) 2006-05-11 2015-02-11 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US8809292B2 (en) 2006-05-11 2014-08-19 Alnylam Pharmaceuticals, Inc Compositions and methods for inhibiting expression of the PCSK9 gene
EP2194128A1 (en) 2006-05-11 2010-06-09 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US8222222B2 (en) 2006-05-11 2012-07-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US9260718B2 (en) 2006-05-11 2016-02-16 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US10501742B2 (en) 2006-05-11 2019-12-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
US9822365B2 (en) 2006-05-11 2017-11-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
EP2584047A1 (en) 2006-05-11 2013-04-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
EP2584048A1 (en) 2006-05-11 2013-04-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the PCSK9 gene
EP2392583A1 (en) 2006-05-19 2011-12-07 Alnylam Europe AG. RNAi modulation of Aha and therapeutic uses thereof
US7812150B2 (en) 2006-05-19 2010-10-12 Alnylam Pharmaceuticals, Inc. RNAi modulation of Aha and therapeutic uses thereof
US7888498B2 (en) 2006-05-22 2011-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
EP2584051A1 (en) 2006-05-22 2013-04-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expressions of IKK-B gene
US8383805B2 (en) 2006-09-18 2013-02-26 Alnylam Pharmaceuticals, Inc. RNAi modulation of SCAP and therapeutic uses thereof
US7737266B2 (en) 2006-09-18 2010-06-15 Board Of Regents, The University Of Texas System RNAi modulation of SCAP and therapeutics uses thereof
US9102940B2 (en) 2006-09-18 2015-08-11 Alnylam Pharmaceuticals, Inc. RNAi modulation of SCAP and therapeutic uses thereof
US7919613B2 (en) 2006-09-18 2011-04-05 Alnylam Pharmaceuticals, Inc. RNAi modulation of SCAP and therapeutic uses thereof
US9506067B2 (en) 2006-09-21 2016-11-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the HAMP gene
US9090895B2 (en) 2006-09-21 2015-07-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the HAMP gene
US8470799B2 (en) 2006-09-21 2013-06-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the HAMP gene
EP2441837A1 (en) 2007-03-26 2012-04-18 Alnylam Pharmaceuticals Inc. dsRNA compositions and methods for treating HPV infections
US10273482B2 (en) 2007-07-05 2019-04-30 Arrowhead Pharmaceuticals, Inc. dsRNA for treating viral infection
EP2319926A1 (en) 2007-07-05 2011-05-11 Novartis AG DSRNA for treating viral infection
EP2316943A1 (en) 2007-07-05 2011-05-04 Novartis AG DSRNA for treating viral infection
EP2848688A1 (en) 2007-12-10 2015-03-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor VII gene
EP2617828A1 (en) 2007-12-10 2013-07-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
EP2712926A2 (en) 2008-03-05 2014-04-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 and VEGF genes
EP2690175A2 (en) 2008-09-02 2014-01-29 Alnylam Pharmaceuticals Compositions and methods for combined inhibition of mutant EGFR gene and IL-6 expression
EP3208337A1 (en) 2008-09-02 2017-08-23 Alnylam Pharmaceuticals, Inc. Compositions for combined inhibition of mutant egfr and il-6 expression
EP3109321A1 (en) 2008-09-25 2016-12-28 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
EP3584320A1 (en) 2008-09-25 2019-12-25 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
EP2937418A1 (en) 2008-10-20 2015-10-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
EP3848461A1 (en) 2008-10-20 2021-07-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
EP3354733A1 (en) 2008-10-20 2018-08-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
WO2010068816A1 (en) 2008-12-10 2010-06-17 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
EP3225281A1 (en) 2008-12-10 2017-10-04 Alnylam Pharmaceuticals, Inc. Gnaq targeted dsrna compositions and methods for inhibiting expression
WO2010089221A1 (en) 2009-02-03 2010-08-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of ptp1b genes
WO2010099341A1 (en) 2009-02-26 2010-09-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mig-12 gene
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
US8273869B2 (en) 2009-06-15 2012-09-25 Alnylam Pharmaceuticals, Inc. Lipid formulated dsRNA targeting the PCSK9 gene
US10053689B2 (en) 2009-06-15 2018-08-21 Arbutus Biopharma Corporation Methods for increasing efficacy of lipid formulated siRNA
US9051567B2 (en) 2009-06-15 2015-06-09 Tekmira Pharmaceuticals Corporation Methods for increasing efficacy of lipid formulated siRNA
WO2011054939A2 (en) 2009-11-09 2011-05-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of kif10 genes
WO2011073218A1 (en) 2009-12-18 2011-06-23 F. Hoffmann-La Roche Ag Compositons and methods for inhibiting expression of il-18 genes
WO2011095495A1 (en) 2010-02-05 2011-08-11 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of ikk2 genes
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
EP3329924A1 (en) 2010-03-29 2018-06-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
EP3456827A2 (en) 2010-06-02 2019-03-20 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
WO2011153323A2 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
US11920134B2 (en) 2010-10-18 2024-03-05 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of RRM2 genes
EP3434772A2 (en) 2010-10-18 2019-01-30 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of rrm2 genes
EP2851426A2 (en) 2010-10-18 2015-03-25 Arrowhead Research Corporation Compositions and methods for inhibiting expression of RRM2 genes
WO2012052258A1 (en) 2010-10-18 2012-04-26 Arrowhead Research Corporation Compositions and methods for inhibiting expression of rrm2 genes
US9938531B2 (en) 2010-10-18 2018-04-10 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of RRM2 genes
US10619160B2 (en) 2010-10-18 2020-04-14 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of RRM2 genes
US8946176B2 (en) 2010-10-18 2015-02-03 Arrowhead Research Corporation Compositions and methods for inhibiting expression of RRM2 genes
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
EP3674409A1 (en) 2011-03-29 2020-07-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of tmprss6 gene
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
EP4092120A1 (en) 2011-06-21 2022-11-23 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (anglptl3) irna compositions and methods of use thereof
EP3444348A1 (en) 2011-06-21 2019-02-20 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2012177784A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Angiopoietin-like 3 (angptl3) irna compostions and methods of use thereof
US9228188B2 (en) 2011-06-21 2016-01-05 Alnylam Pharmaceuticals, Inc. Compositions and method for inhibiting hepcidin antimicrobial peptide (HAMP) or HAMP-related gene expression
WO2012177947A2 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP3564393A1 (en) 2011-06-21 2019-11-06 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
EP3656860A1 (en) 2011-06-21 2020-05-27 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
EP3693464A2 (en) 2011-06-21 2020-08-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP3366312A1 (en) 2011-06-23 2018-08-29 Alnylam Pharmaceuticals, Inc. Serpina 1 sirnas: compositions of matter and methods of treatment
EP3597750A1 (en) 2011-06-23 2020-01-22 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
WO2012178033A2 (en) 2011-06-23 2012-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
EP4134433A1 (en) 2011-06-23 2023-02-15 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
US8809293B2 (en) 2011-06-30 2014-08-19 Arrowhead Madison Inc. Compositions and methods for inhibiting gene expression of hepatitis B virus
EP3901263A1 (en) 2011-06-30 2021-10-27 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting gene expression of hepatitis b virus
EP3418386A2 (en) 2011-06-30 2018-12-26 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting gene expression of hepatitis b virus
USRE48345E1 (en) 2011-06-30 2020-12-08 Arrowhead Pharmaceuticals Inc. Compositions and methods for inhibiting gene expression of hepatitis B virus
WO2013003520A1 (en) 2011-06-30 2013-01-03 Arrowhead Research Corporation Compositions and methods for inhibiting gene expression of hepatitis b virus
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
EP3284824A1 (en) 2012-04-10 2018-02-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2013155204A2 (en) 2012-04-10 2013-10-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
EP3868883A1 (en) 2012-04-10 2021-08-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2013163430A2 (en) 2012-04-26 2013-10-31 Alnylam Pharmaceuticals Serpinc1 irna compositions and methods of use thereof
EP4209592A1 (en) 2012-04-26 2023-07-12 Genzyme Corporation Serpinc1 irna compositions and methods of use thereof
EP4083209A1 (en) 2012-12-05 2022-11-02 Alnylam Pharmaceuticals, Inc. Pcsk9 irna compositions and methods of use thereof
EP3336187A1 (en) 2012-12-05 2018-06-20 Alnylam Pharmaceuticals, Inc. Pcsk9 irna compositions and methods of use thereof
WO2014089313A1 (en) 2012-12-05 2014-06-12 Alnylam Pharmaceuticals PCSK9 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2014130922A1 (en) 2013-02-25 2014-08-28 Trustees Of Boston University Compositions and methods for treating fungal infections
EP3699592A1 (en) 2013-03-13 2020-08-26 Geneweave Biosciences Inc. Non-replicative transduction particles and transduction particle-based reporter systems
EP3312281A2 (en) 2013-03-14 2018-04-25 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
EP3708184A1 (en) 2013-03-27 2020-09-16 The General Hospital Corporation Methods and agents for treating alzheimer s disease
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
WO2014190137A1 (en) 2013-05-22 2014-11-27 Alnylam Pharmaceuticals, Inc. SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3587578A1 (en) 2013-05-22 2020-01-01 Alnylam Pharmaceuticals, Inc. Tmprss6 irna compositions and methods of use thereof
WO2014190157A1 (en) 2013-05-22 2014-11-27 Alnylam Pharmaceuticals, Inc. Tmprss6 compositions and methods of use thereof
EP3828276A1 (en) 2013-05-22 2021-06-02 Alnylam Pharmaceuticals, Inc. Tmprss6 irna compositions and methods of use thereof
WO2015050990A1 (en) 2013-10-02 2015-04-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US9988627B2 (en) 2013-10-04 2018-06-05 Novartis Ag Formats for organic compounds for use in RNA interference
US10519446B2 (en) 2013-10-04 2019-12-31 Novartis Ag Organic compounds to treat hepatitis B virus
EP3693463A1 (en) 2013-10-04 2020-08-12 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
US10227588B2 (en) 2013-10-04 2019-03-12 Novartis Ag 3′end caps for RNAi agents for use in RNA interference
US11008570B2 (en) 2013-10-04 2021-05-18 Novartis Ag 3′ end caps for RNAi agents for use in RNA interference
EP3798306A1 (en) 2013-12-12 2021-03-31 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
EP3960860A2 (en) 2014-02-11 2022-03-02 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2015175510A1 (en) 2014-05-12 2015-11-19 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
WO2015179724A1 (en) 2014-05-22 2015-11-26 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
EP3739048A1 (en) 2014-05-22 2020-11-18 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
WO2016057893A1 (en) 2014-10-10 2016-04-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
EP4039809A1 (en) 2014-10-10 2022-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
EP3647424A1 (en) 2014-11-10 2020-05-06 Alnylam Pharmaceuticals, Inc. Hepatitis b virus (hbv) irna compositions and methods of use thereof
WO2016077321A1 (en) 2014-11-10 2016-05-19 Alnylam Pharmaceuticals, Inc. Hepatitis b virus (hbv) irna compositions and methods of use thereof
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2016130806A2 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2016168286A1 (en) 2015-04-13 2016-10-20 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2016179342A2 (en) 2015-05-06 2016-11-10 Alnylam Pharmaceuticals, Inc. Factor xii (hageman factor) (f12), kallikrein b, plasma (fletcher factor) 1 (klkb1), and kininogen 1 (kng1) irna compositions and methods of use thereof
WO2016201301A1 (en) 2015-06-12 2016-12-15 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
US10806750B2 (en) 2015-08-07 2020-10-20 Arrowhead Pharmaceuticals, Inc. RNAi therapy for hepatitis B virus infection
US10130651B2 (en) 2015-08-07 2018-11-20 Arrowhead Pharmaceuticals, Inc. RNAi Therapy for Hepatitis B Virus Infection
US11534453B2 (en) 2015-08-07 2022-12-27 Arrowhead Pharmaceuticals, Inc. RNAi therapy for hepatitis B virus infection
WO2017040078A1 (en) 2015-09-02 2017-03-09 Alnylam Pharmaceuticals, Inc. PROGRAMMED CELL DEATH 1 LIGAND 1 (PD-L1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2017048843A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2017100236A1 (en) 2015-12-07 2017-06-15 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating a serpinc1-associated disorder
WO2017100542A1 (en) 2015-12-10 2017-06-15 Alnylam Pharmaceuticals, Inc. Sterol regulatory element binding protein (srebp) chaperone (scap) irna compositions and methods of use thereof
US11136597B2 (en) 2016-02-16 2021-10-05 Yale University Compositions for enhancing targeted gene editing and methods of use thereof
WO2017184689A1 (en) 2016-04-19 2017-10-26 Alnylam Pharmaceuticals, Inc. High density lipoprotein binding protein (hdlbp/vigilin) irna compositions and methods of use thereof
WO2017214518A1 (en) 2016-06-10 2017-12-14 Alnylam Pharmaceuticals, Inc. COMPLETMENT COMPONENT C5 iRNA COMPOSTIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
US11590156B2 (en) 2016-08-04 2023-02-28 Arrowhead Pharmaceuticals, Inc. RNAi agents for hepatitis B virus infection
US11517584B2 (en) 2016-08-04 2022-12-06 Arrowhead Pharmaceuticals, Inc. RNAi agents for Hepatitis B virus infection
WO2018098117A1 (en) 2016-11-23 2018-05-31 Alnylam Pharmaceuticals, Inc. SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2018112320A1 (en) 2016-12-16 2018-06-21 Alnylam Pharmaceuticals, Inc. Methods for treating or preventing ttr-associated diseases using transthyretin (ttr) irna compositions
WO2018195165A1 (en) 2017-04-18 2018-10-25 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
WO2019014530A1 (en) 2017-07-13 2019-01-17 Alnylam Pharmaceuticals Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
WO2019089922A1 (en) 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
WO2019099610A1 (en) 2017-11-16 2019-05-23 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
WO2019126097A1 (en) 2017-12-18 2019-06-27 Alnylam Pharmaceuticals, Inc. High mobility group box-1 (hmgb1) irna compositions and methods of use thereof
WO2019122182A1 (en) 2017-12-20 2019-06-27 F. Hoffmann-La Roche Ag Non-replicative transduction particles and transduction particle-based reporter systems
WO2019222166A1 (en) 2018-05-14 2019-11-21 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
EP4324520A2 (en) 2018-05-14 2024-02-21 Alnylam Pharmaceuticals, Inc. Angiotensinogen (agt) irna compositions and methods of use thereof
WO2020037125A1 (en) 2018-08-16 2020-02-20 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2020060986A1 (en) 2018-09-18 2020-03-26 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2020104649A2 (en) 2018-11-23 2020-05-28 Sanofi Novel rna compositions and methods for inhibiting angptl8
WO2020136154A1 (en) 2018-12-27 2020-07-02 F. Hoffmann-La Roche Ag Non-replicative transduction particles and transduction particle-based reporter systems for detection of acinetobacter baumannii
WO2020141124A2 (en) 2018-12-31 2020-07-09 F. Hoffmann-La Roche Ag Non-replicative transduction particles with one or more non-native tail fibers and transduction particle-based reporter systems
WO2021022109A1 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. SERPIN FAMILY F MEMBER 2 (SERPINF2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2021037972A1 (en) 2019-08-27 2021-03-04 Sanofi Compositions and methods for inhibiting pcsk9
WO2021046122A1 (en) 2019-09-03 2021-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021071830A1 (en) 2019-10-07 2021-04-15 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
WO2021076828A1 (en) 2019-10-18 2021-04-22 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
WO2021081026A1 (en) 2019-10-22 2021-04-29 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
WO2021087325A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
WO2021087036A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2021096763A1 (en) 2019-11-13 2021-05-20 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2021102373A1 (en) 2019-11-22 2021-05-27 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
WO2021119226A1 (en) 2019-12-13 2021-06-17 Alnylam Pharmaceuticals, Inc. Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2021136752A1 (en) 2019-12-31 2021-07-08 F. Hoffmann-La Roche Ag Quantitative pcr screening of inducible prophage from bacterial isolates
WO2021151079A1 (en) 2020-01-24 2021-07-29 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
WO2021167841A1 (en) 2020-02-18 2021-08-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2021178736A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021188611A1 (en) 2020-03-18 2021-09-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
WO2021195307A1 (en) 2020-03-26 2021-09-30 Alnylam Pharmaceuticals, Inc. Coronavirus irna compositions and methods of use thereof
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
WO2021207167A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021207189A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
WO2021222065A1 (en) 2020-04-27 2021-11-04 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
WO2021222549A1 (en) 2020-04-30 2021-11-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
WO2021257782A1 (en) 2020-06-18 2021-12-23 Alnylam Pharmaceuticals, Inc. XANTHINE DEHYDROGENASE (XDH) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022066847A1 (en) 2020-09-24 2022-03-31 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
WO2022076291A1 (en) 2020-10-05 2022-04-14 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
WO2022079222A1 (en) 2020-10-16 2022-04-21 Sanofi Novel rna compositions and methods for inhibiting angptl3
WO2022079221A1 (en) 2020-10-16 2022-04-21 Sanofi Rna compositions and methods for inhibiting lipoprotein(a)
WO2022087041A1 (en) 2020-10-21 2022-04-28 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
WO2022087329A1 (en) 2020-10-23 2022-04-28 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
WO2022103999A1 (en) 2020-11-13 2022-05-19 Alnylam Pharmaceuticals, Inc. COAGULATION FACTOR V (F5) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022119873A1 (en) 2020-12-01 2022-06-09 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
WO2022150260A1 (en) 2021-01-05 2022-07-14 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT 9 (C9) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022174000A2 (en) 2021-02-12 2022-08-18 Alnylam Pharmaceuticals, Inc. Superoxide dismutase 1 (sod1) irna compositions and methods of use thereof for treating or preventing superoxide dismutase 1- (sod1-) associated neurodegenerative diseases
WO2022182864A1 (en) 2021-02-25 2022-09-01 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods and methods of use thereof
WO2022182574A1 (en) 2021-02-26 2022-09-01 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2022187435A1 (en) 2021-03-04 2022-09-09 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
WO2022212231A2 (en) 2021-03-29 2022-10-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
WO2022212153A1 (en) 2021-04-01 2022-10-06 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
WO2022231999A1 (en) 2021-04-26 2022-11-03 Alnylam Pharmaceuticals, Inc. Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022256395A1 (en) 2021-06-02 2022-12-08 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2022256290A2 (en) 2021-06-04 2022-12-08 Alnylam Pharmaceuticals, Inc. HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2022260939A2 (en) 2021-06-08 2022-12-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders
WO2023278576A1 (en) 2021-06-30 2023-01-05 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023003805A1 (en) 2021-07-19 2023-01-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
WO2023003922A1 (en) 2021-07-21 2023-01-26 Alnylam Pharmaceuticals, Inc. Metabolic disorder-associated target gene irna compositions and methods of use thereof
WO2023003995A1 (en) 2021-07-23 2023-01-26 Alnylam Pharmaceuticals, Inc. Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
WO2023014677A1 (en) 2021-08-03 2023-02-09 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
WO2023014765A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. iRNA COMPOSITIONS AND METHODS FOR SILENCING ANGIOTENSINOGEN (AGT)
WO2023019246A1 (en) 2021-08-13 2023-02-16 Alnylam Pharmaceuticals, Inc. Factor xii (f12) irna compositions and methods of use thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
WO2023044094A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
WO2023064530A1 (en) 2021-10-15 2023-04-20 Alnylam Pharmaceuticals, Inc. Extra-hepatic delivery irna compositions and methods of use thereof
WO2023076451A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2023076450A2 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof

Also Published As

Publication number Publication date
IL142490A0 (en) 2002-03-10
KR20010099682A (en) 2001-11-09
MXPA01003642A (en) 2003-07-21
WO2000022113A9 (en) 2000-08-24
CA2346155A1 (en) 2000-04-20
EP1117776A1 (en) 2001-07-25
JP2002527061A (en) 2002-08-27
AU6430599A (en) 2000-05-01
BR9914772A (en) 2001-12-11

Similar Documents

Publication Publication Date Title
WO2000022113A1 (en) ENZYMATIC SYNTHESIS OF ssDNA
Izquierdo et al. Regulation of Fas alternative splicing by antagonistic effects of TIA-1 and PTB on exon definition
KR20110128345A (en) Compositions and methods for the delivery of biologically active rnas
KR19980701418A (en) Stabilized outer guide sequence
WO2000022114A9 (en) PRODUCTION OF ssDNA $i(IN VIVO)
JP2012080889A (en) Multiple-compartment eukaryotic expression systems
WO2006039253A2 (en) Sirna-mediated gene silencing of alpha synuclein
EP2111450A2 (en) Nucleic acid constructs and methods for specific silencing of h19
KR20140123054A (en) COMPOSITIONS AND METHODS FOR THE DELIVERY OF BIOLOGICALLY ACTIVE RNAs
WO2007084954A2 (en) Selective inhibition of ig20 splice variants to treat cancers
US20120301449A1 (en) Rna interference target for treating aids
KR20010042848A (en) Insulin-like growth factor ⅱ antisense oligonucleotide sequences and methods of using same to modulate cell growth
WO2006091112A1 (en) Compositions for the delivery of rna interference molecules and methods for their use
KR20020059608A (en) Altering Gene Expression with ssDNA Produced in vivo
US20030082800A1 (en) In vivo ssDNA expression vectors for altering gene expression
CN116113697A (en) Methods and compositions for treating epilepsy
JP2006500017A (en) Adenoviral VA1 PolIII expression system for RNA expression
Yang et al. Lentiviral-mediated RNA interference against TGF-beta receptor type II in renal epithelial and fibroblast cell populations in vitro demonstrates regulated renal fibrogenesis that is more efficient than a nonlentiviral vector
JP7153033B2 (en) Systems and methods for cell-type specific translation of RNA molecules in eukaryotes
EP1119615B1 (en) Production of ssdna inside a cell
AU2004205192B2 (en) Production of ssDNA in vivo
US20070160581A1 (en) Production of ssDNA in vivo
AU2007249158A1 (en) Production of ssDNA in vivo
EP1111057A1 (en) Prodrug ribozyme
US20050260588A1 (en) In vivo ssdna expression vectors for altering gene expression

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 1999 64305

Country of ref document: AU

Kind code of ref document: A

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: C2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 1-27, DESCRIPTION, REPLACED BY NEW PAGES 1-27; PAGES 28-32, CLAIMS, REPLACED BY NEW PAGES 28-32; PAGES 1/4-4/4, DRAWINGS, REPLACED BY NEW PAGES 1/3-3/3; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

WWE Wipo information: entry into national phase

Ref document number: 142490

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2346155

Country of ref document: CA

Kind code of ref document: A

Country of ref document: CA

Ref document number: 2000 576004

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/003642

Country of ref document: MX

Ref document number: 1020017004474

Country of ref document: KR

Ref document number: 64305/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1999951989

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999951989

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1020017004474

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1999951989

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1020017004474

Country of ref document: KR